Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.893
Filtrar
1.
Vet Res ; 55(1): 71, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822398

RESUMO

In the wake of the COVID-19 pandemic caused by SARS-CoV-2, questions emerged about the potential effects of Bacillus Calmette-Guérin (BCG) vaccine on the immune response to SARS-CoV-2 infection, including the neurodegenerative diseases it may contribute to. To explore this, an experimental study was carried out in BCG-stimulated and non-stimulated k18-hACE2 mice challenged with SARS-CoV-2. Viral loads in tissues determined by RT-qPCR, histopathology in brain and lungs, immunohistochemical study in brain (IHC) as well as mortality rates, clinical signs and plasma inflammatory and coagulation biomarkers were assessed. Our results showed BCG-SARS-CoV-2 challenged mice presented higher viral loads in the brain and an increased frequency of neuroinvasion, with the greatest differences observed between groups at 3-4 days post-infection (dpi). Histopathological examination showed a higher severity of brain lesions in BCG-SARS-CoV-2 challenged mice, mainly consisting of neuroinflammation, increased glial cell population and neuronal degeneration, from 5 dpi onwards. This group also presented higher interstitial pneumonia and vascular thrombosis in lungs (3-4 dpi), BCG-SARS-CoV-2 mice showed higher values for TNF-α and D-dimer values, while iNOS values were higher in SARS-CoV-2 mice at 3-4 dpi. Results presented in this study indicate that BCG stimulation could have intensified the inflammatory and neurodegenerative lesions promoting virus neuroinvasion and dissemination in this experimental model. Although k18-hACE2 mice show higher hACE2 expression and neurodissemination, this study suggests that, although the benefits of BCG on enhancing heterologous protection against pathogens and tumour cells have been broadly demonstrated, potential adverse outcomes due to the non-specific effects of BCG should be considered.


Assuntos
Vacina BCG , Encéfalo , COVID-19 , SARS-CoV-2 , Animais , Camundongos , Vacina BCG/administração & dosagem , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/fisiologia , Encéfalo/patologia , Encéfalo/virologia , Carga Viral , Pulmão/patologia , Pulmão/virologia , Pulmão/imunologia , Enzima de Conversão de Angiotensina 2/metabolismo , Camundongos Transgênicos , Feminino
2.
Proc Natl Acad Sci U S A ; 121(24): e2321619121, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38833475

RESUMO

Angiotensin-convertingenzyme 2 (ACE2) has dual functions, regulating cardiovascular physiology and serving as the receptor for coronaviruses. Bats, the only true flying mammals and natural viral reservoirs, have evolved positive alterations in traits related to both functions of ACE2. This suggests significant evolutionary changes in ACE2 during bat evolution. To test this hypothesis, we examine the selection pressure in ACE2 along the ancestral branch of all bats (AncBat-ACE2), where powered flight and bat-coronavirus coevolution occurred, and detect a positive selection signature. To assess the functional effects of positive selection, we resurrect AncBat-ACE2 and its mutant (AncBat-ACE2-mut) created by replacing the positively selected sites. Compared to AncBat-ACE2-mut, AncBat-ACE2 exhibits stronger enzymatic activity, enhances mice's performance in exercise fatigue, and shows lower affinity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our findings indicate the functional pleiotropy of positive selection in the ancient ACE2 of bats, providing an alternative hypothesis for the evolutionary origin of bats' defense against coronaviruses.


Assuntos
Enzima de Conversão de Angiotensina 2 , Quirópteros , Seleção Genética , Quirópteros/virologia , Quirópteros/genética , Animais , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Camundongos , Pleiotropia Genética , Evolução Molecular , SARS-CoV-2/genética , COVID-19/virologia , COVID-19/genética , Coronavirus/genética , Humanos , Filogenia
3.
Clin Exp Hypertens ; 46(1): 2361671, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38841901

RESUMO

BACKGROUND: Fibroblast growth factor 21 (FGF21) has a protective effect against cardiovascular disease. However, the role of FGF21 in hypertension remains elusive. METHODS: Ten-week-old male C57BL/6 mice were randomly divided into normal-salt (NS) group, NS+FGF21 group, deoxycorticosterone acetate-salt (DOCA) group and DOCA+FGF21 group. The mice in NS group underwent uninephrectomy without receiving DOCA and 1% NaCl and the mice in DOCA group were subjected to uninephrectomy and DOCA-salt (DOCA and 1% NaCl) treatment for 6 weeks. At the same time, the mice were infused with vehicle (artificial cerebrospinal fluid, aCSF) or FGF21 (1 mg/kg) into the bilateral paraventricular nucleus (PVN) of mice. RESULTS: Here, we showed that FGF21 treatment lowered DOCA salt-induced inflammation and oxidative stress in the PVN, which reduced sympathetic nerve activity and hypertension. Mechanistically, FGF21 treatment decreased the expression of HNF4α and inhibited the binding activity of HNF4α to the promoter region of ACE2 in the PVN of DOCA salt-treated mice, which further up-regulated ACE2/Ang (1-7) signals in the PVN. In addition, ACE2 deficiency abolished the protective effect of FGF21 in DOCA salt-treated mice, suggesting that FGF21-mediated antihypertensive effect was dependent on ACE2. CONCLUSIONS: The results demonstrate that FGF21 protects against salt-sensitive hypertension via regulating HNF4α/ACE2/Ang (1-7) axis in the PVN of DOCA salt-treated mice via multi-organ crosstalk between liver, brain and blood vessels.


Assuntos
Enzima de Conversão de Angiotensina 2 , Acetato de Desoxicorticosterona , Fatores de Crescimento de Fibroblastos , Fator 4 Nuclear de Hepatócito , Hipertensão , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular , Animais , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/metabolismo , Masculino , Camundongos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Enzima de Conversão de Angiotensina 2/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Estresse Oxidativo/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cloreto de Sódio na Dieta
4.
Sci Adv ; 10(23): eadj4735, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38838150

RESUMO

Why individuals with Down syndrome (DS) are more susceptible to SARS-CoV-2-induced neuropathology remains elusive. Choroid plexus (ChP) plays critical roles in barrier function and immune response modulation and expresses the ACE2 receptor and the chromosome 21-encoded TMPRSS2 protease, suggesting its substantial role in establishing SARS-CoV-2 infection in the brain. To explore this, we established brain organoids from DS and isogenic euploid iPSC that consist of a core of functional cortical neurons surrounded by a functional ChP-like epithelium (ChPCOs). DS-ChPCOs recapitulated abnormal DS cortical development and revealed defects in ciliogenesis and epithelial cell polarity in ChP-like epithelium. We then demonstrated that the ChP-like epithelium facilitates infection and replication of SARS-CoV-2 in cortical neurons and that this is increased in DS. Inhibiting TMPRSS2 and furin activity reduced viral replication in DS-ChPCOs to euploid levels. This model enables dissection of the role of ChP in neurotropic virus infection and euploid forebrain development and permits screening of therapeutics for SARS-CoV-2-induced neuropathogenesis.


Assuntos
Encéfalo , COVID-19 , Plexo Corióideo , Síndrome de Down , Organoides , SARS-CoV-2 , Serina Endopeptidases , Plexo Corióideo/virologia , Plexo Corióideo/metabolismo , Plexo Corióideo/patologia , Organoides/virologia , Organoides/metabolismo , Organoides/patologia , Humanos , SARS-CoV-2/fisiologia , COVID-19/virologia , COVID-19/patologia , COVID-19/metabolismo , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Síndrome de Down/metabolismo , Síndrome de Down/patologia , Síndrome de Down/genética , Encéfalo/virologia , Encéfalo/patologia , Encéfalo/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Neurônios/patologia , Replicação Viral , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/virologia , Furina/metabolismo , Furina/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Tropismo Viral
5.
Int J Nanomedicine ; 19: 4957-4976, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38828198

RESUMO

Background: The "gut-islets axis" is an important endocrine signaling axis that regulates islets function by modulating the gut microbiota and endocrine metabolism within the gut. However, the specific mechanisms and roles of the intestine in islets regulation remain unclear. Recent studies investigated that exosomes derived from gut microbiota can transport signals to remotely regulate islets ß-cell function, suggesting the possibility of novel signaling pathways mediated by gut exosomes in the regulation of the "gut-islet axis.". Methods: The exosomes were isolated from the intestinal enteroendocrine cell-line STC-1cells culture supernatants treated with palmitate acid (PA) or BSA. Metabolic stress models were established by separately subjecting MIN6 cells to PA stimulation and feeding mice with a high-fat diet. Intervention with exosomes in vitro and in vivo to assess the biological effects of exosomes on islets ß cells under metabolic stress. The Mas receptor antagonist A779 and ACE2ko mice were used to evaluate the role of exosomal ACE2. Results: We found ACE2, a molecule that plays a crucial role in the regulation of islets function, is abundantly expressed in exosomes derived from STC-1 under physiological normal condition (NCEO). These exosomes cannot only be taken up by ß-cells in vitro but also selectively transported to the islets in vivo. Following intervention with NCEXO, both Min6 cells in a lipotoxic environment and mice on a high-fat diet exhibited significant improvements in islets ß-cell function and ß-cell mass. Further investigations demonstrated that these protective effects are attributed to exosomal ACE2, as ACE2 inhibits NLRP3 inflammasome activation and reduces ß-cell pyroptosis. Conclusion: ACE2-enriched exosomes from the gut can selectively target islets, subsequently inhibiting NLRP3 inflammasome activation and ß cell pyroptosis, thereby restoring islets ß cell function under metabolic stress. This study provides novel insights into therapeutic strategies for the prevention and treatment of obesity and diabetes.


Assuntos
Enzima de Conversão de Angiotensina 2 , Exossomos , Inflamassomos , Células Secretoras de Insulina , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Animais , Exossomos/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos , Piroptose/efeitos dos fármacos , Piroptose/fisiologia , Enzima de Conversão de Angiotensina 2/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Linhagem Celular , Intestino Delgado/efeitos dos fármacos , Masculino , Dieta Hiperlipídica , Camundongos Knockout , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/metabolismo
6.
Cells ; 13(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38727309

RESUMO

The activation of endothelial cells is crucial for immune defense mechanisms but also plays a role in the development of atherosclerosis. We have previously shown that inflammatory stimulation of endothelial cells on top of elevated lipoprotein/cholesterol levels accelerates atherogenesis. The aim of the current study was to investigate how chronic endothelial inflammation changes the aortic transcriptome of mice at normal lipoprotein levels and to compare this to the inflammatory response of isolated endothelial cells in vitro. We applied a mouse model expressing constitutive active IκB kinase 2 (caIKK2)-the key activator of the inflammatory NF-κB pathway-specifically in arterial endothelial cells and analyzed transcriptomic changes in whole aortas, followed by pathway and network analyses. We found an upregulation of cell death and mitochondrial beta-oxidation pathways with a predicted increase in endothelial apoptosis and necrosis and a simultaneous reduction in protein synthesis genes. The highest upregulated gene was ACE2, the SARS-CoV-2 receptor, which is also an important regulator of blood pressure. Analysis of isolated human arterial and venous endothelial cells supported these findings and also revealed a reduction in DNA replication, as well as repair mechanisms, in line with the notion that chronic inflammation contributes to endothelial dysfunction.


Assuntos
Colesterol , Células Endoteliais , Inflamação , Animais , Humanos , Células Endoteliais/metabolismo , Camundongos , Inflamação/patologia , Inflamação/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Artérias/metabolismo , Artérias/patologia , Transcriptoma/genética , Aorta/metabolismo , Aorta/patologia , Camundongos Endogâmicos C57BL , Aterosclerose/metabolismo , Aterosclerose/patologia , Quinase I-kappa B/metabolismo , Masculino , NF-kappa B/metabolismo
7.
Biol Pharm Bull ; 47(5): 930-940, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38692871

RESUMO

The coronavirus disease 2019 (COVID-19) is caused by the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19, with the recurrent epidemics of new variants of SARS-CoV-2, remains a global public health problem, and new antivirals are still required. Some cholesterol derivatives, such as 25-hydroxycholesterol, are known to have antiviral activity against a wide range of enveloped and non-enveloped viruses, including SARS-CoV-2. At the entry step of SARS-CoV-2 infection, the viral envelope fuses with the host membrane dependent of viral spike (S) glycoproteins. From the screening of cholesterol derivatives, we found a new compound 26,27-dinorcholest-5-en-24-yne-3ß,20-diol (Nat-20(S)-yne) that inhibited the SARS-CoV-2 S protein-dependent membrane fusion in a syncytium formation assay. Nat-20(S)-yne exhibited the inhibitory activities of SARS-CoV-2 pseudovirus entry and intact SARS-CoV-2 infection in a dose-dependent manner. Among the variants of SARS-CoV-2, inhibition of infection by Nat-20(S)-yne was stronger in delta and Wuhan strains, which predominantly invade into cells via fusion at the plasma membrane, than in omicron strains. The interaction between receptor-binding domain of S proteins and host receptor ACE2 was not affected by Nat-20(S)-yne. Unlike 25-hydroxycholesterol, which regulates various steps of cholesterol metabolism, Nat-20(S)-yne inhibited only de novo cholesterol biosynthesis. As a result, plasma membrane cholesterol content was substantially decreased in Nat-20(S)-yne-treated cells, leading to inhibition of SARS-CoV-2 infection. Nat-20(S)-yne having a new mechanism of action may be a potential therapeutic candidate for COVID-19.


Assuntos
Antivirais , COVID-19 , Colesterol , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacologia , Humanos , COVID-19/virologia , Colesterol/metabolismo , Células Vero , Chlorocebus aethiops , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Internalização do Vírus/efeitos dos fármacos , Betacoronavirus/efeitos dos fármacos , Pandemias , Tratamento Farmacológico da COVID-19 , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/virologia
8.
Am J Reprod Immunol ; 91(5): e13861, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38716765

RESUMO

BACKGROUND: Maternal-fetal immunology is intricate, and the effects of mRNA-S maternal vaccination on immune regulation at the maternal-fetal interface require further investigation. Our study endeavors to elucidate these immunological changes, enhancing our comprehension of maternal and fetal health outcomes. By analyzing immune profiles and cytokine responses, we aim to provide valuable insights into the impact of mRNA-S vaccination on the delicate balance of immune regulation during pregnancy, addressing critical questions in the field of reproductive pharmacology. OBJECTIVES: This investigation sought to examine the prospective influence of mRNA-S-based vaccines and extracellular vesicles (EVs) containing the Spike (S) protein at the maternal-fetal interface. Our primary emphasis was on evaluating their effects on maternal decidua cells and fetal chorion trophoblast cells (hFM-CTCs). METHODS: We validated the generation of EVs containing the S protein from small human airway epithelial cell lines (HSAECs) following mRNA-S vaccine exposure. We assessed the expression of angiotensin-converting enzyme 2 (ACE2) gene and protein in fetal membranes and the placenta, with specific attention to decidual cells and fetal membrane chorion cells. To assess cellular functionality, these cells were exposed to both recombinant S protein and EVs loaded with S proteins (eSPs). RESULTS: Our findings revealed that cells and EVs subjected to mRNA-S-based vaccination exhibited altered protein expression levels of S proteins. At the feto-maternal interface, both placental and fetal membrane tissues demonstrated similar ACE-2 expression levels. Among individual cellular layers, syncytiotrophoblast cells in the placenta and chorion cells in the fetal membrane exhibited elevated ACE-2 expression. Notably, EVs derived from HSAECs activated the MAPK pathway in decidual cells. Additionally, decidual cells displayed a substantial increase in gene expression of chemokines like CXCL-10 and CXCL-11, as well as proinflammatory cytokines such as IL-6 in response to eSPs. However, the levels of Ccl-2 and IL-1ß remained unchanged in decidual cells under the same conditions. Conversely, hFM-CTCs demonstrated significant alterations in the proinflammatory cytokines and chemokines with respect to eSPs. CONCLUSION: In conclusion, our study indicates that mRNA-S-based maternal vaccination during pregnancy may influence the maternal-fetal interface's COVID-19 interaction and immune regulation. Further investigation is warranted to assess safety and implications.


Assuntos
Vesículas Extracelulares , Trofoblastos , Humanos , Feminino , Gravidez , Trofoblastos/imunologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Decídua/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Citocinas/metabolismo , Vacinação , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Troca Materno-Fetal , SARS-CoV-2/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Linhagem Celular , Vacinas contra COVID-19/imunologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética
9.
Front Immunol ; 15: 1383612, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38742107

RESUMO

Introduction: SARS-CoV-2, the cause of the COVID pandemic, is an RNA virus with a high propensity to mutate. Successive virus variants, including variants of concern (VOC), have emerged with increased transmission or immune escape. The original pandemic virus and early variants replicated poorly, if at all, in mice at least partly due to a mismatch between the receptor binding domain on the viral spike protein and the murine angiotensin converting enzyme 2 (ACE2). Omicron VOC emerged in late 2021 harboring > 50 new mutations, 35 of them in the spike protein. This variant resulted in a very large wave of infections, even in the face of prior immunity, albeit being inherently less severe than earlier variants. Reflecting the lower severity reported in humans, Omicron displayed attenuated infection in hamsters and also in the K18-hACE2 mouse model. K18-hACE2 mice express both the human ACE2 as well as the endogenous mouse ACE2. Methods: Here we infected hACE2 knock-in mice that express only human ACE2 and no murine ACE2, or C57BL/6 wildtype mice with SARS-CoV-2 D614G (first-wave isolate), Delta or Omicron BA.1 variants and assessed infectivity and downstream innate immune responses. Results: While replication of SARS-CoV-2 Omicron was lower in the lungs of hACE2 knock-in mice compared with SARS-CoV-2 D614G and VOC Delta, it replicated more efficiently than the earlier variants in C57BL/6 wildtype mice. This opens the opportunity to test the effect of host genetics on SARS-CoV-2 infections in wildtype mice. As a proof of principle, we tested Omicron infection in mice lacking expression of the interferon-alpha receptor-1 (IFNAR1). In these mice we found that loss of type I IFN receptor signaling resulted in higher viral loads in the lungs were detected. Finally, using a chimeric virus of first wave SARS-CoV-2 harboring the Omicron spike protein, we show that Omicron spike increase infection of C57BL/6 wildtype mice, but non-spike genes of Omicron confer attenuation of viral replication. Discussion: Since this chimeric virus efficiently infected C57BL/6 wildtype mice, and replicated in their lungs, our findings illustrate a pathway for genetic mapping of virushost interactions during SARS-CoV-2 infection.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Camundongos Endogâmicos C57BL , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Replicação Viral , Animais , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Camundongos , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/imunologia , COVID-19/virologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Humanos , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Camundongos Transgênicos
10.
J Med Virol ; 96(5): e29671, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38747003

RESUMO

The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.


Assuntos
Barreira Hematoencefálica , COVID-19 , Plexo Corióideo , SARS-CoV-2 , Barreira Hematoencefálica/virologia , Animais , Plexo Corióideo/virologia , Plexo Corióideo/patologia , COVID-19/virologia , COVID-19/patologia , COVID-19/complicações , COVID-19/fisiopatologia , Camundongos , Junções Íntimas/virologia , Modelos Animais de Doenças , Enzima de Conversão de Angiotensina 2/metabolismo , Inflamação/virologia , Humanos , Pericitos/virologia , Pericitos/patologia
11.
FASEB J ; 38(10): e23656, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38752523

RESUMO

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Obesity is a major risk factor for the development of COVID-19. Angiotensin-converting enzyme 2 (ACE2) is an essential receptor for cell entry of SARS-CoV-2. The receptor-binding domain of the S1 subunit (S1-RBD protein) in the SARS-CoV-2 spike glycoprotein binds to ACE2 on host cells, through which the virus enters several organs, including the lungs. Considering these findings, recombinant ACE2 might be utilized as a decoy protein to attenuate SARS-CoV-2 infection. Here, we examined whether obesity increases ACE2 expression in the lungs and whether recombinant ACE2 administration diminishes the entry of S1-RBD protein into lung cells. We observed that high-fat diet-induced obesity promoted ACE2 expression in the lungs by increasing serum levels of LPS derived from the intestine. S1-RBD protein entered the lungs specifically through ACE2 expressed in host lungs and that the administration of recombinant ACE2 attenuated this entry. We conclude that obesity makes hosts susceptible to recombinant SARS-CoV-2 spike proteins due to elevated ACE2 expression in lungs, and this model of administering S1-RBD protein can be applied to new COVID-19 treatments.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Dieta Hiperlipídica , Pulmão , Obesidade , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Dieta Hiperlipídica/efeitos adversos , Camundongos , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Pulmão/metabolismo , Pulmão/virologia , SARS-CoV-2/metabolismo , Obesidade/metabolismo , COVID-19/metabolismo , COVID-19/virologia , Camundongos Endogâmicos C57BL , Internalização do Vírus , Masculino , Humanos , Camundongos Obesos , Proteínas Recombinantes/metabolismo
12.
ACS Appl Bio Mater ; 7(5): 3238-3246, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38700999

RESUMO

As the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues its global spread, the exploration of novel therapeutic and diagnostic strategies is still needed. The virus enters host cells by binding the angiotensin-converting enzyme 2 (ACE2) receptor through the spike protein. Here, we develop an engineered, small, stable, and catalytically inactive version of ACE2, termed miniature ACE2 (mACE2), designed to bind the spike protein with high affinity. Employing a magnetic nanoparticle-based assay, we harnessed the strong binding affinity of mACE2 to develop a sensitive and specific platform for the detection or neutralization of SARS-CoV-2. Our findings highlight the potential of engineered mACE2 as a valuable tool in the fight against SARS-CoV-2. The success of developing such a small reagent based on a piecewise molecular design serves as a proof-of-concept approach for the rapid deployment of such agents to diagnose and fight other viral diseases.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/química , SARS-CoV-2/genética , Humanos , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/química , COVID-19/virologia , COVID-19/diagnóstico , Teste de Materiais , Engenharia de Proteínas , Ligação Proteica , Nanopartículas de Magnetita/química
13.
Arch Virol ; 169(6): 122, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38753071

RESUMO

Coronavirus disease 2019 (COVID-19) is still causing hospitalization and death, and vaccination appears to become less effective with each emerging variant. Spike, non-spike, and other possible unrecognized mutations have reduced the efficacy of recommended therapeutic approaches, including monoclonal antibodies, plasma transfusion, and antivirals. SARS-CoV-2 binds to angiotensin-converting enzyme 2 (ACE2) and probably dipeptidyl peptidase 4 (DPP-4) to initiate the process of endocytosis by employing host proteases such as transmembrane serine protease-2 (TMPRSS-2) and ADAM metallopeptidase domain 17 (ADAM17). Spironolactone reduces the amount of soluble ACE2 and antagonizes TMPRSS-2 and ADAM17. DPP-4 inhibitors play immunomodulatory roles and may block viral entry. The efficacy of treatment with a combination of spironolactone and DPP-4 inhibitors does not appear to be affected by viral mutations. Therefore, the combination of spironolactone and DPP-4 inhibitors might improve the clinical outcome for COVID-19 patients by decreasing the efficiency of SARS-CoV-2 entry into cells and providing better anti-inflammatory, antiproliferative, and antifibrotic effects than those achieved using current therapeutic approaches such as antivirals and monoclonal antibodies.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Inibidores da Dipeptidil Peptidase IV , SARS-CoV-2 , Espironolactona , Humanos , Espironolactona/uso terapêutico , Espironolactona/farmacologia , SARS-CoV-2/efeitos dos fármacos , Antivirais/uso terapêutico , Antivirais/farmacologia , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Inibidores da Dipeptidil Peptidase IV/farmacologia , COVID-19/virologia , Internalização do Vírus/efeitos dos fármacos , Quimioterapia Combinada , Dipeptidil Peptidase 4/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Serina Endopeptidases
14.
PLoS Pathog ; 20(5): e1012204, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38709834

RESUMO

Since the COVID-19 outbreak, raccoon dogs have been suggested as a potential intermediary in transmitting SARS-CoV-2 to humans. To understand their role in the COVID-19 pandemic and the species barrier for SARS-CoV-2 transmission to humans, we analyzed how their ACE2 protein interacts with SARS-CoV-2 spike protein. Biochemical data showed that raccoon dog ACE2 is an effective receptor for SARS-CoV-2 spike protein, though not as effective as human ACE2. Structural comparisons highlighted differences in the virus-binding residues of raccoon dog ACE2 compared to human ACE2 (L24Q, Y34H, E38D, T82M, R353K), explaining their varied effectiveness as receptors for SARS-CoV-2. These variations contribute to the species barrier that exists between raccoon dogs and humans regarding SARS-CoV-2 transmission. Identifying these barriers can help assess the susceptibility of other mammals to SARS-CoV-2. Our research underscores the potential of raccoon dogs as SARS-CoV-2 carriers and identifies molecular barriers that affect the virus's ability to jump between species.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Cães Guaxinins , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Cães Guaxinins/virologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Humanos , COVID-19/virologia , COVID-19/transmissão , COVID-19/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/química , Receptores Virais/metabolismo , Receptores Virais/química , Ligação Proteica
15.
Rev Med Virol ; 34(3): e2543, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38782605

RESUMO

COVID-19 as a pan-epidemic is waning but there it is imperative to understand virus interaction with oral tissues and oral inflammatory diseases. We review periodontal disease (PD), a common inflammatory oral disease, as a driver of COVID-19 and oral post-acute-sequelae conditions (PASC). Oral PASC identifies with PD, loss of teeth, dysgeusia, xerostomia, sialolitis-sialolith, and mucositis. We contend that PD-associated oral microbial dysbiosis involving higher burden of periodontopathic bacteria provide an optimal microenvironment for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These pathogens interact with oral epithelial cells activate molecular or biochemical pathways that promote viral adherence, entry, and persistence in the oral cavity. A repertoire of diverse molecules identifies this relationship including lipids, carbohydrates and enzymes. The S protein of SARS-CoV-2 binds to the ACE2 receptor and is activated by protease activity of host furin or TRMPSS2 that cleave S protein subunits to promote viral entry. However, PD pathogens provide additional enzymatic assistance mimicking furin and augment SARS-CoV-2 adherence by inducing viral entry receptors ACE2/TRMPSS, which are poorly expressed on oral epithelial cells. We discuss the mechanisms involving periodontopathogens and host factors that facilitate SARS-CoV-2 infection and immune resistance resulting in incomplete clearance and risk for 'long-haul' oral health issues characterising PASC. Finally, we suggest potential diagnostic markers and treatment avenues to mitigate oral PASC.


Assuntos
COVID-19 , Doenças Periodontais , SARS-CoV-2 , Humanos , COVID-19/imunologia , COVID-19/virologia , Doenças Periodontais/virologia , Doenças Periodontais/microbiologia , Disbiose/microbiologia , Enzima de Conversão de Angiotensina 2/metabolismo , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/metabolismo , Boca/virologia , Boca/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Síndrome de COVID-19 Pós-Aguda
16.
Rev Esc Enferm USP ; 58: e20230365, 2024.
Artigo em Inglês, Português | MEDLINE | ID: mdl-38743953

RESUMO

OBJECTIVE: To map the evidence in the literature about the relationship between gastrointestinal symptoms and COVID-19 in the pediatric population. METHOD: This is a scoping review following the recommendations of the Joanna Briggs Institute and PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. The search was carried out on the following bases: Embase, Google Scholar, PubMed, Scopus, LILACS, CINAHL, Scielo, Web of Science and Virtual Health Library Portal, between July and August 2023. Original studies available in full, in any language, were included. RESULTS: Ten studies were chosen that pointed to three premises: (1) the ACE2 receptor is found in the epithelial cells of the gastrointestinal tract; (2) gastrointestinal symptoms are mediated by stress and infection is justified by the gut-brain axis; (3) it develops the process of Multisystem Inflammatory Syndrome in children, affecting the gastrointestinal tract. CONCLUSION: The synthesis of evidence provided three assumptions which guide the origin of gastrointestinal symptoms. The identification of gastrointestinal symptoms in children affected by COVID-19 can assist in the clinical approach and management of care and treatments.


Assuntos
COVID-19 , Gastroenteropatias , Humanos , COVID-19/complicações , Gastroenteropatias/virologia , Gastroenteropatias/epidemiologia , Criança , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Síndrome de Resposta Inflamatória Sistêmica/diagnóstico , Eixo Encéfalo-Intestino/fisiologia , Enzima de Conversão de Angiotensina 2/metabolismo
17.
Biomolecules ; 14(5)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38785944

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mainly targets the upper respiratory tract. It gains entry by interacting with the host cell receptor angiotensin-converting enzyme 2 (ACE2) via its heavily glycosylated spike glycoprotein. SARS-CoV-2 can also affect the gastrointestinal tract. Given the significant role of glycosylation in the life cycle of proteins and the multisystem target of SARS-CoV-2, the role of glycosylation in the interaction of S1 with ACE2 in Caco-2 cells was investigated after modulation of their glycosylation patterns using N-butyldeoxynojirimycin (NB-DNJ) and 1-deoxymannojirimycin (dMM), in addition to mutant CHO cells harboring mutations at different stages of glycosylation. The data show a substantial reduction in the interactions between the altered glycosylation forms of S1 and ACE2 in the presence of NB-DNJ, while varied outcomes resulted from dMM treatment. These results highlight the promising effects of NB-DNJ and its potential use as an off-label drug to treat SARS-CoV-2 infections.


Assuntos
Enzima de Conversão de Angiotensina 2 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , Células CACO-2 , Enzima de Conversão de Angiotensina 2/metabolismo , Glicosilação , Glicoproteína da Espícula de Coronavírus/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/efeitos dos fármacos , Animais , Células CHO , Cricetulus , Transporte Proteico , COVID-19/metabolismo , COVID-19/virologia , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/análogos & derivados , Ligação Proteica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virologia
18.
Proc Natl Acad Sci U S A ; 121(23): e2407437121, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38814864

RESUMO

The accessory protease transmembrane protease serine 2 (TMPRSS2) enhances severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uptake into ACE2-expressing cells, although how increased entry impacts downstream viral and host processes remains unclear. To investigate this in more detail, we performed infection assays in engineered cells promoting ACE2-mediated entry with and without TMPRSS2 coexpression. Electron microscopy and inhibitor experiments indicated TMPRSS2-mediated cell entry was associated with increased virion internalization into endosomes, and partially dependent upon clathrin-mediated endocytosis. TMPRSS2 increased panvariant uptake efficiency and enhanced early rates of virus replication, transcription, and secretion, with variant-specific profiles observed. On the host side, transcriptional profiling confirmed the magnitude of infection-induced antiviral and proinflammatory responses were linked to uptake efficiency, with TMPRSS2-assisted entry boosting early antiviral responses. In addition, TMPRSS2-enhanced infections increased rates of cytopathology, apoptosis, and necrosis and modulated virus secretion kinetics in a variant-specific manner. On the virus side, convergent signatures of cell-uptake-dependent innate immune induction were recorded in viral genomes, manifesting as switches in dominant coupled Nsp3 residues whose frequencies were correlated to the magnitude of the cellular response to infection. Experimentally, we demonstrated that selected Nsp3 mutations conferred enhanced interferon antagonism. More broadly, we show that TMPRSS2 orthologues from evolutionarily diverse mammals facilitate panvariant enhancement of cell uptake. In summary, our study uncovers previously unreported associations, linking cell entry efficiency to innate immune activation kinetics, cell death rates, virus secretion dynamics, and convergent selection of viral mutations. These data expand our understanding of TMPRSS2's role in the SARS-CoV-2 life cycle and confirm its broader significance in zoonotic reservoirs and animal models.


Assuntos
COVID-19 , Imunidade Inata , SARS-CoV-2 , Serina Endopeptidases , Internalização do Vírus , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , SARS-CoV-2/metabolismo , Humanos , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , COVID-19/virologia , COVID-19/imunologia , COVID-19/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Replicação Viral , Animais , Endocitose , Células HEK293 , Chlorocebus aethiops , Citologia
19.
Front Immunol ; 15: 1374541, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38807598

RESUMO

Objective: The coronavirus disease 2019 (COVID-19) spread rapidly and claimed millions of lives worldwide. Acute respiratory distress syndrome (ARDS) is the major cause of COVID-19-associated deaths. Due to the limitations of current drugs, developing effective therapeutic options that can be used rapidly and safely in clinics for treating severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections is necessary. This study aims to investigate the effects of two food-extracted immunomodulatory agents, ajoene-enriched garlic extract (AGE) and cruciferous vegetables-extracted sulforaphane (SFN), on anti-inflammatory and immune responses in a SARS-CoV-2 acute lung injury mouse model. Methods: In this study, we established a mouse model to mimic the SARS-CoV-2 infection acute lung injury model via intratracheal injection of polyinosinic:polycytidylic acid (poly[I:C]) and SARS-CoV-2 recombinant spike protein (SP). After the different agents treatment, lung sections, bronchoalveolar lavage fluid (BALF) and fresh faeces were harvested. Then, H&E staining was used to examine symptoms of interstitial pneumonia. Flow cytometry was used to examine the change of immune cell populations. Multiplex cytokines assay was used to examine the inflammatory cytokines.16S rDNA high-throughput sequencing was used to examine the change of gut microbiome. Results: Our results showed that AGE and SFN significantly suppressed the symptoms of interstitial pneumonia, effectively inhibited the production of inflammatory cytokines, decreased the percentage of inflammatory cell populations, and elevated T cell populations in the mouse model. Furthermore, we also observed that the gut microbiome of genus Paramuribaculum were enriched in the AGE-treated group. Conclusion: Here, for the first time, we observed that these two novel, safe, and relatively inexpensive immunomodulatory agents exhibited the same effects on anti-inflammatory and immune responses as neutralizing monoclonal antibodies (mAbs) against interleukin 6 receptor (IL-6R), which have been suggested for treating COVID-19 patients. Our results revealed the therapeutic ability of these two immunomodulatory agents in a mouse model of SARS-CoV-2 acute lung injury by promoting anti-inflammatory and immune responses. These results suggest that AGE and SFN are promising candidates for the COVID-19 treatment.


Assuntos
Lesão Pulmonar Aguda , Enzima de Conversão de Angiotensina 2 , Anti-Inflamatórios , Tratamento Farmacológico da COVID-19 , COVID-19 , Modelos Animais de Doenças , Agentes de Imunomodulação , SARS-CoV-2 , Animais , Camundongos , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/etiologia , COVID-19/imunologia , SARS-CoV-2/imunologia , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Isotiocianatos/farmacologia , Isotiocianatos/uso terapêutico , Sulfóxidos , Humanos , Citocinas/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Pulmão/efeitos dos fármacos , Masculino , Poli I-C , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico
20.
FASEB J ; 38(10): e23692, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38786655

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection involves an initial viral infection phase followed by a host-response phase that includes an eicosanoid and cytokine storm, lung inflammation and respiratory failure. While vaccination and early anti-viral therapies are effective in preventing or limiting the pathogenic host response, this latter phase is poorly understood with no highly effective treatment options. Inhibitors of soluble epoxide hydrolase (sEH) increase levels of anti-inflammatory molecules called epoxyeicosatrienoic acids (EETs). This study aimed to investigate the impact of sEH inhibition on the host response to SARS-CoV-2 infection in a mouse model with human angiotensin-converting enzyme 2 (ACE2) expression. Mice were infected with SARS-CoV-2 and treated with either vehicle or the sEH inhibitor 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU). At day 5 post-infection, SARS-CoV-2 induced weight loss, clinical signs, a cytokine storm, an eicosanoid storm, and severe lung inflammation with ~50% mortality on days 6-8 post-infection. SARS-CoV-2 infection induced lung expression of phospholipase A2 (PLA2), cyclooxygenase (COX) and lipoxygenase (LOX) pathway genes, while suppressing expression of most cytochrome P450 genes. Treatment with the sEH inhibitor TPPU delayed weight loss but did not alter clinical signs, lung cytokine expression or overall survival of infected mice. Interestingly, TPPU treatment significantly reversed the eicosanoid storm and attenuated viral-induced elevation of 39 fatty acids and oxylipins from COX, LOX and P450 pathways, which suggests the effects at the level of PLA2 activation. The suppression of the eicosanoid storm by TPPU without corresponding changes in lung cytokines, lung inflammation or mortality reveals a surprising dissociation between systemic oxylipin and cytokine signaling pathways during SARS-CoV-2 infection and suggests that the cytokine storm is primarily responsible for morbidity and mortality in this animal model.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Síndrome da Liberação de Citocina , Eicosanoides , Epóxido Hidrolases , SARS-CoV-2 , Animais , Camundongos , Eicosanoides/metabolismo , COVID-19/imunologia , COVID-19/virologia , COVID-19/metabolismo , SARS-CoV-2/efeitos dos fármacos , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/metabolismo , Síndrome da Liberação de Citocina/tratamento farmacológico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Citocinas/metabolismo , Humanos , Pulmão/virologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/metabolismo , Modelos Animais de Doenças , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Feminino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA