Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
researchsquare; 2022.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-1756189.v2

ABSTRACT

Long-term sequelae after Coronavirus disease (COVID)-19 are frequent and of major concern. SARS-CoV-2 infection affects the host's gut microbiota, which is linked with disease severity in patients with COVID-19. We report here that the gut microbiota of post-COVID subjects had a remarkable predominance of Enterobacteriaceae strains with antibiotic-resistance phenotype compared to healthy controls. Additionally, short-chain fatty acids (SCFA) levels were reduced in their feces. Fecal transplant from post-COVID subjects to germ-free mice led to lung inflammation and worst outcomes during pulmonary infection by multidrug-resistant Klebsiella pneumoniae. Transplanted mice also had poorer cognitive performance. Overall, we show prolonged impacts of SARS-CoV-2 infection in the gut microbiota that persist after subjects have cleared the virus. Together, these data demonstrate that the gut microbiota can directly contribute to post-COVID sequelae, suggesting that it may be a potential therapeutic target.


Subject(s)
Coronavirus Infections , Pulmonary Embolism , Klebsiella Infections , Pneumonia , COVID-19
2.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.10.23.465567

ABSTRACT

Coronaviruses belong to a well-known family of enveloped RNA viruses and are the causative agent of the common cold. Although the seasonal coronaviruses do not pose a threat to human life, three members of this family, i.e., SARS-CoV, MERS-CoV and recently, SARS-CoV2, may cause severe acute respiratory syndrome and lead to death. Unfortunately, COVID-19 has already caused more than 4.4 million deaths worldwide. Although much is better understood about the immunopathogenesis of the lung disease, important information about systemic disease is still missing, mainly concerning neurological parameters. In this context, we sought to evaluate immunometabolic changes using in vitro and in vivo models of hamsters infected with SARS-CoV-2. Here we show that, besides infecting hamsters astrocytes, SARS-CoV-2 induces changes in protein expression and metabolic pathways involved in carbon metabolism, glycolysis, mitochondrial respiration, and synaptic transmission. Interestingly, many of the differentially expressed proteins are concurrent with proteins that correlate with neurological diseases, such as Parkinsons's disease, multiple sclerosis, amyotrophic lateral sclerosis, and Huntington's disease. Metabolic analysis by high resolution real-time respirometry evidenced hyperactivation of glycolysis and mitochondrial respiration. Further metabolomics analysis confirmed the consumption of many metabolites, including glucose, pyruvate, glutamine, and alpha ketoglutarate. Interestingly, we observed that glutamine was significantly reduced in infected cultures, and the blockade of mitochondrial glutaminolysis significantly reduced viral replication and pro-inflammatory response. SARS-CoV-2 was confirmed in vivo as hippocampus, cortex, and olfactory bulb of intranasally infected hamsters were positive for viral genome several days post-infection. Altogether, our data reveals important changes in overall protein expression, mostly of those related to carbon metabolism and energy generation, causing an imbalance in important metabolic molecules and neurotransmitters. This may suggest that some of the neurological features observed during COVID-19, as memory and cognitive impairment, may rely on altered energetic profile of brain cells, as well as an unbalanced glutamine/glutamate levels, whose importance for adequate brain function is unquestionable.


Subject(s)
Huntington Disease , Lung Diseases , Severe Acute Respiratory Syndrome , Multiple Sclerosis , Heredodegenerative Disorders, Nervous System , Tooth, Impacted , Parkinson Disease , COVID-19 , Amyotrophic Lateral Sclerosis , Cognition Disorders
3.
Fernanda Crunfli; Victor Corasolla Carregari; Flavio Protásio Veras; Pedro Henrique Vendramini; Aline Gazzola Fragnani Valença; André Saraiva Leão Marcelo Antunes; Caroline Brandão-Teles; Giuliana da Silva Zuccoli; Guilherme Reis-de-Oliveira; Lícia C.Silva-Costa; Verônica Monteiro Saia-Cereda; Ana Campos Codo; Pierina Lorencini Parise; Daniel A. Toledo-Teixeira; Gabriela Fabiano de Souza; Stéfanie Primon Muraro; Bruno Marcel Silva Melo; Glaucia M. Almeida; Egidi Mayara Silva Firmino; Isadora Marques Paiva; Bruna Manuella Souza Silva; Raíssa Guimarães Ludwig; Gabriel Palermo Ruiz; Thiago Leite Knittel; Gustavo Gastão Davanzo; Jaqueline Aline Gerhardt; Patrícia Brito Rodrigues; Julia Forato; Mariene Ribeiro Amorim; Natália Brunetti Silva; Matheus Cavalheiro Martini; Maíra Nilson Benatti; Sabrina Batah; Li Siyuan; Rafael Batista João; Lucas Scardua Silva; Mateus Henrique Nogueira; Ítalo Karmann Aventurato; Mariana Rabelo de Brito; Marina Alvim; José Roberto da Silva Júnior; Lívia Liviane Damião; Maria Ercilia de Paula Castilho Stefano; Iêda Maria Pereira de Sousa; Elessandra Dias da Rocha; Solange Maria Gonçalves; Luiz Henrique Lopes da Silva; Vanessa Bettini; Brunno de Campos; Guilherme Ludwing; Rosa Maria Mendes Viana; Ronaldo Martins; Andre Schwambach Vieira; José Carlos Alves-Filho; Eurico Arruda; Adriano Sebollela; Fernando Cendes; Fernando Cunha; André Ricardo de Lima Damásio; Marco Aurélio Ramirez Vinolo; Carolina Munhoz; Stevens K. Rehen; Thais Mauad; Amaro Duarte-Neto; Luiz Fernando Ferraz da Silva; Marisa Dolhnikoff; Paulo Saldiva; Alexandre Fabro; Alessandro S. Farias; Pedro Manoel M. Moraes-Vieira; José Luiz Proença Módena; Clarissa Yasuda; Marcelo A. Mori; Thiago Mattar Cunha; Daniel Martins de Souza.
researchsquare; 2020.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-104944.v1

ABSTRACT

COVID-19 patients may exhibit neuropsychiatric and neurological symptoms. We found that anxiety and cognitive impairment are manifested by 28-56% of SARS-CoV-2-infected individuals with mild respiratory symptoms and are associated with altered cerebral cortical thickness. Using an independent cohort, we found histopathological signs of brain damage in 25% of individuals who died of COVID-19. All of the affected brain tissues exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Infection of neural stem cell-derived astrocytes changed energy metabolism, altered key proteins and metabolites used to fuel neurons and for biogenesis of neurotransmitters, and elicited a secretory phenotype that reduces neuronal viability. Our data support the model where SARS-CoV-2 reaches the brain, infects astrocytes and triggers neuropathological changes that contribute to the structural and functional alterations in the brain of COVID-19 patients.


Subject(s)
Anxiety Disorders , Signs and Symptoms, Respiratory , Brain Injury, Chronic , Astrocytoma , Severe Acute Respiratory Syndrome , Mental Disorders , COVID-19 , Cognition Disorders
4.
Fernanda Crunfli; Victor Corasolla Carregari; Flavio Protasio Veras; Pedro Henrique Vendramini; Aline Gazzola Fragnani Valenca; Andre Saraiva Leao Marcelo Antunes; Carolina Brandao-Teles; Giuliana da Silva Zuccoli; Guilherme Reis-de-Oliveira; Licia C. Silva-Costa; Verônica Monteiro Saia-Cereda; Ana Campos Codo; Pierina Lorencini Parise; Daniel A. Toledo-Teixeira; Gabriela Fabiano de Souza; Stéfanie Primon Muraro; Bruno Marcel Silva Melo; Glaucia M. Almeida; Egidi Mayara Silva Firmino; Isadora Marques Paiva; Bruna Manuella Souza Silva; Raíssa Guimarães Ludwig; Gabriel Palermo Ruiz; Thiago Leite Knittel; Gustavo Gastão Davanzo; Jaqueline Aline Gerhardt; Patrícia Brito Rodrigues; Julia Forato; Mariene Ribeiro Amorim; Natália Brunetti Silva; Matheus Cavalheiro Martini; Maíra Nilson Benatti; Sabrina Batah; Li Siyuan; Rafael Batista João; Lucas Scardua Silva; Mateus Henrique Nogueira; ítalo Karmann Aventurato; Mariana Rabelo de Brito; Marina Koutsodontis Machado Alvim; José Roberto da Silva Junior; Lívia Liviane Damião; Maria Ercilia de Paula Castilho Stefano; Iêda Maria Pereira de Sousa; Elessandra Dias da Rocha; Solange Maria Gonçalves; Luiz Henrique Lopes da Silva; Vanessa Bettini; Brunno Machado de Campos; Guilherme Ludwig; Rosa Maria Mendes Viana; Ronaldo Martins; Andre S. Vieira; José Carlos Alves-Filho; Eurico de Arruda Neto; Adriano Sebollela; Fernando Cendes; Fernando Q Cunha Sr.; André Damásio; Marco Aurélio Ramirez Vinolo; Carolina Demarchi Munhoz; Stevens K Rehen Sr.; Thais Mauad; Amaro Nunes Duarte-Neto; Luiz Fernando Ferraz da Silva; Marisa Dolhnikoff; Paulo Saldiva; Alexandre Todorovic Fabro; Alessandro S Farias; Pedro Manoel M. Moraes-Vieira; José Luiz Proença Módena; Clarissa Lin Yasuda; Marcelo A. Mori; Thiago Mattar Cunha; Daniel Martins-de-Souza.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.10.09.20207464

ABSTRACT

COVID-19 patients may exhibit neuropsychiatric and/or neurological symptoms. We found that anxiety and cognitive impairment are manifested by 28-56% of SARS-CoV-2-infected individuals with mild or no respiratory symptoms and are associated with altered cerebral cortical thickness. Using an independent cohort, we found histopathological signs of brain damage in 19% of individuals who died of COVID-19. All of the affected brain tissues exhibited foci of SARS-CoV-2 infection, particularly in astrocytes. Infection of neural stem cell-derived astrocytes changed energy metabolism, altered key proteins and metabolites used to fuel neurons and for biogenesis of neurotransmitters, and elicited a secretory phenotype that reduces neuronal viability. Our data support the model where SARS-CoV-2 reaches the brain, infects astrocytes and triggers neuropathological changes that contribute to the structural and functional alterations in the brain of COVID-19 patients.


Subject(s)
Anxiety Disorders , Brain Injury, Chronic , Astrocytoma , Severe Acute Respiratory Syndrome , COVID-19 , Cognition Disorders
5.
Jordan J. Clark; Rebekah Penrice-Randal; Parul Sharma; Anja Kipar; Xiaofeng Dong; Andrew D. Davidson; Maia Kavanagh Williamson; David A Matthews; Lance Turtle; Tessa Prince; Grant Hughes; Edward I Patterson; Krishanthi Subramaniam; Jo Sharp; Lynn McLaughlin; En-Min Zhou; Joseph D Turner; Amy E Marriott; Stefano Colombo; Shaun Pennington; Giancarlo Biagini; Andrew Owen; Julian Alexander Hiscox; James P Stewart; Jinghe Huang; Auke C Reidinga; Daisy Rusch; Kim CE Sigaloff; Renee A Douma; Lianne de Haan; Egill A Fridgeirsson; Niels C Gritters van de Oever; Roger JMW Rennenberg; Guido van Wingen; Marcel JH Aries; Martijn Beudel; ítalo Karmann Aventurato; Mariana Rabelo de Brito; Marina Koutsodontis Machado Alvim; José Roberto da Silva Junior; Lívia Liviane Damião; Maria Ercilia de Paula Castilho Stefano; Iêda Maria Pereira de Sousa; Elessandra Dias da Rocha; Solange Maria Gonçalves; Luiz Henrique Lopes da Silva; Vanessa Bettini; Brunno Machado de Campos; Guilherme Ludwig; Rosa Maria Mendes Viana; Ronaldo Martins; Andre S. Vieira; José Carlos Alves-Filho; Eurico de Arruda Neto; Adriano Sebollela; Fernando Cendes; Fernando Q Cunha Sr.; André Damásio; Marco Aurélio Ramirez Vinolo; Carolina Demarchi Munhoz; Stevens K Rehen Sr.; Thais Mauad; Amaro Nunes Duarte-Neto; Luiz Fernando Ferraz da Silva; Marisa Dolhnikoff; Paulo Saldiva; Alexandre Todorovic Fabro; Alessandro S Farias; Pedro Manoel M. Moraes-Vieira; José Luiz Proença Módena; Clarissa Lin Yasuda; Marcelo A. Mori; Thiago Mattar Cunha; Daniel Martins-de-Souza.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.13.334532

ABSTRACT

COVID-19 is a spectrum of clinical symptoms in humans caused by infection with SARS-CoV-2, a recently emerged coronavirus that has rapidly caused a pandemic. Coalescence of a second wave of this virus with seasonal respiratory viruses, particularly influenza virus is a possible global health concern. To investigate this, transgenic mice expressing the human ACE2 receptor driven by the epithelial cell cytokeratin-18 gene promoter (K18-hACE2) were first infected with IAV followed by SARS-CoV-2. The host response and effect on virus biology was compared to K18-hACE2 mice infected with IAV or SARS-CoV-2 only. Infection of mice with each individual virus resulted in a disease phenotype compared to control mice. Although, SARS-CoV-2 RNA synthesis appeared significantly reduced in the sequentially infected mice, these mice had a more rapid weight loss, more severe lung damage and a prolongation of the innate response compared to singly infected or control mice. The sequential infection also exacerbated the extrapulmonary manifestations associated with SARS-CoV-2. This included a more severe encephalitis. Taken together, the data suggest that the concept of "twinfection" is deleterious and mitigation steps should be instituted as part of a comprehensive public health response to the COVID-19 pandemic.


Subject(s)
Lung Diseases , Infections , Encephalitis , Weight Loss , COVID-19
6.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.13.308676

ABSTRACT

The recent COVID-19 pandemic has sparked a global public health crisis. Vital to the development of informed treatments for this disease is a comprehensive understanding of the molecular interactions involved in disease pathology. One lens through which we can better understand this pathology is through the network of protein-protein interactions between its viral agent, SARS-CoV-2, and its human host. For instance, increased infectivity of SARS-CoV-2 compared to SARS-CoV can be explained by rapid evolution along the interface between the Spike protein and its human receptor (ACE2) leading to increased binding affinity. Sequence divergences that modulate other protein-protein interactions may further explain differences in transmission and virulence in this novel coronavirus. To facilitate these comparisons, we combined homology-based structural modeling with the ECLAIR pipeline for interface prediction at residue resolution, and molecular docking with PyRosetta. This enabled us to compile a novel 3D structural interactome meta-analysis for the published interactome network between SARS-CoV-2 and human. This resource includes docked structures for all interactions with protein structures, enrichment analysis of variation along interfaces, predicted {Delta}{Delta}G between SARS-CoV and SARS-CoV-2 variants for each interaction, predicted impact of natural human population variation on binding affinity, and a further prioritized set of drug repurposing candidates predicted to overlap with protein interfaces. All predictions are available online for easy access and are continually updated when new interactions are published. NOTE: Some sections of this pre-print have been redacted to comply with current bioRxiv policy restricting the dissemination of purely in silico results predicting potential therapies for SARS-CoV-2 that have not undergone thorough peer-review. The results section titled 'Prioritization of Candidate Inhibitors of SARS-CoV-2-Human Interactions Through Binding Site Comparison,' Figure 4, Supplemental Table 9, and all links to our web resource have been removed. Blank headers left in place to preserve structure and item numbering. Our full manuscript will be published in an appropriate journal following peer-review.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome
7.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.12.336099

ABSTRACT

In a try to understand the pathogenesis, evolution, and epidemiology of the SARS-CoV-2 virus, scientists from all over the world are tracking its genomic changes in real-time. Genomic studies can be helpful in understanding the disease dynamics. We have downloaded 324 complete and near-complete SARS-CoV-2 genomes submitted in the GISAID database from Bangladesh which were isolated between 30 March to 7 September 2020. We then compared these genomes with the Wuhan reference sequence and found 4160 mutation events including 2253 missense single nucleotide variations, 38 deletions, and 10 insertions. The C>T nucleotide change was most prevalent possibly due to selective mutation pressure to reduce CpG sites to evade CpG targeted host immune response. The most frequent mutation that occurred in 98% of the isolates was 3037C>T which is a synonymous change that almost always accompanied 3 other mutations that include 241C>T, 14408C>T (P323L in RdRp), and 23403A>G (D614G in spike protein). The P323L was reported to increase mutation rate and D614G is associated with increased viral replication and currently the most prevalent variant circulating all over the world. We identified multiple missense mutations in B-cell and T-cell predicted epitope regions and/or PCR target regions (including R203K and G204R that occurred in 86% of the isolates) that may impact immunogenicity and/or RT-PCR based diagnosis. Our analysis revealed 5 large deletion events in ORF7a and ORF8 gene products that may be associated with less severity of the disease and increased viral clearance. Our phylogeny analysis identified most of the isolates belonged to the Nextstrain clade 20B (86%) and GISAID clade GR (88%). Most of our isolates shared common ancestors either directly with European countries or jointly with middle eastern countries as well as Australia and India. Interestingly, the 19B clade (GISAID S clade) was unique to Chittagong which was originally prevalent in China. This reveals possible multiple introductions of the virus in Bangladesh via different routes. Hence more genome sequencing and analysis with related clinical data are needed to interpret the functional significance and better predict the disease dynamics that may be helpful for policymakers to control the COVID-19 pandemic in Bangladesh.


Subject(s)
COVID-19
8.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.13.336800

ABSTRACT

Neutralizing monoclonal antibodies (nAbs) to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) represent promising candidates for clinical intervention against coronavirus virus diseases 2019 (COVID-19). We isolated a large number of nAbs from SARS-CoV-2 infected individuals capable of disrupting proper interaction between the receptor binding domain (RBD) of the viral spike (S) protein and the receptor angiotensin converting enzyme 2 (ACE2). In order to understand the mechanism of these nAbs on neutralizing SARS-CoV-2 virus infections, we have performed cryo-EM analysis and here report cryo-EM structures of the ten most potent nAbs in their native full-length IgG or Fab forms bound to the trimeric S protein of SARS-CoV-2. The bivalent binding of the full-length IgG is found to associate with more RBD in the "up" conformation than the monovalent binding of Fab, perhaps contributing to the enhanced neutralizing activity of IgG and triggering more shedding of the S1 subunit from the S protein. Comparison of large number of nAbs identified common and unique structural features associated with their potent neutralizing activities. This work provides structural basis for further understanding the mechanism of nAbs, especially through revealing the bivalent binding and their correlation with more potent neutralization and the shedding of S1 subunit.


Subject(s)
Severe Acute Respiratory Syndrome , COVID-19
9.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.13.337774

ABSTRACT

An unexpected observation among the COVID-19 pandemic is that smokers constituted only 1.4-18.5% of hospitalized adults, calling for an urgent investigation to determine the role of smoking in SARS-CoV-2 infection. Here, we show that cigarette smoke extract (CSE) and carcinogen benzo(a)pyrene (BaP) increase ACE2 mRNA but trigger ACE2 protein catabolism. BaP induces an aryl hydrocarbon receptor (AhR)-dependent upregulation of the ubiquitin E3 ligase Skp2 for ACE2 ubiquitination. ACE2 in lung tissues of non-smokers is higher than in smokers, consistent with the findings that tobacco carcinogens downregulate ACE2 in mice. Tobacco carcinogens inhibit SARS-CoV-2 Spike protein pseudovirions infection of the cells. Given that tobacco smoke accounts for 8 million deaths including 2.1 million cancer deaths annually and Skp2 is an oncoprotein, tobacco use should not be recommended and cessation plan should be prepared for smokers in COVID-19 pandemic.


Subject(s)
COVID-19 , Severe Acute Respiratory Syndrome
10.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.10.13.337980

ABSTRACT

Substitution for aspartic acid by glycine at position 614 in the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the ongoing pandemic, appears to facilitate rapid viral spread. The G614 variant has now replaced the D614-carrying virus as the dominant circulating strain. We report here cryo-EM structures of a full-length S trimer carrying G614, which adopts three distinct prefusion conformations differing primarily by the position of one receptor-binding domain (RBD). A loop disordered in the D614 S trimer wedges between domains within a protomer in the G614 spike. This added interaction appears to prevent premature dissociation of the G614 trimer, effectively increasing the number of functional spikes and enhancing infectivity. The loop transition may also modulate structural rearrangements of S protein required for membrane fusion. These findings extend our understanding of viral entry and suggest an improved immunogen for vaccine development.

11.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.09.25.20200329

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the agent of a major global outbreak of respiratory tract disease known as coronavirus disease-2019 (COVID-19). SARS-CoV-2 infects the lungs and may cause several immune-related complications such as lymphocytopenia and cytokine storm which are associated with the severity of the disease and predict mortality . The mechanism by which SARS-CoV-2 infection may result in immune system dysfunction is not fully understood. Here we show that SARS-CoV-2 infects human CD4+ T helper cells, but not CD8+ T cells, and is present in blood and bronchoalveolar lavage T helper cells of severe COVID-19 patients. We demonstrated that SARS-CoV-2 spike glycoprotein (S) directly binds to the CD4 molecule, which in turn mediates the entry of SARS- CoV-2 in T helper cells in a mechanism that also requires ACE2 and TMPRSS2. Once inside T helper cells, SARS-CoV-2 assembles viral factories, impairs cell function and may cause cell death. SARS-CoV-2 infected T helper cells express higher amounts of IL-10, which is associated with viral persistence and disease severity. Thus, CD4-mediated SARS-CoV-2 infection of T helper cells may explain the poor adaptive immune response of many COVID- 19 patients.


Subject(s)
Ataxia Telangiectasia , Severe Acute Respiratory Syndrome , COVID-19 , Lymphopenia
12.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.09.27.315796

ABSTRACT

The current COVID-19 pandemic has a devastating global impact and is caused by the SARS-CoV-2 virus. SARS-CoV-2 attaches to human host cells through interaction of its receptor binding domain (RBD) located on the viral Spike (S) glycoprotein with angiotensin converting enzyme-2 (ACE2) on the surface of host cells. RBD binding to ACE2 is a critical first step in SARS-CoV-2 infection. Viral attachment occurs in dynamic environments where forces act on the binding partners and multivalent interactions play central roles, creating an urgent need for assays that can quantitate SARS-CoV-2 interactions with ACE2 under mechanical load and in defined geometries. Here, we introduce a tethered ligand assay that comprises the RBD and the ACE2 ectodomain joined by a flexible peptide linker. Using specific molecular handles, we tether the fusion proteins between a functionalized flow cell surface and magnetic beads in magnetic tweezers. We observe repeated interactions of RBD and ACE2 under constant loads and can fully quantify the force dependence and kinetics of the binding interaction. Our results suggest that the SARS-CoV-2 ACE2 interaction has higher mechanical stability, a larger free energy of binding, and a lower off-rate than that of SARS-CoV-1, the causative agents of the 2002-2004 SARS outbreak. In the absence of force, the SARS-CoV-2 RBD rapidly (within [≤]1 ms) engages the ACE2 receptor if held in close proximity and remains bound to ACE2 for 400-800 s, much longer than what has been reported for other viruses engaging their cellular receptors. We anticipate that our assay will be a powerful tool investigate the roles of mutations in the RBD that might alter the infectivity of the virus and to test the modes of action of neutralizing antibodies and other agents designed to block RBD binding to ACE2 that are currently developed as potential COVID-19 therapeutics.


Subject(s)
COVID-19
13.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.09.27.316174

ABSTRACT

SARS-CoV-2 neutralizing antibodies represent an important component of the ongoing search for effective treatment of and protection against COVID-19. We report here on the use of a naive phage display antibody library to identify a panel of fully human SARS-CoV-2 neutralizing antibodies. Following functional profiling in vitro against an early pandemic isolate as well as a recently emerged isolate bearing the D614G Spike mutation, the clinical candidate antibody, STI-1499, and the affinity-engineered variant, STI-2020, were evaluated for in vivo efficacy in the Syrian golden hamster model of COVID-19. Both antibodies demonstrated potent protection against the pathogenic effects of the disease and a dose-dependent reduction of virus load in the lungs, reaching undetectable levels following a single dose of 500 micrograms of STI-2020. These data support continued development of these antibodies as therapeutics against COVID-19 and future use of this approach to address novel emerging pandemic disease threats.


Subject(s)
COVID-19 , Emergencies
14.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.09.27.312538

ABSTRACT

A recent study by Wilk et al. of the transcriptome of peripheral blood mononuclear cells (PBMCs) in seven patients hospitalized with COVID-19 described a population of 'developing neutrophils' that were 'phenotypically related by dimensionality reduction' to plasmablasts, and that these two cell populations represent a 'linear continuum of cellular phenotype'. The authors suggest that, in the setting of acute respiratory distress syndrome (ARDS) secondary to severe COVID-19, a 'differentiation bridge from plasmablasts to developing neutrophils' connected these distantly related cell types. This conclusion is controversial as it appears to violate several basic principles in cell biology relating to cell lineage identity and fidelity. Correctly classifying cells and their developmental history is an important issue in cell biology and we suggest that this conclusion is not supported by the data as we show here that: (1) regressing out covariates such as unique molecular identifiers (UMIs) can lead to overfitting; and (2) that UMAP embeddings may reflect the expression of similar genes but not necessarily direct cell lineage relationships.


Subject(s)
COVID-19 , Respiratory Distress Syndrome
15.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.09.28.316281

ABSTRACT

The ongoing COVID-19 pandemic is responsible for worldwide economic damage and nearly one million deaths. Potent drugs for the treatment of severe SARS-CoV-2 infections are not yet available. To identify host factors that support coronavirus infection, we performed genome-wide functional genetic screens with SARS-CoV-2 and the common cold virus HCoV-229E in non-transgenic human cells. These screens identified PI3K type 3 as a potential drug target against multiple coronaviruses. We discovered that the lysosomal protein TMEM106B is an important host factor for SARS-CoV-2 infection. Furthermore, we show that TMEM106B is required for replication in multiple human cell lines derived from liver and lung and is expressed in relevant cell types in the human airways. Our results identify new coronavirus host factors that may potentially serve as drug targets against SARS-CoV-2 or to quickly combat future zoonotic coronavirus outbreaks.


Subject(s)
Coronavirus Infections , Severe Acute Respiratory Syndrome , COVID-19
16.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.09.28.316307

ABSTRACT

Membrane proteins play numerous physiological roles and are thus of tremendous interest in pharmacology. Nevertheless, stable and homogeneous sample preparation is one of the bottlenecks in biophysical and pharmacological studies of membrane proteins because membrane proteins are typically unstable and poorly expressed. To overcome such obstacles, GFP fusion-based Fluorescence-detection Size-Exclusion Chromatography (FSEC) has been widely employed for membrane protein expression screening for over a decade. However, fused GFP itself may occasionally affect the expression and/or stability of the targeted membrane protein, leading to both false-positive and false-negative results in expression screening. Furthermore, GFP fusion technology is not well suited for some membrane proteins depending on their membrane topology. Here, we developed an FSEC assay utilizing nanobody (Nb) technology, named FSEC-Nb, in which targeted membrane proteins are fused to a small peptide tag and recombinantly expressed. The whole-cell extracts are solubilized, mixed with anti-peptide Nb fused to GFP and applied to a size-exclusion chromatography column attached to a fluorescence detector for FSEC analysis. FSEC-Nb enables one to evaluate the expression, monodispersity and thermostability of membrane proteins without the need of purification by utilizing the benefits of the GFP fusion-based FSEC method, but does not require direct GFP fusion to targeted proteins. We applied FSEC-Nb to screen zinc-activated ion channel (ZAC) family proteins in the Cys-loop superfamily and membrane proteins from SARS-CoV-2 as examples of the practical application of FSEC-Nb. We successfully identified a ZAC ortholog with high monodispersity but moderate expression levels that could not be identified with the previously developed GFP fusion-free FSEC method. Consistent with the results of FSEC-Nb screening, the purified ZAC ortholog showed monodispersed particles by both negative staining EM and cryo-EM. Furthermore, we identified two membrane proteins from SARS-CoV-2 with high monodispersity and expression level by FSEC-Nb, which may facilitate structural and functional studies of SARS-CoV-2. Overall, our results show FSEC-Nb as a powerful tool for membrane protein expression screening that can provide further opportunity to prepare well-behaved membrane proteins for structural and functional studies.

17.
ssrn; 2020.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3606770

ABSTRACT

COVID-19 can result in severe lung injury. It remained to be determined why diabetic individuals with uncontrolled glucose levels are more prone to develop the severe form of COVID-19. The molecular mechanism underlying SARS-CoV-2 infection and what determines the onset of the cytokine storm found in severe COVID-19 patients are unknown. Monocytes/macrophages are the most enriched immune cell types in the lungs of COVID-19 patients and appear to have a central role in the pathogenicity of the disease. These cells adapt their metabolism upon infection and become highly glycolytic, which facilitates SARS-CoV-2 replication. The infection triggers mitochondrial ROS production, which induces stabilization of hypoxia-inducible factor- 1α (HIF - 1α) and consequently promotes glycolysis. HIF- 1α-induced changes in monocyte metabolism by SARS-CoV-2 infection directly inhibit T cell response and reduce epithelial cell survival. Targeting HIF-1 ɑ may have great therapeutic potential for the development of novel drugs to treat COVID-19.


Subject(s)
COVID-19 , Lung Injury
SELECTION OF CITATIONS
SEARCH DETAIL