Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
J Comp Pathol ; 212: 27-31, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968671

ABSTRACT

The tissue tropism and the wide host range of influenza A viruses are determined by the presence of sialic acid (SA) α2,3-Gal and SA α2,6-Gal receptors. Recent studies have shown that animals possessing both receptors allow for the rearrangement and emergence of new viral strains of public health importance. This study aimed to evaluate the expression and distribution of human and avian influenza A receptors in nine Neotropical snake species using lectin immunohistochemistry. We selected 17 snakes that were examined post mortem at the Veterinary Pathology Sector of the Universidade Federal de Minas Gerais between 2019 and 2023. Sections of nasal turbinate, trachea, lung, oral mucosa, stomach and intestine were subjected to immunohistochemical analysis using the lectins Maackia amurensis and Sambucus nigra. This research detected, for the first time, co-expression of SA α2,3-Gal and SA α2,6-Gal receptors in the respiratory and digestive tracts of snakes, indicating the possible susceptibility of these species to influenza A virus of avian and human origin. Consequently, snakes can be considered important species for monitoring influenza A in wild, urban and peri-urban environments. More studies should be conducted to investigate the role of snakes in influenza A epidemiology.


Subject(s)
Influenza A virus , Receptors, Cell Surface , Snakes , Animals , Receptors, Cell Surface/metabolism , Orthomyxoviridae Infections/veterinary , Receptors, Virus/metabolism
2.
FASEB J ; 38(10): e23691, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38780525

ABSTRACT

Heme is a prosthetic group of proteins involved in vital physiological processes. It participates, for example, in redox reactions crucial for cell metabolism due to the variable oxidation state of its central iron atom. However, excessive heme can be cytotoxic due to its prooxidant properties. Therefore, the control of intracellular heme levels ensures the survival of organisms, especially those that deal with high concentrations of heme during their lives, such as hematophagous insects. The export of heme initially attributed to the feline leukemia virus C receptor (FLVCR) has recently been called into question, following the discovery of choline uptake by the same receptor in mammals. Here, we found that RpFLVCR is a heme exporter in the midgut of the hematophagous insect Rhodnius prolixus, a vector for Chagas disease. Silencing RpFLVCR decreased hemolymphatic heme levels and increased the levels of intracellular dicysteinyl-biliverdin, indicating heme retention inside midgut cells. FLVCR silencing led to increased expression of heme oxygenase (HO), ferritin, and mitoferrin mRNAs while downregulating the iron importers Malvolio 1 and 2. In contrast, HO gene silencing increased FLVCR and Malvolio expression and downregulated ferritin, revealing crosstalk between heme degradation/export and iron transport/storage pathways. Furthermore, RpFLVCR silencing strongly increased oxidant production and lipid peroxidation, reduced cytochrome c oxidase activity, and activated mitochondrial biogenesis, effects not observed in RpHO-silenced insects. These data support FLVCR function as a heme exporter, playing a pivotal role in heme/iron metabolism and maintenance of redox balance, especially in an organism adapted to face extremely high concentrations of heme.


Subject(s)
Heme , Mitochondria , Oxidation-Reduction , Rhodnius , Animals , Heme/metabolism , Rhodnius/metabolism , Mitochondria/metabolism , Receptors, Virus/metabolism , Receptors, Virus/genetics , Leukemia Virus, Feline/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics
3.
J Mol Evol ; 92(3): 329-337, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777906

ABSTRACT

The spike protein determines the host-range specificity of coronaviruses. In particular, the Receptor-Binding Motif in the spike protein from SARS-CoV-2 contains the amino acids involved in molecular recognition of the host Angiotensin Converting Enzyme 2. Therefore, to understand how SARS-CoV-2 acquired its capacity to infect humans it is necessary to reconstruct the evolution of this important motif. Early during the pandemic, it was proposed that the SARS-CoV-2 Receptor-Binding Domain was acquired via recombination with a pangolin infecting coronavirus. This proposal was challenged by an alternative explanation that suggested that the Receptor-Binding Domain from SARS-CoV-2 did not originated via recombination with a coronavirus from a pangolin. Instead, this alternative hypothesis proposed that the Receptor-Binding Motif from the bat coronavirus RaTG13, was acquired via recombination with an unidentified coronavirus. And as a consequence of this event, the Receptor-Binding Domain from the pangolin coronavirus appeared as phylogenetically closer to SARS-CoV-2. Recently, the genomes from coronaviruses from Cambodia (bat_RShST182/200) and Laos (BANAL-20-52/103/247) which are closely related to SARS-CoV-2 were reported. However, no detailed analysis of the evolution of the Receptor-Binding Motif from these coronaviruses was reported. Here we revisit the evolution of the Receptor-Binding Domain and Motif in the light of the novel coronavirus genome sequences. Specifically, we wanted to test whether the above coronaviruses from Cambodia and Laos were the source of the Receptor-Binding Domain from RaTG13. We found that the Receptor-Binding Motif from these coronaviruses is phylogenetically closer to SARS-CoV-2 than to RaTG13. Therefore, the source of the Receptor-Binding Domain from RaTG13 is still unidentified. In accordance with previous studies, our results are consistent with the hypothesis that the Receptor-Binding Motif from SARS-CoV-2 evolved by vertical inheritance from a bat-infecting population of coronaviruses.


Subject(s)
Evolution, Molecular , Phylogeny , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Humans , Animals , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/chemistry , Amino Acid Motifs , COVID-19/virology , Protein Binding , Betacoronavirus/genetics , Chiroptera/virology , Pangolins/virology , Binding Sites , Genome, Viral , Receptors, Virus/metabolism , Receptors, Virus/genetics , Receptors, Virus/chemistry
4.
J Mol Biol ; 436(11): 168577, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38642883

ABSTRACT

The Red Queen Hypothesis (RQH), derived from Lewis Carroll's "Through the Looking-Glass", postulates that organisms must continually adapt in response to each other to maintain relative fitness. Within the context of host-pathogen interactions, the RQH implies an evolutionary arms race, wherein viruses evolve to exploit hosts and hosts evolve to resist viral invasion. This study delves into the dynamics of the RQH in the context of virus-cell interactions, specifically focusing on virus receptors and cell receptors. We observed multiple virus-host systems and noted patterns of co-evolution. As viruses evolved receptor-binding proteins to effectively engage with cell receptors, cells countered by altering their receptor genes. This ongoing mutual adaptation cycle has influenced the molecular intricacies of receptor-ligand interactions. Our data supports the RQH as a driving force behind the diversification and specialization of both viral and host cell receptors. Understanding this co-evolutionary dance offers insights into the unpredictability of emerging viral diseases and potential therapeutic interventions. Future research is crucial to dissect the nuanced molecular changes and the broader ecological consequences of this ever-evolving battle. Here, we combine phylogenetic inferences, structural modeling, and molecular dynamics analyses to describe the epidemiological characteristics of major Brazilian DENV strains that circulated from 1990 to 2022 from a combined perspective, thus providing us with a more detailed picture on the dynamics of such interactions over time.


Subject(s)
Cell Adhesion Molecules , Dengue Virus , Evolution, Molecular , Host-Pathogen Interactions , Receptors, Cell Surface , Viral Envelope Proteins , Viral Envelope , Humans , Brazil , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/chemistry , Dengue/virology , Dengue Virus/genetics , Dengue Virus/metabolism , Host-Pathogen Interactions/genetics , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/chemistry , Molecular Dynamics Simulation , Phylogeny , Protein Binding , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/chemistry , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Viral Envelope/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/chemistry
5.
Int J Mol Sci ; 23(17)2022 Aug 30.
Article in English | MEDLINE | ID: mdl-36077240

ABSTRACT

Sialic acids and heparan sulfates make up the outermost part of the cell membrane and the extracellular matrix. Both structures are characterized by being negatively charged, serving as receptors for various pathogens, and are highly expressed in the respiratory and digestive tracts. Numerous viruses use heparan sulfates as receptors to infect cells; in this group are HSV, HPV, and SARS-CoV-2. Other viruses require the cell to express sialic acids, as is the case in influenza A viruses and adenoviruses. This review aims to present, in a general way, the participation of glycoconjugates in viral entry, and therapeutic strategies focused on inhibiting the interaction between the virus and the glycoconjugates. Interestingly, there are few studies that suggest the participation of both glycoconjugates in the viruses addressed here. Considering the biological redundancy that exists between heparan sulfates and sialic acids, we propose that it is important to jointly evaluate and design strategies that contemplate inhibiting the interactions of both glycoconjugates. This approach will allow identifying new receptors and lead to a deeper understanding of interspecies transmission.


Subject(s)
COVID-19 , Viruses , Glycoconjugates/metabolism , Heparitin Sulfate/metabolism , Humans , N-Acetylneuraminic Acid/metabolism , Receptors, Virus/metabolism , SARS-CoV-2 , Sialic Acids/metabolism , Sulfates , Virus Attachment , Viruses/metabolism
6.
Front Immunol ; 13: 871874, 2022.
Article in English | MEDLINE | ID: mdl-35898497

ABSTRACT

The COVID-19 pandemic caused by the severe acute syndrome virus 2 (SARS-CoV-2) has been around since November 2019. As of early June 2022, more than 527 million cases were diagnosed, with more than 6.0 million deaths due to this disease. Coronaviruses accumulate mutations and generate greater diversity through recombination when variants with different mutations infect the same host. Consequently, this virus is predisposed to constant and diverse mutations. The SARS-CoV-2 variants of concern/interest (VOCs/VOIs) such as Alpha (B.1.1.7), Beta (B.1.351), Gamma (B.1.1.28/P.1), Delta (B.1.617.2), and Omicron (B.1.1.529) have quickly spread across the world. These VOCs and VOIs have accumulated mutations within the spike protein receptor-binding domain (RBD) which interacts with the angiotensin-2 converting enzyme (ACE-2) receptor, increasing cell entry and infection. The RBD region is the main target for neutralizing antibodies; however, other notable mutations have been reported to enhance COVID-19 infectivity and lethality. Considering the urgent need for alternative therapies against this virus, an anti-SARS-CoV-2 equine immunoglobulin F(ab')2, called ECIG, was developed by the Butantan Institute using the whole gamma-irradiated SARS-CoV-2 virus. Surface plasmon resonance experiments revealed that ECIG binds to wild-type and mutated RBD, S1+S2 domains, and nucleocapsid proteins of known VOCs, including Alpha, Gamma, Beta, Delta, Delta Plus, and Omicron. Additionally, it was observed that ECIG attenuates the binding of RBD (wild-type, Beta, and Omicron) to human ACE-2, suggesting that it could prevent viral entry into the host cell. Furthermore, the ability to concomitantly bind to the wild-type and mutated nucleocapsid protein likely enhances its neutralizing activity of SARS-CoV-2. We postulate that ECIG benefits COVID-19 patients by reducing the infectivity of the original virus and existing variants and may be effective against future ones. Impacting the course of the disease, mainly in the more vulnerable, reduces infection time and limits the appearance of new variants by new recombination.


Subject(s)
COVID-19 , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Animals , Antibodies, Viral , Horses , Humans , Nucleocapsid Proteins , Pandemics , Receptors, Virus/metabolism , Spike Glycoprotein, Coronavirus
8.
Nature ; 598(7882): 677-681, 2021 10.
Article in English | MEDLINE | ID: mdl-34646021

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an enveloped RNA virus that causes encephalitis and potentially mortality in infected humans and equines1. At present, no vaccines or drugs are available that prevent or cure diseases caused by VEEV. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) was recently identified as a receptor for the entry of VEEV into host cells2. Here we present the cryo-electron microscopy structure of the LDLRAD3 extracellular domain 1 (LDLRAD3-D1) in complex with VEEV virus-like particles at a resolution of 3.0 Å. LDLRAD3-D1 has a cork-like structure and is inserted into clefts formed between adjacent VEEV E2-E1 heterodimers in the viral-surface trimer spikes through hydrophobic and polar contacts. Mutagenesis studies of LDLRAD3-D1 identified residues that are involved in the key interactions with VEEV. Of note, some of the LDLRAD3-D1 mutants showed a significantly increased binding affinity for VEEV, suggesting that LDLRAD3-D1 may serve as a potential scaffold for the development of inhibitors of VEEV entry. Our structures provide insights into alphavirus assembly and the binding of receptors to alphaviruses, which may guide the development of therapeutic countermeasures against alphaviruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/chemistry , Receptors, LDL/chemistry , Receptors, Virus/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Structure, Secondary , Virus Internalization
9.
Nature ; 598(7882): 672-676, 2021 10.
Article in English | MEDLINE | ID: mdl-34646020

ABSTRACT

LDLRAD3 is a recently defined attachment and entry receptor for Venezuelan equine encephalitis virus (VEEV)1, a New World alphavirus that causes severe neurological disease in humans. Here we present near-atomic-resolution cryo-electron microscopy reconstructions of VEEV virus-like particles alone and in a complex with the ectodomains of LDLRAD3. Domain 1 of LDLRAD3 is a low-density lipoprotein receptor type-A module that binds to VEEV by wedging into a cleft created by two adjacent E2-E1 heterodimers in one trimeric spike, and engages domains A and B of E2 and the fusion loop in E1. Atomic modelling of this interface is supported by mutagenesis and anti-VEEV antibody binding competition assays. Notably, VEEV engages LDLRAD3 in a manner that is similar to the way that arthritogenic alphaviruses bind to the structurally unrelated MXRA8 receptor, but with a much smaller interface. These studies further elucidate the structural basis of alphavirus-receptor interactions, which could inform the development of therapies to mitigate infection and disease against multiple members of this family.


Subject(s)
Encephalitis Virus, Venezuelan Equine/chemistry , Receptors, LDL/chemistry , Receptors, Virus/chemistry , Amino Acid Sequence , Animals , Cell Line , Cryoelectron Microscopy , Humans , Mice , Models, Molecular , Protein Structure, Secondary , Sequence Alignment , Virus Internalization
10.
Viruses ; 13(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204224

ABSTRACT

Pestivirus envelope protein E2 is crucial to virus infection and accomplishes virus-receptor interaction during entry. However, mapping of E2 residues mediating these interactions has remained unexplored. In this study, to investigate the structure-function relationship for a ß-hairpin motif exposed to the solvent in the crystal structure of bovine viral diarrhea virus (BVDV) E2, we designed two amino acidic substitutions that result in a change of electrostatic potential. First, using wild type and mutant E2 expressed as soluble recombinant proteins, we found that the mutant protein had reduced binding to susceptible cells compared to wild type and diminished ability to inhibit BVDV infection, suggesting a lower affinity for BVDV receptors. We then analyzed the effect of ß-hairpin mutations in the context of recombinant viral particles. Mutant viruses recovered from cell culture supernatant after transfection of recombinant RNA had almost completely inhibited ability to re-infect susceptible cells, indicating an impact of mutations on BVDV infectivity. Finally, sequential passaging of the mutant virus resulted in the selection of a viral population in which ß-hairpin mutations reverted to the wild type sequence to restore infectivity. Taken together, our results show that this conserved region of the E2 protein is critical for the interaction with host cell receptors.


Subject(s)
Diarrhea Viruses, Bovine Viral/genetics , Diarrhea Viruses, Bovine Viral/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization , Amino Acid Substitution , Animals , Cattle , Cell Line , Diarrhea Viruses, Bovine Viral/chemistry , Inverted Repeat Sequences/physiology , Protein Binding , Viral Envelope Proteins/genetics
11.
J Mol Model ; 27(8): 222, 2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34236527

ABSTRACT

The crescent evolution of a global pandemic COVID-19 and its respiratory syndrome (SARS-Cov-2) has been a constant concern (Ghosh 2021; Khan et al. 2021; Alazmi and Motwalli 2020; Vargas et al. 2020). The absence of a proven and effective medication has compelled all the scientific community to search for a new drug. The use of known drugs is a faster way to develop new therapies. Molecular docking is a powerful tool (Gao et al. J Mol Model 10: 44-54, 2004; Singh et al. J Mol Model 18: 39-51, 2012; Schulz-Gasch and Stahl J Mol Model 9:47-57, 2003) to study the interaction of potential drugs with SARS-CoV-2, Alsalme et al. (2020) and Sanders et al. (2020) spike protein as a consequence the main goal of this article is to present the result of the study of an interaction between (R and S)-Linezolid with receptor-binding domain (RBD) of SARS-Cov-2 spike protein complexed with human Angiostensin-converting enzyme 2 (ACE2) (6vW1 - from PDB). The Linezolid enantiomers were optimized at B3LYP/6-311++G(2d,p) level of theory. Molecular docking of the system (S)-Linezolid⋯RBD⋯ACE2 and (R)-Linezolid⋯RBD⋯ACE2 was performed, the analysis was made using LigPlot+ and NCIplot software packages, to understand the intermolecular interactions. The UV-Vis and ECD of the complexes - (R and S)-Linezolid⋯RBD⋯ACE2 were performed in two layers with DFT/6-311++G(3df,2p) and DFT/6-31G(d), respectively. The results showed that only the (S)-Linezolid had a stable interaction with - 8.05 kcal.mol- 1, whereas all the R-enantiomeric configurations had positive values of binding energy. The (S)-Linezolid had the same interactions as in the (S)-Linezolid ⋯ Haluarcula morismortui Ribosomal system, where it is well-known the fact that the latter has biological activity. A specific interaction on the fluorine ring justified an attenuation on the ECD signal, in comparison to isolated species. Therefore, some biological activity of (S)-Linezolid with SARS-CoV-2 RBD was expected, indicated by the modification of its ECD signal and justified by a similar interaction in the S-Linezolid⋯Haluarcula marismortui Ribosomal system.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Linezolid/pharmacology , Molecular Docking Simulation , SARS-CoV-2/drug effects , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/metabolism , Binding Sites , COVID-19/virology , Host-Pathogen Interactions , Humans , Kinetics , Linezolid/metabolism , Protein Binding , Protein Conformation , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Structure-Activity Relationship
12.
Curr Opin Virol ; 49: 164-175, 2021 08.
Article in English | MEDLINE | ID: mdl-34171540

ABSTRACT

The flavivirus are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than 70 viruses, and despite genomic and structural similarities, infections by different flaviviruses result in different clinical presentations. In the absence of a safe and effective vaccine against these infections, the search for new strategies to inhibit viral infection is necessary. The life cycle of arboviruses begins with the entry process composed of multiple steps: attachment, internalization, endosomal escape and capsid uncoating. This mini-review describes factors and mechanisms involved in the viral entry as events required to take over the cellular machinery and host factors and cellular pathways commonly used by flaviviruses as possible approaches for developing broad-spectrum antiviral drugs.


Subject(s)
Flavivirus Infections/virology , Flavivirus/physiology , Virus Internalization , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Endocytosis , Flavivirus/drug effects , Flavivirus/pathogenicity , Flavivirus Infections/drug therapy , Host-Pathogen Interactions , Humans , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Virus Replication
13.
IUBMB Life ; 73(6): 843-854, 2021 06.
Article in English | MEDLINE | ID: mdl-33960608

ABSTRACT

The 78 kDa glucose-regulated protein (GRP78) is an endoplasmic reticulum (ER)-resident molecular chaperone. GRP78 is a member of the 70 kDa heat shock family of proteins involved in correcting and clearing misfolded proteins in the ER. In response to cellular stress, GRP78 escapes from the ER and moves to the plasma membrane where it (a) functions as a receptor for many ligands, and (b) behaves as an autoantigen for autoantibodies that contribute to human disease and cancer. Cell surface GRP78 (csGRP78) associates with the major histocompatibility complex class I (MHC-I), and is the port of entry for several viruses, including the predictive binding of the novel SARS-CoV-2. Furthermore, csGRP78 is found in association with partners as diverse as the teratocarcinoma-derived growth factor 1 (Cripto), the melanocortin-4 receptor (MC4R) and the DnaJ-like protein MTJ-1. CsGRP78 also serves as a receptor for a large variety of ligands including activated α2 -macroglobulin (α2 M*), plasminogen kringle 5 (K5), microplasminogen, the voltage-dependent anion channel (VDAC), tissue factor (TF), and the prostate apoptosis response-4 protein (Par-4). In this review, we discuss the mechanisms involved in the translocation of GRP78 from the ER to the cell surface, and the role of secreted GRP78 and its autoantibodies in cancer and neurological disorders.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , COVID-19/transmission , Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Receptors, Virus/physiology , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases of the Nervous System/metabolism , Cell Survival , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Exosomes , GPI-Linked Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/immunology , Humans , Ligands , Neoplasm Invasiveness , Neoplasm Proteins/immunology , Nerve Tissue Proteins/immunology , Protein Domains , Protein Transport , Signal Transduction , Tumor Microenvironment , Unfolded Protein Response/physiology , Virus Internalization
14.
Biochem Biophys Res Commun ; 562: 89-93, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34049205

ABSTRACT

New SARS-CoV-2 variants emerged in the United Kingdom and South Africa in December 2020 in concomitant with the Brazillian variant in February 2021 (B.1.1.248 lineage) and currently sparking worldwide during the last few months. The new strain 501.V2 in South Africa bears three mutations in the spike receptor-binding domain (RBD); K417 N, E484K, and N501Y, while the Brazilian B.1.1.248 lineage has 12 mutations. In the current study, we simulate the complex ACE2-SARS-CoV-2 spike RBD system in which the RBD is in the wild-type and mutated isoforms. Additionally, the cell-surface Glucose Regulated Protein 78 (CS-GRP78) associated with the ACE2-SARS-CoV-2 spike RBD complex (ACE2-S RBD) is modeled at the presence of these mutant variants of the viral spike. The results showed that E484K and N501Y are critical in viral spike recognition through either ACE2 or CS-GRP78. The mutated variants (the UK, South African, and Brazilian) of the spike RBD tightly bind to GRP78 more than in the case of the wild-type RBD. These results point to the potent role of GRP78 with ACE2 in the attachment of the new variants, which could be a key for the design of inhibitors to block SARS-CoV-2 attachment and entry to the host cell.


Subject(s)
Computer Simulation , Heat-Shock Proteins/metabolism , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Brazil , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/chemistry , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Receptors, Virus/chemistry , Receptors, Virus/metabolism , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , South Africa , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Substrate Specificity , United Kingdom , Virus Internalization
15.
Curr Med Chem ; 28(34): 7157-7184, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-33820514

ABSTRACT

OBJECTIVE: This review aims to study the receptor's family and functions most related to COVID-19 infection and also suggest the tissue and cell location on which the majority of COVID-19 receptors are mainly expressed. METHODS: This systematic review is according to PRISMA guidelines. PubMed, Cochrane, SciELO, Lilacs, Web of Science, and DOAJ databases were used. Clinical trials and research articles studying receptors related to COVID-19 were included in this review. R programming language was used to elaborate charts and receptors network, and SPSS(26v) software was used to perform statistical analysis (PROSPERO: CRD42020210643). RESULTS: The majority of studies on the involvement of receptors in COVID-19 included plasma receptors and G protein-coupled receptor families (p<0.05). These receptors are highly expressed in the brain (24%) and 80% of them can interact with each other in a protein network, exerting some regulatory effects on various tissues. The main influential receptor in the network of receptors involved in the COVID-19 was the EGFR and the majority of receptors were associated with pathological processes of the disease (p<0.05), including the amplification of inflammatory responses in COVID-19, which may be related to neurological disorders in some cases. Studies on receptors involved in the COVID-19 included mainly patients from the United States, Spain, and Brazil (p<0.05). CONCLUSION: Plasma receptors and G protein-coupled receptors, especially the EGFR, involved in pathological effects of the COVID-19 inflammatory process in the brain have shown significant importance in this review.


Subject(s)
COVID-19 , Nervous System Diseases , Receptors, G-Protein-Coupled , Receptors, Virus , Brain , Humans , Nervous System Diseases/virology
16.
Infect Genet Evol ; 91: 104832, 2021 07.
Article in English | MEDLINE | ID: mdl-33812037

ABSTRACT

MicroRNAs are gene expression regulators, associated with several human pathologies, including the ones caused by virus infections. Although their role in infection diseases is not completely known, they can exert double functions in the infected cell, by mediating the virus infection and/or regulating the immunity-related gene targets through complex networks of virus-host cell interactions. In this systematic review, the Pubmed, EMBASE, Scopus, Lilacs, Scielo, and EBSCO databases were searched for research articles published until October 22nd, 2020 that focused on describing the role, function, and/or association of miRNAs in SARS-CoV-2 human infection and COVID-19. Following the PRISMA 2009 protocol, 29 original research articles were selected. Most of the studies reported miRNA data based on the genome sequencing of SARS-CoV-2 isolates and computational prediction analysis. The latter predicted, by at least one independent study, 1266 host miRNAs to target the viral genome. Thirteen miRNAs were identified by four independent studies to target SARS-CoV-2 specific genes, suggested to act by interfering with their cleavage and/or translation process. The studies selected also reported on viral and host miRNAs that targeted host genes, on the expression levels of miRNAs in biological specimens of COVID-19 patients, and on the impact of viral genome mutations on miRNA function. Also, miRNAs that regulate the expression levels of the ACE2 and TMPRSS2 proteins, which are critical for the virus entrance in the host cells, were reported. In conclusion, despite the limited number of studies identified, based on the search terms and eligibility criteria applied, this systematic review provides evidence on the impact of miRNAs on SARS-CoV-2 infection and COVID-19. Although most of the reported viral/host miRNAs interactions were based on in silico prediction analysis, they demonstrate the relevance of the viral/host miRNA interaction for viral activity and host responses. In addition, the identified studies highlight the potential use of miRNAs as therapeutic targets against COVID-19, and other viral human diseases (This review was registered at the International Prospective Register of Systematic Reviews (PROSPERO) database (#CRD42020199290).


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , Genome, Viral , MicroRNAs/genetics , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Spike Glycoprotein, Coronavirus/genetics , Angiotensin-Converting Enzyme 2/immunology , COVID-19/immunology , COVID-19/pathology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , MicroRNAs/classification , MicroRNAs/immunology , Mutation , Protein Binding , Receptors, Virus/genetics , Receptors, Virus/immunology , SARS-CoV-2/immunology , Serine Endopeptidases/immunology , Severity of Illness Index , Spike Glycoprotein, Coronavirus/immunology
17.
Physiol Rep ; 9(2): e14707, 2021 01.
Article in English | MEDLINE | ID: mdl-33463909

ABSTRACT

The COVID-19 has originated from Wuhan, China, in December 2019 and has been affecting the public health system, society, and economy in an unheard-of manner. There is no specific treatment or vaccine available for COVID-19. Previous data showed that men are more affected than women by COVID-19, then we hypothesized whether sex hormones could be protecting the female organism against the infection. VERO E6 cells have been commonly used as in vitro model for SARS-CoV-2 infection. In our experimental approach, we have treated VERO E6 cells with 17ß-estradiol to evaluate the modulation of SARS-CoV-2 infection in this cell line. Here we demonstrated that estrogen protein receptors ERα, ERß, and GPER1 are expressed by VERO E6 cells and could be used to study the effects of this steroid hormone. Previous and 24-hours post-infection, cells treated with 17ß-estradiol revealed a reduction in the viral load. Afterward, we found that SARS-CoV-2 infection per se results in ACE2 and TMPRSS2 increased gene expression in VERO E6-cell, which could be generating a cycle of virus infection in host cells. The estrogen treatment reduces the levels of the TMPRSS2, which are involved with SARS-CoV-2 infectiveness capacity, and hence, reducing the pathogenicity/genesis. These data suggest that estrogen could be a potential therapeutic target promoting cell protection against SARS-CoV-2. This opens new possibilities for further studies on 17ß-estradiol in human cell lines infected by SARS-CoV-2 and at least in part, explain why men developed a more severe COVID-19 compared to women.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Estradiol/pharmacology , SARS-CoV-2/drug effects , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , COVID-19/virology , Chlorocebus aethiops , Host-Pathogen Interactions , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vero Cells
18.
Chembiochem ; 22(4): 724-732, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32986926

ABSTRACT

The magnified infectious power of the SARS-CoV-2 virus compared to its precursor SARS-CoV is intimately linked to an enhanced ability in the mutated virus to find available hydrogen-bond sites in the host cells. This characteristic is acquired during virus evolution because of the selective pressure exerted at the molecular level. We pinpoint the specific residue (in the virus) to residue (in the cell) contacts during the initial recognition and binding and show that the virus⋅⋅⋅cell interaction is mainly due to an extensive network of hydrogen bonds and to a large surface of noncovalent interactions. In addition to the formal quantum characterization of bonding interactions, computation of absorption spectra for the specific virus⋅⋅⋅cell interacting residues yields significant shifts of Δλmax =47 and 66 nm in the wavelength for maximum absorption in the complex with respect to the isolated host and virus, respectively.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/prevention & control , Receptors, Virus/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Amino Acid Sequence , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , COVID-19/epidemiology , COVID-19/virology , Humans , Molecular Dynamics Simulation , Pandemics , Protein Binding , Protein Domains , Receptors, Virus/chemistry , Receptors, Virus/genetics , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics
19.
Front Immunol ; 11: 569760, 2020.
Article in English | MEDLINE | ID: mdl-33362758

ABSTRACT

The World Health Organization (WHO) announced in March a pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). This new infectious disease was named Coronavirus Disease 19 (COVID-19), and at October 2020, more than 39,000,000 cases of SARS-CoV-2 have been detected worldwide leading to near 1,100,000 deaths. Clinically, COVID-19 is characterized by clinical manifestations, such as fever, dry cough, headache, and in more severe cases, respiratory distress. Moreover, neurological-, cardiac-, and renal-related symptoms have also been described. Clinical evidence suggests that migration of immune cells to the affected organs can produce an exacerbated release of proinflammatory mediators that contribute to disease and render the immune response as a major player during the development of the COVID-19 disease. Due to the current sanitary situation, the development of vaccines is imperative. Up to the date, 42 prototypes are being tested in humans in different clinical stages, with 10 vaccine candidates undergoing evaluation in phase III clinical trials. In the same way, the search for an effective treatment to approach the most severe cases is also in constant advancement. Several potential therapies have been tested since COVID-19 was described, including antivirals, antiparasitic and immune modulators. Recently, clinical trials with hydroxychloroquine-a promising drug in the beginning-were suspended. In addition, the Food and Drug Administration (FDA) approved convalescent serum administration as a treatment for SARS-CoV-2 patients. Moreover, monoclonal antibody therapy is also under development to neutralize the virus and prevent infection. In this article, we describe the clinical manifestations and the immunological information available about COVID-19 disease. Furthermore, we discuss current therapies under study and the development of vaccines to prevent this disease.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , COVID-19 Vaccines/immunology , COVID-19/pathology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Monoclonal/therapeutic use , COVID-19/prevention & control , COVID-19/therapy , Female , Humans , Immunization, Passive/methods , Male , RNA, Viral/genetics , Receptors, Virus/metabolism , T-Lymphocytes/immunology , COVID-19 Serotherapy
20.
Front Cell Infect Microbiol ; 10: 589505, 2020.
Article in English | MEDLINE | ID: mdl-33364201

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemics is a challenge without precedent for the modern science. Acute Respiratory Discomfort Syndrome (ARDS) is the most common immunopathological event in SARS-CoV-2, SARS-CoV, and MERS-CoV infections. Fast lung deterioration results of cytokine storm determined by a robust immunological response leading to ARDS and multiple organ failure. Here, we show cysteine protease Cathepsin L (CatL) involvement with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and COVID-19 from different points of view. CatL is a lysosomal enzyme that participates in numerous physiological processes, including apoptosis, antigen processing, and extracellular matrix remodeling. CatL is implicated in pathological conditions like invasion and metastasis of tumors, inflammatory status, atherosclerosis, renal disease, diabetes, bone diseases, viral infection, and other diseases. CatL expression is up-regulated during chronic inflammation and is involved in degrading extracellular matrix, an important process for SARS-CoV-2 to enter host cells. In addition, CatL is probably involved in processing SARS-CoV-2 spike protein. As its inhibition is detrimental to SARS-CoV-2 infection and possibly exit from cells during late stages of infection, CatL could have been considered a valuable therapeutic target. Therefore, we describe here some drugs already in the market with potential CatL inhibiting capacity that could be used to treat COVID-19 patients. In addition, we discuss the possible role of host genetics in the etiology and spreading of the disease.


Subject(s)
COVID-19/complications , Cathepsin L/physiology , Pandemics , Respiratory Distress Syndrome/enzymology , SARS-CoV-2/physiology , Acute Kidney Injury/etiology , Amantadine/therapeutic use , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/epidemiology , Cathepsin L/antagonists & inhibitors , Cathepsin L/genetics , Chloroquine/therapeutic use , Cysteine Proteinase Inhibitors/therapeutic use , Genetic Predisposition to Disease , Heparin/therapeutic use , Humans , Hydroxychloroquine/therapeutic use , Lysosomes/enzymology , Molecular Targeted Therapy , Receptors, Virus/metabolism , Respiratory Distress Syndrome/etiology , SARS-CoV-2/ultrastructure , Serine Endopeptidases/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Teicoplanin/therapeutic use , Virus Internalization , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL