Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.241
Filtrer
1.
Signal Transduct Target Ther ; 9(1): 142, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38825657

RÉSUMÉ

Radiotherapy combined with immune checkpoint blockade holds great promise for synergistic antitumor efficacy. Targeted radionuclide therapy delivers radiation directly to tumor sites. LNC1004 is a fibroblast activation protein (FAP)-targeting radiopharmaceutical, conjugated with the albumin binder Evans Blue, which has demonstrated enhanced tumor uptake and retention in previous preclinical and clinical studies. Herein, we demonstrate that 68Ga/177Lu-labeled LNC1004 exhibits increased uptake and prolonged retention in MC38/NIH3T3-FAP and CT26/NIH3T3-FAP tumor xenografts. Radionuclide therapy with 177Lu-LNC1004 induced a transient upregulation of PD-L1 expression in tumor cells. The combination of 177Lu-LNC1004 and anti-PD-L1 immunotherapy led to complete eradication of all tumors in MC38/NIH3T3-FAP tumor-bearing mice, with mice showing 100% tumor rejection upon rechallenge. Immunohistochemistry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing revealed that combination therapy reprogrammed the tumor microenvironment in mice to foster antitumor immunity by suppressing malignant progression and increasing cell-to-cell communication, CD8+ T-cell activation and expansion, M1 macrophage counts, antitumor activity of neutrophils, and T-cell receptor diversity. A preliminary clinical study demonstrated that 177Lu-LNC1004 was well-tolerated and effective in patients with refractory cancers. Further, scRNA-seq of peripheral blood mononuclear cells underscored the importance of addressing immune evasion through immune checkpoint blockade treatment. This was emphasized by the observed increase in antigen processing and presentation juxtaposed with T cell inactivation. In conclusion, our data supported the efficacy of immunotherapy combined with 177Lu-LNC1004 for cancer patients with FAP-positive tumors.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Animaux , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Humains , Protéines membranaires/génétique , Protéines membranaires/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Endopeptidases/génétique , Cellules NIH 3T3 , Radiopharmaceutiques/usage thérapeutique , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Immunothérapie , Gelatinases/génétique , Gelatinases/immunologie , Lutétium/pharmacologie , Lignée cellulaire tumorale
2.
Nat Commun ; 15(1): 4056, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38744813

RÉSUMÉ

The fusion peptide of SARS-CoV-2 spike protein is functionally important for membrane fusion during virus entry and is part of a broadly neutralizing epitope. However, sequence determinants at the fusion peptide and its adjacent regions for pathogenicity and antigenicity remain elusive. In this study, we perform a series of deep mutational scanning (DMS) experiments on an S2 region spanning the fusion peptide of authentic SARS-CoV-2 in different cell lines and in the presence of broadly neutralizing antibodies. We identify mutations at residue 813 of the spike protein that reduced TMPRSS2-mediated entry with decreased virulence. In addition, we show that an F823Y mutation, present in bat betacoronavirus HKU9 spike protein, confers resistance to broadly neutralizing antibodies. Our findings provide mechanistic insights into SARS-CoV-2 pathogenicity and also highlight a potential challenge in developing broadly protective S2-based coronavirus vaccines.


Sujet(s)
Anticorps neutralisants , COVID-19 , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Pénétration virale , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Anticorps neutralisants/immunologie , COVID-19/virologie , COVID-19/immunologie , Animaux , Anticorps antiviraux/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Chlorocebus aethiops , Cellules HEK293 , Cellules Vero , Épitopes/immunologie , Épitopes/génétique , Lignée cellulaire , Souris
3.
J Virol ; 98(5): e0195723, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38557247

RÉSUMÉ

Zoonotic coronaviruses pose a continuous threat to human health, with newly identified bat-borne viruses like swine acute diarrhea syndrome coronavirus (SADS-CoV) causing high mortality in piglets. In vitro studies indicate that SADS-CoV can infect cell lines from diverse species, including humans, highlighting its potential risk to human health. However, the lack of tools to study viral entry, along with the absence of vaccines or antiviral therapies, perpetuates this threat. To address this, we engineered an infectious molecular clone of Vesicular Stomatitis Virus (VSV), replacing its native glycoprotein (G) with SADS-CoV spike (S) and inserting a Venus reporter at the 3' leader region to generate a replication-competent rVSV-Venus-SADS S virus. Serial passages of rVSV-Venus-SADS S led to the identification of an 11-amino-acid truncation in the cytoplasmic tail of the S protein, which allowed more efficient viral propagation due to increased cell membrane anchoring of the S protein. The S protein was integrated into rVSV-Venus-SADS SΔ11 particles, susceptible to neutralization by sera from SADS-CoV S1 protein-immunized rabbits. Additionally, we found that TMPRSS2 promotes SADS-CoV spike-mediated cell entry. Furthermore, we assessed the serum-neutralizing ability of mice vaccinated with rVSV-Venus-SADS SΔ11 using a prime-boost immunization strategy, revealing effective neutralizing antibodies against SADS-CoV infection. In conclusion, we have developed a safe and practical tool for studying SADS-CoV entry and exploring the potential of a recombinant VSV-vectored SADS-CoV vaccine.IMPORTANCEZoonotic coronaviruses, like swine acute diarrhea syndrome coronavirus (SADS-CoV), pose a continual threat to human and animal health. To combat this, we engineered a safe and efficient tool by modifying the Vesicular Stomatitis Virus (VSV), creating a replication-competent rVSV-Venus-SADS S virus. Through serial passages, we optimized the virus for enhanced membrane anchoring, a key factor in viral propagation. This modified virus, rVSV-Venus-SADS SΔ11, proved susceptible to neutralization, opening avenues for potential vaccines. Additionally, our study revealed the role of TMPRSS2 in SADS-CoV entry. Mice vaccinated with rVSV-Venus-SADS SΔ11 developed potent neutralizing antibodies against SADS-CoV. In conclusion, our work presents a secure and practical tool for studying SADS-CoV entry and explores the promise of a recombinant VSV-vectored SADS-CoV vaccine.


Sujet(s)
Alphacoronavirus , Pénétration virale , Réplication virale , Animaux , Humains , Souris , Lapins , Alphacoronavirus/génétique , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Lignée cellulaire , Chlorocebus aethiops , Infections à coronavirus/virologie , Infections à coronavirus/prévention et contrôle , Cellules HEK293 , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Serine endopeptidases/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme , Suidae , Cellules Vero , Virus de la stomatite vésiculeuse de type Indiana/génétique , Vesiculovirus/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique
4.
Clin Cancer Res ; 30(9): 1859-1877, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38393682

RÉSUMÉ

PURPOSE: Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN: Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS: We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS: CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.


Sujet(s)
Antigènes CD3 , Endopeptidases , Protéines liées au GPI , Immunothérapie adoptive , Mésothéline , Tumeurs du pancréas , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Souris , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Microenvironnement tumoral/immunologie , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Protéines liées au GPI/immunologie , Protéines liées au GPI/métabolisme , Lignée cellulaire tumorale , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/thérapie , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Protéines membranaires/immunologie , Protéines membranaires/métabolisme , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Adénocarcinome/immunologie , Adénocarcinome/thérapie , Adénocarcinome/anatomopathologie
5.
Front Immunol ; 13: 832306, 2022.
Article de Anglais | MEDLINE | ID: mdl-36091026

RÉSUMÉ

Neutrophils play major roles against bacteria and fungi infections not only due to their microbicide properties but also because they release mediators like Interleukin-1 beta (IL-1ß) that contribute to orchestrate the inflammatory response. This cytokine is a leaderless protein synthesized in the cytoplasm as a precursor (pro-IL-1ß) that is proteolytically processed to its active isoform and released from human neutrophils by secretory autophagy. In most myeloid cells, pro-IL-1ß is processed by caspase-1 upon inflammasome activation. Here we employed neutrophils from both healthy donors and patients with a gain-of-function (GOF) NLRP3-mutation to dissect IL-1ß processing in these cells. We found that although caspase-1 is required for IL-1ß secretion, it undergoes rapid inactivation, and instead, neutrophil serine proteases play a key role in pro-IL-1ß processing. Our findings bring to light distinctive features of the regulation of caspase-1 activity in human neutrophils and reveal new molecular mechanisms that control human neutrophil IL-1ß secretion.


Sujet(s)
Autophagie , Caspase-1 , Interleukine-1 bêta , Granulocytes neutrophiles , Protéases à sérine , Autophagie/génétique , Autophagie/immunologie , Caspase-1/génétique , Caspase-1/métabolisme , Humains , Inflammasomes/génétique , Inflammasomes/immunologie , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Granulocytes neutrophiles/enzymologie , Granulocytes neutrophiles/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Protéases à sérine/génétique , Protéases à sérine/immunologie
6.
Front Immunol ; 13: 872047, 2022.
Article de Anglais | MEDLINE | ID: mdl-35585971

RÉSUMÉ

An effective COVID-19 vaccine against broad SARS-CoV-2 variants is still an unmet need. In the study, the vesicular stomatitis virus (VSV)-based vector was used to express the SARS-CoV-2 Spike protein to identify better vaccine designs. The replication-competent of the recombinant VSV-spike virus with C-terminal 19 amino acid truncation (SΔ19 Rep) was generated. A single dose of SΔ19 Rep intranasal vaccination is sufficient to induce protective immunity against SARS-CoV-2 infection in hamsters. All the clones isolated from the SΔ19 Rep virus contained R682G mutation located at the Furin cleavage site. An additional S813Y mutation close to the TMPRSS2 cleavage site was identified in some clones. The enzymatic processing of S protein was blocked by these mutations. The vaccination of the R682G-S813Y virus produced a high antibody response against S protein and a robust S protein-specific CD8+ T cell response. The vaccinated animals were protected from the lethal SARS-CoV-2 (delta variant) challenge. The S antigen with resistance to enzymatic processes by Furin and TMPRSS2 will provide better immunogenicity for vaccine design.


Sujet(s)
COVID-19 , Furine , SARS-CoV-2 , Serine endopeptidases , Animaux , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Vaccins contre la COVID-19 , Furine/génétique , Furine/métabolisme , Humains , Immunité cellulaire , SARS-CoV-2/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Glycoprotéine de spicule des coronavirus/immunologie
7.
Aging (Albany NY) ; 14(1): 73-108, 2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-35017320

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread around the world and became a global pandemic in 2020. One promising drug target for SARS-CoV-2 is the transmembrane protease serine 2 (TMPRSS2). This study was designed to explore the expression status, prognostic significance and molecular functions of TMPRSS2 in lung cancer. TMPRSS2 expression was investigated using the TIMER, Oncomine, UALCAN, GEO, HPA and TCGA databases. The prognostic value of TMPRSS2 was examined using Cox regression and a nomogram. KEGG, GO and GSEA were performed to investigate the cellular function of TMPRSS2 in lung cancer. The relationship between TMPRSS2 and immune infiltration was determined using the TIMER and CIBERSORT algorithms. TMPRSS2 mRNA and protein expression was significantly reduced in lung cancer. Decreased TMPRSS2 expression and increased DNA methylation of TMPRSS2 were associated with various clinicopathological parameters in patients with lung cancer. Low TMPRSS2 mRNA expression also correlated with poor outcome in lung cancer patients. Moreover, a nomogram was constructed and exhibited good predictive power for the overall survival of lung cancer patients. KEGG and GO analyses and GSEA implied that multiple immune- and metabolism-related pathways were significantly linked with TMPRSS2 expression. Intriguingly, TMPRSS2 expression associated with immune cell infiltration in lung cancer. More importantly, TMPRSS2 expression was markedly decreased in SARS-CoV-infected cells. These findings indicate that TMPRSS2 may be a promising prognostic biomarker and therapeutic target for lung cancer through metabolic pathways and immune cell infiltration.


Sujet(s)
COVID-19/génétique , Système immunitaire/immunologie , Tumeurs du poumon/génétique , SARS-CoV-2/physiologie , Serine endopeptidases/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , COVID-19/complications , COVID-19/immunologie , COVID-19/virologie , Femelle , Interactions hôte-pathogène , Humains , Tumeurs du poumon/complications , Tumeurs du poumon/immunologie , Tumeurs du poumon/mortalité , Mâle , Voies et réseaux métaboliques , Adulte d'âge moyen , SARS-CoV-2/génétique , Serine endopeptidases/immunologie , Jeune adulte
8.
Am J Respir Cell Mol Biol ; 66(2): 206-222, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34731594

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected more than 180 million people since the onset of the pandemic. Despite similar viral load and infectivity rates between children and adults, children rarely develop severe illness. Differences in the host response to the virus at the primary infection site are among the mechanisms proposed to account for this disparity. Our objective was to investigate the host response to SARS-CoV-2 in the nasal mucosa in children and adults and compare it with the host response to respiratory syncytial virus (RSV) and influenza virus. We analyzed clinical outcomes and gene expression in the nasal mucosa of 36 children with SARS-CoV-2, 24 children with RSV, 9 children with influenza virus, 16 adults with SARS-CoV-2, and 7 healthy pediatric and 13 healthy adult controls. In both children and adults, infection with SARS-CoV-2 led to an IFN response in the nasal mucosa. The magnitude of the IFN response correlated with the abundance of viral reads, not the severity of illness, and was comparable between children and adults infected with SARS-CoV-2 and children with severe RSV infection. Expression of ACE2 and TMPRSS2 did not correlate with age or presence of viral infection. SARS-CoV-2-infected adults had increased expression of genes involved in neutrophil activation and T-cell receptor signaling pathways compared with SARS-CoV-2-infected children, despite similar severity of illness and viral reads. Age-related differences in the immune response to SARS-CoV-2 may place adults at increased risk of developing severe illness.


Sujet(s)
Vieillissement/immunologie , COVID-19/immunologie , Régulation de l'expression des gènes/immunologie , Immunité muqueuse , Muqueuse nasale/immunologie , SARS-CoV-2/immunologie , Adolescent , Facteurs âges , Angiotensin-converting enzyme 2/immunologie , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Muqueuse nasale/virologie , Infections à virus respiratoire syncytial/immunologie , Virus respiratoires syncytiaux/immunologie , Serine endopeptidases/immunologie
9.
J Cell Mol Med ; 26(1): 25-34, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34904376

RÉSUMÉ

Transmission electron microscopy has historically been indispensable for virology research, as it offers unique insight into virus function. In the past decade, as cryo-electron microscopy (cryo-EM) has matured and become more accessible, we have been able to peer into the structure of viruses at the atomic level and understand how they interact with the host cell, with drugs or with antibodies. Perhaps, there was no time in recent history where cryo-EM was more needed, as SARS-CoV-2 has spread around the globe, causing millions of deaths and almost unquantifiable economic devastation. In this concise review, we aim to mark the most important contributions of cryo-EM to understanding the structure and function of SARS-CoV-2 proteins, from surface spikes to the virus core and from virus-receptor interactions to antibody binding.


Sujet(s)
Angiotensin-converting enzyme 2/composition chimique , Anticorps antiviraux/composition chimique , Vaccins contre la COVID-19/composition chimique , COVID-19/prévention et contrôle , Récepteurs viraux/composition chimique , Glycoprotéine de spicule des coronavirus/composition chimique , Angiotensin-converting enzyme 2/immunologie , Angiotensin-converting enzyme 2/métabolisme , Anticorps antiviraux/biosynthèse , COVID-19/immunologie , COVID-19/virologie , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/biosynthèse , Cryomicroscopie électronique , Épitopes/composition chimique , Épitopes/immunologie , Épitopes/métabolisme , Humains , Modèles moléculaires , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Structure secondaire des protéines , Récepteurs viraux/immunologie , Récepteurs viraux/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/pathogénicité , SARS-CoV-2/ultrastructure , Serine endopeptidases/composition chimique , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/métabolisme , Virion/effets des médicaments et des substances chimiques , Virion/pathogénicité , Virion/ultrastructure
10.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34902435

RÉSUMÉ

BACKGROUND: Treatments for coronavirus disease 2019, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are urgently needed but remain limited. SARS-CoV-2 infects cells through interactions of its spike (S) protein with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) on host cells. Multiple cells and organs are targeted, particularly airway epithelial cells. OM-85, a standardized lysate of human airway bacteria with strong immunomodulating properties and an impeccable safety profile, is widely used to prevent recurrent respiratory infections. We found that airway OM-85 administration inhibits Ace2 and Tmprss2 transcription in the mouse lung, suggesting that OM-85 might hinder SARS-CoV-2/host cell interactions. OBJECTIVES: We sought to investigate whether and how OM-85 treatment protects nonhuman primate and human epithelial cells against SARS-CoV-2. METHODS: ACE2 and TMPRSS2 mRNA and protein expression, cell binding of SARS-CoV-2 S1 protein, cell entry of SARS-CoV-2 S protein-pseudotyped lentiviral particles, and SARS-CoV-2 cell infection were measured in kidney, lung, and intestinal epithelial cell lines, primary human bronchial epithelial cells, and ACE2-transfected HEK293T cells treated with OM-85 in vitro. RESULTS: OM-85 significantly downregulated ACE2 and TMPRSS2 transcription and surface ACE2 protein expression in epithelial cell lines and primary bronchial epithelial cells. OM-85 also strongly inhibited SARS-CoV-2 S1 protein binding to, SARS-CoV-2 S protein-pseudotyped lentivirus entry into, and SARS-CoV-2 infection of epithelial cells. These effects of OM-85 appeared to depend on SARS-CoV-2 receptor downregulation. CONCLUSIONS: OM-85 inhibits SARS-CoV-2 epithelial cell infection in vitro by downregulating SARS-CoV-2 receptor expression. Further studies are warranted to assess whether OM-85 may prevent and/or reduce the severity of coronavirus disease 2019.


Sujet(s)
Adjuvants immunologiques/administration et posologie , COVID-19/prévention et contrôle , Extrait cellulaire/administration et posologie , Récepteurs viraux/antagonistes et inhibiteurs , Récepteurs viraux/immunologie , SARS-CoV-2/immunologie , Angiotensin-converting enzyme 2/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Animaux , COVID-19/immunologie , COVID-19/virologie , Cellules Caco-2 , Extrait cellulaire/immunologie , Cellules cultivées , Chlorocebus aethiops , Régulation négative/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Cellules HEK293 , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/immunologie , Humains , Techniques in vitro , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Poumon/virologie , Souris , Souris de lignée BALB C , Serine endopeptidases/effets des médicaments et des substances chimiques , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Transcription génétique/effets des médicaments et des substances chimiques , Transcription génétique/immunologie , Cellules Vero
11.
J Biosci ; 462021.
Article de Anglais | MEDLINE | ID: mdl-34785628

RÉSUMÉ

Since the start of the pandemic, SARS-CoV-2 has infected almost 200 million human hosts and is set to encounter and gain entry in many more in the coming months. As the coronavirus flourish, the evolutionary pressure selects those variants that can complete the infection cycle faster and reproduce in large numbers compared to others. This increase in infectivity and transmissibility coupled with the immune response from high viral load may cause moderate to severe disease. Whether this leads to enhanced virulence in the prevalent Alpha and Delta variants is still not clear. This review describes the different types of SARS-CoV-2 variants that are now prevalent, their emergence, the mutations responsible for their growth advantages, and how they affect vaccine efficacy and increase chances of reinfection. Finally, we have also summarized the efforts made to recognize and predict the mutations, which can cause immune escape and track their emergence through impactful genomic surveillance.


Sujet(s)
Anticorps neutralisants/composition chimique , COVID-19/épidémiologie , Génome viral , Échappement immunitaire/génétique , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/composition chimique , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Anticorps neutralisants/génétique , Anticorps neutralisants/immunologie , Sites de fixation , COVID-19/anatomopathologie , COVID-19/transmission , COVID-19/virologie , Vaccins contre la COVID-19 , Humains , Modèles moléculaires , Mutation , Phylogenèse , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Récepteurs viraux/composition chimique , Récepteurs viraux/génétique , Récepteurs viraux/immunologie , SARS-CoV-2/classification , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , Serine endopeptidases/composition chimique , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Virulence
12.
Clin Immunol ; 233: 108879, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34798239

RÉSUMÉ

COVID-19 is a pandemic requiring immediate solution for treatment because of its complex pathophysiology. Exploration of novel targets and thus treatment will be life savers which is the need of the hour. 2 host factors- TMPRSS2 and ACE2 are responsible for the way the virus will enter and replicate in the host. Also NRF2 is an important protein responsible for its anti-inflammatory role by multiple mechanisms of action like inhibition of NF-kB, suppression of pro-inflammatory genes, etc. NRF2 is deacetylated by Sirtuins and therefore both have a direct association. Absence of SIRT indicates inhibition of NRF2 expression and thus no anti-oxidative and anti-inflammatory protection for the cell. Therefore, we propose that NRF2 activators and/or SIRT activators can be evaluated to check their efficacy in ameliorating the symptoms of COVID-19.


Sujet(s)
COVID-19/immunologie , Facteur-2 apparenté à NF-E2/immunologie , SARS-CoV-2/immunologie , Sirtuines/immunologie , Angiotensin-converting enzyme 2/immunologie , Angiotensin-converting enzyme 2/métabolisme , Animaux , COVID-19/métabolisme , COVID-19/virologie , Interactions hôte-pathogène/immunologie , Humains , Facteur-2 apparenté à NF-E2/métabolisme , SARS-CoV-2/métabolisme , SARS-CoV-2/physiologie , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Sirtuines/métabolisme , Attachement viral
13.
Biosci Rep ; 41(10)2021 10 29.
Article de Anglais | MEDLINE | ID: mdl-34647577

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has become a global health emergency. Although new vaccines have been generated and being implicated, discovery and application of novel preventive and control measures are warranted. We aimed to identify compounds that may possess the potential to either block the entry of virus to host cells or attenuate its replication upon infection. Using host cell surface receptor expression (angiotensin-converting enzyme 2 (ACE2) and Transmembrane protease serine 2 (TMPRSS2)) analysis as an assay, we earlier screened several synthetic and natural compounds and identified candidates that showed ability to down-regulate their expression. Here, we report experimental and computational analyses of two small molecules, Mortaparib and MortaparibPlus that were initially identified as dual novel inhibitors of mortalin and PARP-1, for their activity against SARS-CoV-2. In silico analyses showed that MortaparibPlus, but not Mortaparib, stably binds into the catalytic pocket of TMPRSS2. In vitro analysis of control and treated cells revealed that MortaparibPlus caused down-regulation of ACE2 and TMPRSS2; Mortaparib did not show any effect. Furthermore, computational analysis on SARS-CoV-2 main protease (Mpro) that also predicted the inhibitory activity of MortaparibPlus. However, cell-based antiviral drug screening assay showed 30-60% viral inhibition in cells treated with non-toxic doses of either MortaparibPlus or Mortaparib. The data suggest that these two closely related compounds possess multimodal anti-COVID-19 activities. Whereas MortaparibPlus works through direct interactions/effects on the host cell surface receptors (ACE2 and TMPRSS2) and the virus protein (Mpro), Mortaparib involves independent mechanisms, elucidation of which warrants further studies.


Sujet(s)
Antiviraux/pharmacologie , Traitements médicamenteux de la COVID-19 , Biologie informatique/méthodes , Angiotensin-converting enzyme 2/immunologie , Angiotensin-converting enzyme 2/métabolisme , Antiviraux/immunologie , COVID-19/immunologie , Lignée cellulaire tumorale , Évaluation préclinique de médicament/méthodes , Protéines du choc thermique HSP70/antagonistes et inhibiteurs , Humains , Protéines mitochondriales/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , SARS-CoV-2/immunologie , Serine endopeptidases/immunologie , Serine endopeptidases/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques
14.
Eur J Pharmacol ; 912: 174548, 2021 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-34606834

RÉSUMÉ

The importance of sex differences is increasingly acknowledged in the incidence and treatment of disease. Accumulating clinical evidence demonstrates that sex differences are noticeable in COVID-19, and the prevalence, severity, and mortality rate of COVID-19 are higher among males than females. Sex-related genetic and hormonal factors and immunological responses may underlie the sex bias in COVID-19 patients. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2) are essential proteins involved in the cell entry of SARS-CoV-2. Since ACE2 is encoded on the X-chromosome, a double copy of ACE2 in females may compensate for virus-mediated downregulation of ACE2, and thus ACE2-mediated cellular protection is greater in females. The X chromosome also contains the largest immune-related genes leading females to develop more robust immune responses than males. Toll-like receptor-7 (TLR-7), one of the key players in innate immunity, is linked to sex differences in autoimmunity and vaccine efficacy, and its expression is greater in females. Sex steroids also affect immune cell function. Estrogen contributes to higher CD4+ and CD8+ T cell activation levels, and females have more B cells than males. Sex differences not only affect the severity and progression of the disease, but also alter the efficacy of pharmacological treatment and adverse events related to the drugs/vaccines used against COVID-19. Administration of different drugs/vaccines in different doses or intervals may be useful to eliminate sex differences in efficacy and side/adverse effects. It should be noted that studies should include sex-specific analyses to develop further sex-specific treatments for COVID-19.


Sujet(s)
COVID-19/étiologie , COVID-19/génétique , Prédisposition génétique à une maladie/génétique , Adulte , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , COVID-19/immunologie , Oestrogènes/génétique , Oestrogènes/immunologie , Femelle , Humains , Immunité innée/génétique , Immunité innée/immunologie , Mâle , Adulte d'âge moyen , SARS-CoV-2/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Caractères sexuels , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
15.
PLoS One ; 16(9): e0257705, 2021.
Article de Anglais | MEDLINE | ID: mdl-34534255

RÉSUMÉ

SARS-CoV-2 enters host cells when the viral spike protein is cleaved by transmembrane protease serine 2 (TMPRSS2) after binding to the host angiotensin-converting enzyme 2 (ACE2). Since ACE2 and TMPRSS2 are expressed in the tongue and gingival mucosa, the oral cavity is a potential entry point for SARS-CoV-2. This study evaluated the inhibitory effects of general ingredients of toothpastes and mouthwashes on the spike protein-ACE2 interaction and the TMPRSS2 protease activity using an in vitro assay. Both assays detected inhibitory effects of sodium tetradecene sulfonate, sodium N-lauroyl-N-methyltaurate, sodium N-lauroylsarcosinate, sodium dodecyl sulfate, and copper gluconate. Molecular docking simulations suggested that these ingredients could bind to inhibitor-binding site of ACE2. Furthermore, tranexamic acid exerted inhibitory effects on TMPRSS2 protease activity. Our findings suggest that these toothpaste and mouthwash ingredients could help prevent SARS-CoV-2 infection.


Sujet(s)
COVID-19/prévention et contrôle , Bains de bouche/pharmacologie , Hygiène buccodentaire/méthodes , SARS-CoV-2/effets des médicaments et des substances chimiques , Pâtes dentifrices/pharmacologie , Pénétration virale/effets des médicaments et des substances chimiques , Angiotensin-converting enzyme 2/immunologie , Humains , Serine endopeptidases/immunologie , Glycoprotéine de spicule des coronavirus/immunologie
16.
Int J Mol Sci ; 22(16)2021 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-34445373

RÉSUMÉ

Human ACE2 and the serine protease TMPRSS2 of novel SARS-CoV-2 are primary entry receptors in host cells. Expression of these genes at the transcriptional level has not been much discussed in detail. The ISRE elements of the ACE2 promoter are a binding site for the ISGF3 complex of the JAK/STAT signaling pathway. TMPRSS2, including IFNß, STAT1, and STAT2, has the PARP1 binding site near to TSS either up or downstream promoter region. It is well documented that PARP1 regulates gene expression at the transcription level. Therefore, to curb virus infection, both promoting type I IFN signaling to boost innate immunity and prevention of virus entry by inhibiting PARP1, ACE2 or TMPRSS2 are safe options. Most importantly, our aim is to attract the attention of the global scientific community towards the codon 72 Single Nucleotide Polymorphism (SNP) of p53 and its underneath role in the innate immune response against SARS-CoV-2. Here, we discuss codon 72 SNP of human p53's role in the different innate immune response to restrict virus-mediated mortality rate only in specific parts of the world. In addition, we discuss potential targets and emerging therapies using bioengineered bacteriophage, anti-sense, or CRISPR strategies.


Sujet(s)
Angiotensin-converting enzyme 2/génétique , COVID-19/génétique , COVID-19/immunologie , SARS-CoV-2/génétique , Serine endopeptidases/génétique , Angiotensin-converting enzyme 2/composition chimique , Angiotensin-converting enzyme 2/immunologie , Sites de fixation , COVID-19/virologie , Humains , Immunité innée , Poly (ADP-Ribose) polymerase-1/composition chimique , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/immunologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Polymorphisme de nucléotide simple , SARS-CoV-2/physiologie , Serine endopeptidases/composition chimique , Serine endopeptidases/immunologie , Vaccination , Pénétration virale
17.
Int J Biol Macromol ; 186: 984-993, 2021 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-34293361

RÉSUMÉ

Zika virus (ZIKV) infections were associated with neurological disorders only after the Brazilian outbreak in 2015. The lack of vaccines and precise diagnosis requires a precise method to detect ZIKV infection. This study aimed to evaluate three ZIKV recombinant proteins for the development of ZIKV infections. Here, it was purified stable recombinant ZIKV Capsid (r-ZIKV-c), non-structural proteins NS1 (r-ZIKV-NS1), and NS3 (r-ZIKV-NS3) for detection of the infection by ZIKV in blood sera of patients. A commercial polyclonal antibody recognized the r-ZIKV-NS1. Here, among three proteins, NS1 showed the best result for diagnostic purposes using serum samples, despite the high similarity with NS1 from DENV, and could differentiate the infections. The recombinant NS1 was used to produce a monoclonal antibody to differentiate between DENV and ZIKV NS1. As for recombinant proteins, the result for r-ZIKV-NS1 values showed 77% and 100% sensitivity and specificity, respectively, in the IgM assay. Our data showed the protein could successfully differentiate between sera of ZIKV infected patients from sera of those not infected with the virus and differentiate from sera of DENV infected patients. Thus, the generated recombinant proteins have great potential for serological diagnosis of ZIKV in Brazil, where it is indispensable.


Sujet(s)
Anticorps antiviraux/sang , Test ELISA , Tests sérologiques , Protéines virales non structurales/immunologie , Infection par le virus Zika/diagnostic , Virus Zika/immunologie , Anticorps monoclonaux/immunologie , Spécificité des anticorps , Marqueurs biologiques/sang , Protéines de capside/génétique , Protéines de capside/immunologie , Études cas-témoins , Dengue/diagnostic , Dengue/virologie , Diagnostic différentiel , Femelle , Humains , Valeur prédictive des tests , Grossesse , Protéines recombinantes/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Protéines virales non structurales/sang , Protéines virales non structurales/génétique , Protéines virales/génétique , Protéines virales/immunologie , Virus Zika/génétique , Infection par le virus Zika/sang , Infection par le virus Zika/immunologie , Infection par le virus Zika/virologie
18.
EBioMedicine ; 70: 103500, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-34311326

RÉSUMÉ

BACKGROUND: The outbreak of Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection has become a global health emergency. We aim to decipher SARS-CoV-2 infected cell types, the consequent host immune response and their interplay in lung of COVID-19 patients. METHODS: We analyzed single-cell RNA sequencing (scRNA-seq) data of bronchoalveolar lavage fluid (BALF) samples from 10 healthy donors, 6 severe COVID-19 patients and 3 mild recovered patients. The expressions of SARS-CoV-2 receptors (ACE2 and TMPRSS2) were examined among different cell types. The immune cells infiltration patterns, their expression profiles, and interplays between immune cells and SARS-CoV-2 target cells were further investigated. FINDINGS: Compared to healthy controls, ACE2 and TMPRSS2 expressions were significantly higher in lung epithelial cells of COVID-19 patients, in particular club and ciliated cells. SARS-CoV-2 activated pro-inflammatory genes and interferon/cytokine signaling in these cells. In severe COVID-19 patients, significantly higher neutrophil, but lower macrophage in lung was observed along with markedly increased cytokines expression compared with healthy controls and mild patients. By contrast, neutrophil and macrophage returned to normal level whilst more T and NK cells accumulation were observed in mild patients. Moreover, SARS-CoV-2 infection altered the community interplays of lung epithelial and immune cells: interactions between the club and immune cells were higher in COVID-19 patients compared to healthy donors; on the other hand, immune-immune cells interactions appeared the strongest in mild patients. INTERPRETATION: SARS-CoV-2 could infect lung epithelium, alter communication patterns between lung epithelial cells and immune system, and drive dysregulated host immune response in COVID-19 patients. FUNDING: This project was supported by National Key R&D Program of China (No. 2018YFC1315000/2018YFC1315004), Science and Technology Program Grant Shenzhen (JCYJ20170413161534162), HMRF Hong Kong (17160862), RGC-CRF Hong Kong (C4039-19G), RGC-GRF Hong Kong (14163817), Vice-Chancellor's Discretionary Fund CUHK and CUHK direct grant, Shenzhen Virtual University Park Support Scheme to CUHK Shenzhen Research Institute.


Sujet(s)
COVID-19/immunologie , Cellules épithéliales/immunologie , Inflammation/immunologie , Poumon/immunologie , SARS-CoV-2/immunologie , Transduction du signal/immunologie , Cellules A549 , Angiotensin-converting enzyme 2/immunologie , COVID-19/virologie , Études cas-témoins , Lignée cellulaire , Lignée cellulaire tumorale , Cytokines/immunologie , Humains , Inflammation/virologie , Cellules tueuses naturelles/immunologie , Poumon/virologie , Macrophages/immunologie , Granulocytes neutrophiles/immunologie , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/virologie , Serine endopeptidases/immunologie , Lymphocytes T/immunologie
19.
Vet Microbiol ; 257: 109058, 2021 Jun.
Article de Anglais | MEDLINE | ID: mdl-33862332

RÉSUMÉ

Actinobacillus pleuropneumoniae (A.pleuropneumoniae) causes serious economic loss for the swine industry. A high-temperature requirements A (HtrA)-like protease and its homologs have been reported to be involved in protein quality control and expression of important immunoprotective antigens in many pathogens. In this study, we showed that HtrA of A.pleuropneumoniae exhibited both chaperone and proteolytic activities. Moreover, Outer membrane protein P5 (OmpP5) in A.pleuropneumoniae and Heat shock protein 90 (Hsp90) in porcine lung tissues were first discovered and identified as specific proteolytic substrates for rHtrA. The maximum cleavage activity occurs at 50 ℃ in a time-dependent manner. In addition, rHtrA mainly induced IgG 2a subtype of IgG and Th1 (IFN-γ, IL-2) response in a mice model, and promoted a significant proliferation of spleen lymphocytes compare with negative control (P < 0.05). The survival rates of 37.5 % were observed against A.pleuropneumoniae strain. Together, these data demonstrate that rHtrA plays a multi-functional role in A.pleuropneumoniae.


Sujet(s)
Actinobacillus pleuropneumoniae/génétique , Protéines de la membrane externe bactérienne/génétique , Protéines de la membrane externe bactérienne/métabolisme , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Actinobacillus pleuropneumoniae/composition chimique , Animaux , Protéines de la membrane externe bactérienne/immunologie , Modèles animaux de maladie humaine , Femelle , Protéines du choc thermique HSP90/métabolisme , Immunoglobuline G/immunologie , Souris de lignée BALB C , Protéolyse , Serine endopeptidases/immunologie , Suidae , Maladies des porcs/immunologie , Maladies des porcs/microbiologie , Lymphocytes auxiliaires Th1/immunologie
20.
Infect Genet Evol ; 91: 104832, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33812037

RÉSUMÉ

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).


Sujet(s)
Angiotensin-converting enzyme 2/génétique , COVID-19/génétique , Génome viral , microARN/génétique , SARS-CoV-2/génétique , Serine endopeptidases/génétique , Glycoprotéine de spicule des coronavirus/génétique , Angiotensin-converting enzyme 2/immunologie , COVID-19/immunologie , COVID-19/anatomopathologie , Régulation de l'expression des gènes , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Humains , Immunité innée , microARN/classification , microARN/immunologie , Mutation , Liaison aux protéines , Récepteurs viraux/génétique , Récepteurs viraux/immunologie , SARS-CoV-2/immunologie , Serine endopeptidases/immunologie , Indice de gravité de la maladie , Glycoprotéine de spicule des coronavirus/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...