Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.522
Filtrar
1.
Biochem Soc Trans ; 51(3): 1143-1155, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37306404

RESUMO

Coxsackievirus and adenovirus receptor (CAR) is a transmembrane cell-cell adhesion receptor that forms homodimers across junctions and plays a key role in mediating epithelial barrier integrity. CAR can also heterodimerise with receptors on the surface of leukocytes and thus plays an additional role in mediating immune cell transmigration across epithelial tissues. Given the importance of both biological processes in cancer, CAR is emerging as a potential mediator of tumorigenesis as well as a target on cancer cells for viral therapy delivery. However, the emerging, often conflicting, evidence suggests that CAR function is tightly regulated and that contributions to disease progression are likely to be context specific. Here, we summarise reported roles for CAR in the context of cancer and draw on observations in other disease settings to offer a perspective on the potential relevance of this receptor as a therapeutic target for solid tumours.


Assuntos
Carcinogênese , Receptores Virais , Humanos , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Receptores Virais/fisiologia , Adesão Celular/fisiologia , Transformação Celular Neoplásica
2.
Mol Biol Rep ; 49(3): 2321-2324, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35102475

RESUMO

Numerous studies demonstrate parallels between CVD, type 2 diabetes mellitus (T2DM) and COVID-19 pathology, which accentuate pre-existing complications in patients infected with COVID-19 and potentially exacerbate the infection course. Antidiabetic drugs such as sodium-glucose transporter-2 (SGLT-2) inhibitors have garnered substantial attention recently due to their efficacy in reducing the severity of cardiorenal disease. The effect of SGLT-2 inhibitors in patients with COVID-19 remains unclear particularly since SGLT-2 inhibitors contribute to altering the RAAS cascade activity, which includes ACE-2, the major cell entry receptor for SARS-CoV2. A study, DARE-19, was carried out to unveil the effects of SGLT-2 inhibitor treatment on comorbid disease complications and concomitant COVID-19 outcomes and demonstrated no statistical significance. However, the need for further studies is essential to provide conclusive clinical findings.


Assuntos
Compostos Benzidrílicos/uso terapêutico , COVID-19/complicações , Glucosídeos/uso terapêutico , Sistema Renina-Angiotensina/efeitos dos fármacos , Insuficiência Respiratória/tratamento farmacológico , SARS-CoV-2 , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Enzima de Conversão de Angiotensina 2/fisiologia , Ensaios Clínicos Fase III como Assunto , Método Duplo-Cego , Reposicionamento de Medicamentos , Cardiopatias/prevenção & controle , Humanos , Nefropatias/prevenção & controle , Mitocôndrias/efeitos dos fármacos , Estudos Multicêntricos como Assunto , Estresse Oxidativo/efeitos dos fármacos , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores Virais/fisiologia , Insuficiência Respiratória/etiologia , Transportador 2 de Glucose-Sódio/fisiologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
3.
J Mol Cell Cardiol ; 164: 69-82, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34838588

RESUMO

The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.


Assuntos
COVID-19/patologia , Endotélio Vascular/patologia , SARS-CoV-2 , Adolescente , Adulto , Idoso , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , COVID-19/sangue , COVID-19/complicações , COVID-19/fisiopatologia , COVID-19/terapia , Ensaios Clínicos como Assunto , Células Endoteliais/patologia , Células Endoteliais/virologia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Proteína HMGB1/fisiologia , Humanos , Macaca mulatta , Camundongos , Neuropilina-1/fisiologia , Estresse Oxidativo , Espécies Reativas de Oxigênio , Receptores Virais/fisiologia , Receptores Depuradores Classe B/fisiologia , Índice de Gravidade de Doença , Transdução de Sinais , Síndrome de Resposta Inflamatória Sistêmica/patologia , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Trombofilia/etiologia , Trombofilia/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Vasculite/etiologia , Vasculite/imunologia , Vasculite/fisiopatologia , Adulto Jovem
5.
Adv Sci (Weinh) ; 9(2): e2103240, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34761549

RESUMO

The outbreak of 2019 coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in a global pandemic. Despite intensive research, the current treatment options show limited curative efficacies. Here the authors report a strategy incorporating neutralizing antibodies conjugated to the surface of a photothermal nanoparticle (NP) to capture and inactivate SARS-CoV-2. The NP is comprised of a semiconducting polymer core and a biocompatible polyethylene glycol surface decorated with high-affinity neutralizing antibodies. The multifunctional NP efficiently captures SARS-CoV-2 pseudovirions and completely blocks viral infection to host cells in vitro through the surface neutralizing antibodies. In addition to virus capture and blocking function, the NP also possesses photothermal function to generate heat following irradiation for inactivation of virus. Importantly, the NPs described herein significantly outperform neutralizing antibodies at treating authentic SARS-CoV-2 infection in vivo. This multifunctional NP provides a flexible platform that can be readily adapted to other SARS-CoV-2 antibodies and extended to novel therapeutic proteins, thus it is expected to provide a broad range of protection against original SARS-CoV-2 and its variants.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , COVID-19/terapia , Imunoconjugados/administração & dosagem , Nanopartículas , SARS-CoV-2/imunologia , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/imunologia , Reações Antígeno-Anticorpo , COVID-19/imunologia , COVID-19/virologia , Avaliação Pré-Clínica de Medicamentos , Temperatura Alta , Humanos , Imunoconjugados/imunologia , Imunoconjugados/uso terapêutico , Luz , Camundongos , Nanopartículas/uso terapêutico , Fosfatidiletanolaminas , Polietilenoglicóis , Polímeros , Receptores Virais/fisiologia , Semicondutores , Glicoproteína da Espícula de Coronavírus/imunologia , Tiadiazóis , Inativação de Vírus
6.
J Cell Biochem ; 123(2): 347-358, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34741481

RESUMO

As per the World Health Organization report, around 226 844 344 confirmed positive cases and 4 666 334 deaths are reported till September 17, 2021 due to the recent viral outbreak. A novel coronavirus (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]) is responsible for the associated coronavirus disease (COVID-19), which causes serious or even fatal respiratory tract infection and yet no approved therapeutics or effective treatment is currently available to combat the outbreak. Due to the emergency, the drug repurposing approach is being explored for COVID-19. In this study, we attempt to understand the potential mechanism and also the effect of the approved antiviral drugs against the SARS-CoV-2 main protease (Mpro). To understand the mechanism of inhibition of the malaria drug hydroxychloroquine (HCQ) against SARS-CoV-2, we performed molecular interaction studies. The studies revealed that HCQ docked at the active site of the Human ACE2 receptor as a possible way of inhibition. Our in silico analysis revealed that the three drugs Lopinavir, Ritonavir, and Remdesivir showed interaction with the active site residues of Mpro. During molecular dynamics simulation, based on the binding free energy contributions, Lopinavir showed better results than Ritonavir and Remdesivir.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Proteases 3C de Coronavírus/antagonistas & inibidores , Hidroxicloroquina/farmacologia , Lopinavir/farmacologia , Receptores Virais/efeitos dos fármacos , Ritonavir/farmacologia , SARS-CoV-2/efeitos dos fármacos , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Alanina/farmacologia , Alanina/uso terapêutico , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/fisiologia , Antivirais/uso terapêutico , Sítios de Ligação , Domínio Catalítico/efeitos dos fármacos , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/fisiologia , Conjuntos de Dados como Assunto , Reposicionamento de Medicamentos , Transferência de Energia , Humanos , Hidroxicloroquina/uso terapêutico , Lopinavir/uso terapêutico , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Receptores Virais/fisiologia , Ritonavir/uso terapêutico
7.
Viruses ; 13(12)2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34960774

RESUMO

The chicken Tva cell surface protein, a member of the low-density lipoprotein receptor family, has been identified as an entry receptor for avian leukosis virus of classic subgroup A and newly emerging subgroup K. Because both viruses represent an important concern for the poultry industry, we introduced a frame-shifting deletion into the chicken tva locus with the aim of knocking-out Tva expression and creating a virus-resistant chicken line. The tva knock-out was prepared by CRISPR/Cas9 gene editing in chicken primordial germ cells and orthotopic transplantation of edited cells into the testes of sterilized recipient roosters. The resulting tva -/- chickens tested fully resistant to avian leukosis virus subgroups A and K, both in in vitro and in vivo assays, in contrast to their susceptible tva +/+ and tva +/- siblings. We also found a specific disorder of the cobalamin/vitamin B12 metabolism in the tva knock-out chickens, which is in accordance with the recently recognized physiological function of Tva as a receptor for cobalamin in complex with transcobalamin transporter. Last but not least, we bring a new example of the de novo resistance created by CRISPR/Cas9 editing of pathogen dependence genes in farm animals and, furthermore, a new example of gene editing in chicken.


Assuntos
Vírus da Leucose Aviária/fisiologia , Proteínas Aviárias/fisiologia , Galinhas/virologia , Receptores Virais/fisiologia , Vitamina B 12/metabolismo , Animais , Vírus da Leucose Aviária/classificação , Proteínas Aviárias/genética , Embrião de Galinha , Feminino , Mutação da Fase de Leitura , Edição de Genes , Técnicas de Inativação de Genes , Masculino , Ácido Metilmalônico/sangue , Receptores Virais/genética
8.
PLoS Comput Biol ; 17(11): e1009560, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34793437

RESUMO

Severe acute respiratory coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, is of zoonotic origin. Evolutionary analyses assessing whether coronaviruses similar to SARS-CoV-2 infected ancestral species of modern-day animal hosts could be useful in identifying additional reservoirs of potentially dangerous coronaviruses. We reasoned that if a clade of species has been repeatedly exposed to a virus, then their proteins relevant for viral entry may exhibit adaptations that affect host susceptibility or response. We perform comparative analyses across the mammalian phylogeny of angiotensin-converting enzyme 2 (ACE2), the cellular receptor for SARS-CoV-2, in order to uncover evidence for selection acting at its binding interface with the SARS-CoV-2 spike protein. We uncover that in rodents there is evidence for adaptive amino acid substitutions at positions comprising the ACE2-spike interaction interface, whereas the variation within ACE2 proteins in primates and some other mammalian clades is not consistent with evolutionary adaptations. We also analyze aminopeptidase N (APN), the receptor for the human coronavirus 229E, a virus that causes the common cold, and find evidence for adaptation in primates. Altogether, our results suggest that the rodent and primate lineages may have had ancient exposures to viruses similar to SARS-CoV-2 and HCoV-229E, respectively.


Assuntos
COVID-19/genética , COVID-19/virologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/virologia , SARS-CoV-2/genética , Adaptação Fisiológica/genética , Substituição de Aminoácidos , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Antígenos CD13/genética , Antígenos CD13/fisiologia , Resfriado Comum/genética , Resfriado Comum/virologia , Biologia Computacional , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/fisiologia , Evolução Molecular , Genômica , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Especificidade de Hospedeiro/genética , Especificidade de Hospedeiro/fisiologia , Humanos , Mamíferos/genética , Mamíferos/virologia , Filogenia , Domínios e Motivos de Interação entre Proteínas/genética , Receptores Virais/genética , Receptores Virais/fisiologia , SARS-CoV-2/fisiologia , Seleção Genética , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/fisiologia , Internalização do Vírus
9.
J Clin Invest ; 131(14)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34263736

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic is among the most important public health crises of our generation. Despite the promise of prevention offered by effective vaccines, patients with severe COVID-19 will continue to populate hospitals and intensive care units for the foreseeable future. The most common clinical presentation of severe COVID-19 is hypoxemia and respiratory failure, typical of the acute respiratory distress syndrome (ARDS). Whether the clinical features and pathobiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia differ from those of pneumonia secondary to other pathogens is unclear. This uncertainty has created variability in the application of historically proven therapies for ARDS to patients with COVID-19. We review the available literature and find many similarities between patients with ARDS from pneumonia attributable to SARS-CoV-2 versus other respiratory pathogens. A notable exception is the long duration of illness among patients with COVID-19, which could result from its unique pathobiology. Available data support the use of care pathways and therapies proven effective for patients with ARDS, while pointing to unique features that might be therapeutically targeted for patients with severe SARS-CoV-2 pneumonia.


Assuntos
COVID-19/etiologia , Pneumonia Viral/etiologia , Síndrome do Desconforto Respiratório/etiologia , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/fisiologia , Autopsia , COVID-19/epidemiologia , COVID-19/patologia , Citocinas/biossíntese , Humanos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , Modelos Biológicos , Pandemias , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Receptores Virais/fisiologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Índice de Gravidade de Doença
10.
Cells ; 10(6)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201214

RESUMO

In late 2019, the betacoronavirus SARS-CoV-2 was identified as the viral agent responsible for the coronavirus disease 2019 (COVID-19) pandemic. Coronaviruses Spike proteins are responsible for their ability to interact with host membrane receptors and different proteins have been identified as SARS-CoV-2 interactors, among which Angiotensin-converting enzyme 2 (ACE2), and Basigin2/EMMPRIN/CD147 (CD147). CD147 plays an important role in human immunodeficiency virus type 1, hepatitis C virus, hepatitis B virus, Kaposi's sarcoma-associated herpesvirus, and severe acute respiratory syndrome coronavirus infections. In particular, SARS-CoV recognizes the CD147 receptor expressed on the surface of host cells by its nucleocapsid protein binding to cyclophilin A (CyPA), a ligand for CD147. However, the involvement of CD147 in SARS-CoV-2 infection is still debated. Interference with both the function (blocking antibody) and the expression (knock down) of CD147 showed that this receptor partakes in SARS-CoV-2 infection and provided additional clues on the underlying mechanism: CD147 binding to CyPA does not play a role; CD147 regulates ACE2 levels and both receptors are affected by virus infection. Altogether, these findings suggest that CD147 is involved in SARS-CoV-2 tropism and represents a possible therapeutic target to challenge COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/fisiologia , Basigina/fisiologia , SARS-CoV-2/fisiologia , Internalização do Vírus , Células A549 , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Basigina/antagonistas & inibidores , Basigina/genética , COVID-19/patologia , COVID-19/prevenção & controle , COVID-19/virologia , Células CACO-2 , Linhagem Celular , Chlorocebus aethiops , Células Hep G2 , Interações Hospedeiro-Patógeno , Humanos , Terapia de Alvo Molecular , Interferência de RNA/fisiologia , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Receptores Virais/metabolismo , Receptores Virais/fisiologia , SARS-CoV-2/metabolismo , Células Vero , Tropismo Viral/fisiologia
11.
Blood ; 138(4): 344-349, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34075401

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with the hypercoagulable state. Tissue factor (TF) is the primary cellular initiator of coagulation. Most of the TF expressed on cell surfaces remains cryptic. Sphingomyelin (SM) is responsible for maintaining TF in the encrypted state, and hydrolysis of SM by acid sphingomyelinase (ASMase) increases TF activity. ASMase was shown to play a role in virus infection biology. In the present study, we investigated the role of ASMase in SARS-CoV-2 infection-induced TF procoagulant activity. Infection of human monocyte-derived macrophages (MDMs) with SARS-CoV-2 spike protein pseudovirus (SARS-CoV-2-SP-PV) markedly increased TF procoagulant activity at the cell surface and released TF+ extracellular vesicles. The pseudovirus infection did not increase either TF protein expression or phosphatidylserine externalization. SARS-CoV-2-SP-PV infection induced the translocation of ASMase to the outer leaflet of the plasma membrane, which led to the hydrolysis of SM in the membrane. Pharmacologic inhibitors or genetic silencing of ASMase attenuated SARS-CoV-2-SP-PV-induced increased TF activity. Inhibition of the SARS-CoV-2 receptor, angiotensin-converting enzyme-2, attenuated SARS-CoV-2-SP-PV-induced increased TF activity. Overall, our data suggest that SARS-CoV-2 infection activates the coagulation by decrypting TF through activation of ASMase. Our data suggest that the US Food and Drug Administration-approved functional inhibitors of ASMase may help treat hypercoagulability in patients with COVID-19.


Assuntos
COVID-19/sangue , Macrófagos/virologia , Proteínas de Membrana/fisiologia , SARS-CoV-2 , Esfingomielina Fosfodiesterase/fisiologia , Glicoproteína da Espícula de Coronavírus/fisiologia , Trombofilia/etiologia , Tromboplastina/fisiologia , Enzima de Conversão de Angiotensina 2/fisiologia , COVID-19/complicações , Micropartículas Derivadas de Células , Ativação Enzimática , Humanos , Hidrólise , Macrófagos/enzimologia , Terapia de Alvo Molecular , Plasmídeos , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores Virais/fisiologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielinas/fisiologia , Trombofilia/sangue , Trombofilia/tratamento farmacológico , Trombofilia/enzimologia
12.
Mol Neurobiol ; 58(9): 4535-4563, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34089508

RESUMO

Coronavirus disease 2019 (COVID-19) is a devastating viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The incidence and mortality of COVID-19 patients have been increasing at an alarming rate. The mortality is much higher in older individuals, especially the ones suffering from respiratory distress, cardiac abnormalities, renal diseases, diabetes, and hypertension. Existing evidence demonstrated that SARS-CoV-2 makes its entry into human cells through angiotensin-converting enzyme 2 (ACE-2) followed by the uptake of virions through cathepsin L or transmembrane protease serine 2 (TMPRSS2). SARS-CoV-2-mediated abnormalities in particular cardiovascular and neurological ones and the damaged coagulation systems require extensive research to develop better therapeutic modalities. As SARS-CoV-2 uses its S-protein to enter into the host cells of several organs, the S-protein of the virus is considered as the ideal target to develop a potential vaccine. In this review, we have attempted to highlight the landmark discoveries that lead to the development of various vaccines that are currently under different stages of clinical progression. Besides, a brief account of various drug candidates that are being tested to mitigate the burden of COVID-19 was also covered. Further, in a dedicated section, the impact of SARS-CoV-2 infection on neuronal inflammation and neuronal disorders was discussed. In summary, it is expected that the content covered in this article help to understand the pathophysiology of COVID-19 and the impact on neuronal complications induced by SARS-CoV-2 infection while providing an update on the vaccine development.


Assuntos
Vacinas contra COVID-19 , COVID-19/complicações , Inflamação/etiologia , Transtornos do Neurodesenvolvimento/etiologia , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Antivirais/uso terapêutico , COVID-19/fisiopatologia , COVID-19/prevenção & controle , COVID-19/terapia , Vacinas contra COVID-19/efeitos adversos , Linhagem Celular , Comorbidade , Síndrome da Liberação de Citocina/etiologia , Feminino , Hormese , Humanos , Imunização Passiva , Transmissão Vertical de Doenças Infecciosas , Camundongos , Modelos Neurológicos , Vírus da Hepatite Murina/patogenicidade , Sistema Nervoso/virologia , Doenças do Sistema Nervoso/epidemiologia , Doenças do Sistema Nervoso/etiologia , Especificidade de Órgãos , Organoides , Gravidez , Complicações Infecciosas na Gravidez/virologia , Receptores Virais/fisiologia , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Serina Endopeptidases/fisiologia , Glicoproteína da Espícula de Coronavírus/fisiologia , Soroterapia para COVID-19 , Tratamento Farmacológico da COVID-19
13.
IUBMB Life ; 73(6): 843-854, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33960608

RESUMO

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.


Assuntos
Doenças Autoimunes do Sistema Nervoso/imunologia , COVID-19/transmissão , Proteínas de Choque Térmico/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores Virais/fisiologia , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes do Sistema Nervoso/metabolismo , Sobrevivência Celular , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/fisiologia , Exossomos , Proteínas Ligadas por GPI/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/imunologia , Humanos , Ligantes , Invasividade Neoplásica , Proteínas de Neoplasias/imunologia , Proteínas do Tecido Nervoso/imunologia , Domínios Proteicos , Transporte Proteico , Transdução de Sinais , Microambiente Tumoral , Resposta a Proteínas não Dobradas/fisiologia , Internalização do Vírus
14.
J Hematol Oncol ; 14(1): 72, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33926500

RESUMO

Evidence suggests that platelets may directly interact with SARS-CoV-2, raising the concern whether ACE2 receptor plays a role in this interaction. The current study showed that SARS-CoV-2 interacts with both platelets and megakaryocytes despite the limited efficiency. Abundance of the conventional receptor ACE2 and alternative receptors or co-factors for SARS-CoV-2 entry was characterized in platelets from COVID-19 patients and healthy persons as well as human megakaryocytes based on laboratory tests or previously reported RNA-seq data. The results suggest that SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism and may regulate alternative receptor expression associated with COVID-19 coagulation dysfunction.


Assuntos
Plaquetas/virologia , COVID-19/sangue , Megacariócitos/virologia , Receptores Virais/fisiologia , SARS-CoV-2/fisiologia , Ligação Viral , Enzima de Conversão de Angiotensina 2/análise , Plaquetas/metabolismo , COVID-19/complicações , COVID-19/genética , Linhagem Celular , Humanos , Megacariócitos/metabolismo , Especificidade de Órgãos , Ativação Plaquetária , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Viral/isolamento & purificação , SARS-CoV-2/isolamento & purificação , Trombofilia/etiologia , Trombofilia/virologia , Transcrição Gênica , Internalização do Vírus
16.
J Virol ; 95(9)2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33568504

RESUMO

Pestiviruses such as bovine viral diarrhea virus (BVDV) and classical swine fever virus (CSFV) belong to the family Flaviviridae and represent pathogens of outstanding veterinary relevance. Pestiviruses enter cells via receptor-mediated endocytosis. For entry in bovine cells, complement regulatory protein CD46bov serves as a cellular receptor for BVDV. In this study, the role of porcine CD46pig in cellular entry was investigated for the recently discovered atypical porcine pestivirus (APPV), CSFV, and Bungowannah virus (BuPV) in order to elucidate the observed differences in host cell tropism. A cell culture-adapted APPV variant, which shows enhanced viral replication in vitro, was generated and demonstrated a strict tropism of APPV for porcine cells. One of the porcine cell lines displayed areas of CD46pig-expressing cells and areas of nonexpressing cells, and one single cell line revealed not to express any CD46pig The CD46pig-deficient porcine lymphoma cell line, known to facilitate CSFV replication, was the only porcine cell line nonpermissive to APPV, indicating a significant difference in the entry mechanism of APPV and CSFV. Infection experiments with a set of genetically engineered CD46pig knockout cells confirmed that CD46pig is a major receptor of APPV as CD46bov is for BVDV. In contrast, it is apparently not an essential determinant in host cell entry of other porcine pestiviruses such as CSFV and BuPV. Existence of a CD46pig-independent entry mechanism illustrates that the pestiviral entry process is more diverse than previously recognized.IMPORTANCE Pestiviruses comprise animal pathogens such as classical swine fever virus (CSFV) and bovine viral diarrhea virus (BVDV) that cause notifiable diseases with great economic impact. Several additional pestivirus species affecting animal health were recently identified, including atypical porcine pestivirus (APPV). APPV is associated with health problems in piglets and is highly abundant in pig populations worldwide. Complement control protein CD46 serves as a receptor for diverse bacterial and viral pathogens, including particular adenoviruses, herpesviruses, measles virus (MeV), and BVDV. Porcine CD46 (CD46pig) was suggested to be a major receptor for CSFV. Here, we identified remarkable differences in relevance of CD46pig during entry of porcine pestiviruses. Resembling BVDV, efficient APPV infection in cell culture depends on CD46pig, while other porcine pestiviruses can efficiently enter and infect cells in the absence of CD46pig Thus, the study provides insights into the entry process of these pathogens and may help to understand differences in their biology.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Peste Suína Clássica/virologia , Proteína Cofatora de Membrana/fisiologia , Receptores Virais/fisiologia , Tropismo Viral , Internalização do Vírus , Animais , Linhagem Celular , Proteína Cofatora de Membrana/imunologia , Suínos
17.
J Infect Dev Ctries ; 15(1): 1-8, 2021 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-33571140

RESUMO

An innate immune response is essential to mobilize protective immunity upon the infection of respiratory epithelial cells with influenza A virus (IAV). The response is classified as early (nonspecific effectors), local systematic (effector cells recruitment) and late (antigen to lymphoid organ transport, naive B and T cells recognition, effector cells clonal expansion and differentiation). Virus particles are detected by the host cells as non-self by various sensors that are present on the cell surface, endosomes and cytosol. These sensors are collectively termed as pattern recognition receptors (PRRs). The PRRs distinguish unique molecular signatures known as pathogen-associated molecular pattern, which are present either on the cell surface or within intracellular compartments. PRRs have been classified into five major groups: C-Type Lectin Receptor (CLR), Toll-like receptor (TLR), Nod-like receptor (NLR), Retinoic acid-inducible gene-I-like receptor (RLR), which play a role in innate immunity to IAV infection, and the pyrin and hematopoietic interferon-inducible nuclear (PYHIN) domain protein. Here, we discuss the role of PRRs in cellular infectivity of IAV and highlight the recent progress.


Assuntos
Vírus da Influenza A/fisiologia , Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Influenza Humana/virologia , Receptores de Reconhecimento de Padrão/fisiologia , Transdução de Sinais , Animais , Proteína DEAD-box 58/fisiologia , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Lectinas Tipo C/fisiologia , Proteína Adaptadora de Sinalização NOD1/fisiologia , Proteínas Nucleares/fisiologia , Receptores Imunológicos/fisiologia , Receptores Virais/fisiologia , Receptores Toll-Like/fisiologia
18.
J Mol Neurosci ; 71(11): 2192-2209, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33464535

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic is an issue of global significance that has taken the lives of many across the world. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus responsible for its pathogenesis. The pulmonary manifestations of COVID-19 have been well described in the literature. Initially, it was thought to be limited to the respiratory system; however, we now recognize that COVID-19 also affects several other organs, including the nervous system. Two similar human coronaviruses (CoV) that cause severe acute respiratory syndrome (SARS-CoV-1) and Middle East respiratory syndrome (MERS-CoV) are also known to cause disease in the nervous system. The neurological manifestations of SARS-CoV-2 infection are growing rapidly, as evidenced by several reports. There are several mechanisms responsible for such manifestations in the nervous system. For instance, post-infectious immune-mediated processes, direct virus infection of the central nervous system (CNS), and virus-induced hyperinflammatory and hypercoagulable states are commonly involved. Guillain-Barré syndrome (GBS) and its variants, dysfunction of taste and smell, and muscle injury are numerous examples of COVID-19 PNS (peripheral nervous system) disease. Likewise, hemorrhagic and ischemic stroke, encephalitis, meningitis, encephalopathy acute disseminated encephalomyelitis, endothelialitis, and venous sinus thrombosis are some instances of COVID-19 CNS disease. Due to multifactorial and complicated pathogenic mechanisms, COVID-19 poses a large-scale threat to the whole nervous system. A complete understanding of SARS-CoV-2 neurological impairments is still lacking, but our knowledge base is rapidly expanding. Therefore, we anticipate that this comprehensive review will provide valuable insights and facilitate the work of neuroscientists in unfolding different neurological dimensions of COVID-19 and other CoV associated abnormalities.


Assuntos
COVID-19/complicações , Doenças do Sistema Nervoso/etiologia , Pandemias , SARS-CoV-2/patogenicidade , Adolescente , Adulto , Enzima de Conversão de Angiotensina 2/fisiologia , Infecções Assintomáticas , Doenças Autoimunes do Sistema Nervoso/etiologia , Barreira Hematoencefálica , COVID-19/imunologia , COVID-19/fisiopatologia , Transtornos Cerebrovasculares/etiologia , Criança , Doenças Transmissíveis Emergentes , Infecções por Coronavirus/complicações , Humanos , Hipóxia/etiologia , Hipóxia/fisiopatologia , Sistema Nervoso/virologia , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/fisiopatologia , Especificidade de Órgãos , Receptores Virais/fisiologia , Síndrome Respiratória Aguda Grave/complicações , Sinapses/virologia , Tropismo Viral , Adulto Jovem
19.
Sci Signal ; 14(665)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436498

RESUMO

The spike protein of SARS-CoV-2 binds the angiotensin-converting enzyme 2 (ACE2) on the host cell surface and subsequently enters host cells through receptor-mediated endocytosis. Additional cell receptors may be directly or indirectly involved, including integrins. The cytoplasmic tails of ACE2 and integrins contain several predicted short linear motifs (SLiMs) that may facilitate internalization of the virus as well as its subsequent propagation through processes such as autophagy. Here, we measured the binding affinity of predicted interactions between SLiMs in the cytoplasmic tails of ACE2 and integrin ß3 with proteins that mediate endocytic trafficking and autophagy. We validated that a class I PDZ-binding motif mediated binding of ACE2 to the scaffolding proteins SNX27, NHERF3, and SHANK, and that a binding site for the clathrin adaptor AP2 µ2 in ACE2 overlaps with a phospho-dependent binding site for the SH2 domains of Src family tyrosine kinases. Furthermore, we validated that an LC3-interacting region (LIR) in integrin ß3 bound to the ATG8 domains of the autophagy receptors MAP1LC3 and GABARAP in a manner enhanced by LIR-adjacent phosphorylation. Our results provide molecular links between cell receptors and mediators of endocytosis and autophagy that may facilitate viral entry and propagation.


Assuntos
Enzima de Conversão de Angiotensina 2/fisiologia , COVID-19/virologia , Integrina beta3/fisiologia , Receptores Virais/fisiologia , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , Internalização do Vírus , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Autofagia/fisiologia , Endocitose/fisiologia , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Integrina beta3/química , Integrina beta3/genética , Modelos Moleculares , Pandemias , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Receptores Virais/química , Receptores Virais/genética , SARS-CoV-2/genética
20.
Sci Signal ; 14(665)2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436497

RESUMO

The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the µ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin ß3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.


Assuntos
COVID-19/virologia , Interações entre Hospedeiro e Microrganismos/fisiologia , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , Internalização do Vírus , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , COVID-19/terapia , Sequência Conservada , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Integrinas/química , Integrinas/genética , Integrinas/fisiologia , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/fisiologia , Modelos Biológicos , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/fisiologia , Domínios e Motivos de Interação entre Proteínas/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/fisiologia , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...