Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
1.
Curr Med Chem ; 31(13): 1634-1645, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38666504

RESUMEN

Immune checkpoint inhibitors (ICIs) have shown unprecedented efficacy in treating many advanced cancers. Although FDA-approved ICIs have shown promising efficacy in treating many advanced cancers, their application is greatly limited by the low response rate, immune-related adverse events (irAE), and drug resistance. Developing novel ICIs holds great promise to improve the survival and prognosis of advanced cancer patients. T-Cell immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed on T cells, natural killer (NK) cells, and T regulatory cells. Increasing reports have shown that the disrupting CD155-TIGIT axis could activate the immune system and restore antitumor immune response. This review briefly summarized the role of TIGIT in tumor immune escape and targeting CD155-TIGIT axis drugs in preclinical and clinical trials for cancer immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias , Receptores Inmunológicos , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/inmunología , Receptores Virales/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales
2.
J Virol ; 97(10): e0069623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37796129

RESUMEN

IMPORTANCE: Human cytomegalovirus (HCMV) infection is the leading cause of non-heritable birth defects worldwide. HCMV readily infects the early progenitor cell population of the developing brain, and we have found that infection leads to significantly downregulated expression of key neurodevelopmental transcripts. Currently, there are no approved therapies to prevent or mitigate the effects of congenital HCMV infection. Therefore, we used human-induced pluripotent stem cell-derived organoids and neural progenitor cells to elucidate the glycoproteins and receptors used in the viral entry process and whether antibody neutralization was sufficient to block viral entry and prevent disruption of neurodevelopmental gene expression. We found that blocking viral entry alone was insufficient to maintain the expression of key neurodevelopmental genes, but neutralization combined with neurotrophic factor treatment provided robust protection. Together, these studies offer novel insight into mechanisms of HCMV infection in neural tissues, which may aid future therapeutic development.


Asunto(s)
Anticuerpos Neutralizantes , Infecciones por Citomegalovirus , Citomegalovirus , Expresión Génica , Factores de Crecimiento Nervioso , Humanos , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Citomegalovirus/efectos de los fármacos , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Células Madre Pluripotentes Inducidas/citología , Factores de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/uso terapéutico , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/virología , Organoides/citología , Organoides/metabolismo , Organoides/virología , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
5.
Cell Mol Biol Lett ; 27(1): 10, 2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35109786

RESUMEN

The novel coronavirus disease 2019 (COVID-19) pandemic has spread worldwide, and finding a safe therapeutic strategy and effective vaccine is critical to overcoming severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, elucidation of pathogenesis mechanisms, especially entry routes of SARS-CoV-2 may help propose antiviral drugs and novel vaccines. Several receptors have been demonstrated for the interaction of spike (S) protein of SARS-CoV-2 with host cells, including angiotensin-converting enzyme (ACE2), ephrin ligands and Eph receptors, neuropilin 1 (NRP-1), P2X7, and CD147. The expression of these entry receptors in the central nervous system (CNS) may make the CNS prone to SARS-CoV-2 invasion, leading to neurodegenerative diseases. The present review provides potential pathological mechanisms of SARS-CoV-2 infection in the CNS, including entry receptors and cytokines involved in neuroinflammatory conditions. Moreover, it explains several neurodegenerative disorders associated with COVID-19. Finally, we suggest inflammasome and JaK inhibitors as potential therapeutic strategies for neurodegenerative diseases.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Sistema Nervioso Central/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Receptores Virales/genética , SARS-CoV-2/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/uso terapéutico , Basigina/genética , Basigina/metabolismo , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/virología , Efrinas/genética , Efrinas/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Factores Inmunológicos/uso terapéutico , Inflamasomas/genética , Inflamasomas/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/virología , Neuropilina-1/genética , Neuropilina-1/metabolismo , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35074895

RESUMEN

The development of small-molecules targeting different components of SARS-CoV-2 is a key strategy to complement antibody-based treatments and vaccination campaigns in managing the COVID-19 pandemic. Here, we show that two thiol-based chemical probes that act as reducing agents, P2119 and P2165, inhibit infection by human coronaviruses, including SARS-CoV-2, and decrease the binding of spike glycoprotein to its receptor, the angiotensin-converting enzyme 2 (ACE2). Proteomics and reactive cysteine profiling link the antiviral activity to the reduction of key disulfides, specifically by disruption of the Cys379-Cys432 and Cys391-Cys525 pairs distal to the receptor binding motif in the receptor binding domain (RBD) of the spike glycoprotein. Computational analyses provide insight into conformation changes that occur when these disulfides break or form, consistent with an allosteric role, and indicate that P2119/P2165 target a conserved hydrophobic binding pocket in the RBD with the benzyl thiol-reducing moiety pointed directly toward Cys432. These collective findings establish the vulnerability of human coronaviruses to thiol-based chemical probes and lay the groundwork for developing compounds of this class, as a strategy to inhibit the SARS-CoV-2 infection by shifting the spike glycoprotein redox scaffold.


Asunto(s)
Amino Alcoholes/farmacología , Enzima Convertidora de Angiotensina 2/química , Antivirales/farmacología , Éteres Fenílicos/farmacología , Receptores Virales/química , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/química , Compuestos de Sulfhidrilo/farmacología , Regulación Alostérica , Amino Alcoholes/química , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Sitios de Unión , COVID-19/virología , Línea Celular , Disulfuros/antagonistas & inhibidores , Disulfuros/química , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/virología , Oxidación-Reducción , Éteres Fenílicos/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Receptores Virales/antagonistas & inhibidores , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Compuestos de Sulfhidrilo/química , Tratamiento Farmacológico de COVID-19
7.
J Biomol Struct Dyn ; 40(20): 10023-10032, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34229582

RESUMEN

The novel coronavirus SARS-CoV-2, responsible for the present COVID-19 global pandemic, is known to bind to the angiotensin converting enzyme-2 (ACE2) receptor in human cells. A possible treatment of COVID-19 could involve blocking ACE2 and/or disabling the spike protein on the virus. Here, molecular dynamics simulations were performed to test the binding affinities of nine candidate compounds. Of these, three drugs showed significant therapeutic potential that warrant further investigation: SN35563, a ketamine ester analogue, was found to bind strongly to the ACE2 receptor but weakly within the spike receptor-binding domain (RBD); in contrast, arbidol and hydroxychloroquine bound preferentially with the spike RBD rather than ACE2. A fourth drug, remdesivir, bound approximately equally to both the ACE2 and viral spike RBD, thus potentially increasing risk of viral infection by bringing the spike protein into closer proximity to the ACE2 receptor. We suggest more experimental investigations to test that SN35563-in combination with arbidol or hydroxychloroquine-might act synergistically to block viral cell entry by providing therapeutic blockade of the host ACE2 simultaneous with reduction of viral spike receptor-binding; and that this combination therapy would allow the use of smaller doses of each drug.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Antivirales , Receptores Virales , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/química , Sitios de Unión , COVID-19 , Hidroxicloroquina/farmacología , Simulación de Dinámica Molecular , Unión Proteica , Receptores Virales/antagonistas & inhibidores , Receptores Virales/química , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/química , Antivirales/farmacología
8.
J Sep Sci ; 45(2): 456-467, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34729910

RESUMEN

Chloroquine and hydroxychloroquine have been studied since the early clinical treatment of SARS-CoV-2 outbreak. Considering these two chiral drugs are currently in use as the racemate, high-expression angiotensin-converting enzyme 2 cell membrane chromatography was established for investigating the differences of two paired enantiomers binding to angiotensin-converting enzyme 2 receptor. Molecular docking assay and detection of SARS-CoV-2 spike pseudotyped virus entry into angiotensin-converting enzyme 2-HEK293T cells were also conducted for further investigation. Results showed that each single enantiomer could bind well to angiotensin-converting enzyme 2, but there were differences between the paired enantiomers and corresponding racemate in frontal analysis. R-Chloroquine showed better angiotensin-converting enzyme 2 receptor binding ability compared to S-chloroquine/chloroquine (racemate). S-Hydroxychloroquine showed better angiotensin-converting enzyme 2 receptor binding ability than R-hydroxychloroquine/hydroxychloroquine. Moreover, each single enantiomer was proved effective compared with the control group; compared with S-chloroquine or the racemate, R-chloroquine showed better inhibitory effects at the same concentration. As for hydroxychloroquine, R-hydroxychloroquine showed better inhibitory effects than S-hydroxychloroquine, but it slightly worse than the racemate. In conclusion, R-chloroquine showed better angiotensin-converting enzyme 2 receptor binding ability and inhibitory effects compared to S-chloroquine/chloroquine (racemate). S-Hydroxychloroquine showed better angiotensin-converting enzyme 2 receptor binding ability than R-hydroxychloroquine/hydroxychloroquine (racemate), while the effect of preventing SARS-CoV-2 pseudovirus from entering cells was weaker than R-hydroxychloroquine/hydroxychloroquine (racemate).


Asunto(s)
Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , Cloroquina/química , Cloroquina/farmacología , Cromatografía Líquida de Alta Presión/métodos , Hidroxicloroquina/química , Hidroxicloroquina/farmacología , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Antivirales/química , Antivirales/farmacología , COVID-19/virología , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/virología , Células HEK293 , Humanos , Técnicas In Vitro , Simulación del Acoplamiento Molecular , Receptores Virales/antagonistas & inhibidores , Receptores Virales/química , Receptores Virales/efectos de los fármacos , SARS-CoV-2/química , SARS-CoV-2/efectos de los fármacos , Solventes , Estereoisomerismo , Pseudotipado Viral , Internalización del Virus , Tratamiento Farmacológico de COVID-19
9.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34902435

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , COVID-19/prevención & control , Extractos Celulares/administración & dosificación , Receptores Virales/antagonistas & inhibidores , Receptores Virales/inmunología , SARS-CoV-2/inmunología , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Animales , COVID-19/inmunología , COVID-19/virología , Células CACO-2 , Extractos Celulares/inmunología , Células Cultivadas , Chlorocebus aethiops , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Células HEK293 , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/inmunología , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/inmunología , Células Vero
10.
Pharmacol Res ; 175: 105982, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34798263

RESUMEN

All the different coronavirus SARS-CoV-2 variants isolated so far share the same mechanism of infection mediated by the interaction of their spike (S) glycoprotein with specific residues on their cellular receptor: the angiotensin converting enzyme 2 (ACE2). Therefore, the steric hindrance on this cellular receptor created by a bulk macromolecule may represent an effective strategy for the prevention of the viral spreading and the onset of severe forms of Corona Virus disease 19 (COVID-19). Here, we applied a systematic evolution of ligands by exponential enrichment (SELEX) procedure to identify two single strand DNA molecules (aptamers) binding specifically to the region surrounding the K353, the key residue in human ACE2 interacting with the N501 amino acid of the SARS-CoV-2 S. 3D docking in silico experiments and biochemical assays demonstrated that these aptamers bind to this region, efficiently prevent the SARS-CoV-2 S/human ACE2 interaction and the viral infection in the nanomolar range, regardless of the viral variant, thus suggesting the possible clinical development of these aptamers as SARS-CoV-2 infection inhibitors. Our approach brings a significant innovation to the therapeutic paradigm of the SARS-CoV-2 pandemic by protecting the target cell instead of focusing on the virus; this is particularly attractive in light of the increasing number of viral mutants that may potentially escape the currently developed immune-mediated neutralization strategies.


Asunto(s)
Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Aptámeros de Nucleótidos/farmacología , Tratamiento Farmacológico de COVID-19 , Receptores Virales/antagonistas & inhibidores , SARS-CoV-2/patogenicidad , Internalización del Virus/efectos de los fármacos , Células A549 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/metabolismo , COVID-19/enzimología , COVID-19/genética , COVID-19/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Mutación , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/genética , Técnica SELEX de Producción de Aptámeros
11.
Cell Rep ; 36(9): 109628, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469726

RESUMEN

Hendra virus and Nipah virus (NiV), members of the Henipavirus (HNV) genus, are zoonotic paramyxoviruses known to cause severe disease across six mammalian orders, including humans. We isolated a panel of human monoclonal antibodies (mAbs) from the B cells of an individual with prior exposure to equine Hendra virus (HeV) vaccine, targeting distinct antigenic sites. The most potent class of cross-reactive antibodies achieves neutralization by blocking viral attachment to the host cell receptors ephrin-B2 and ephrin-B3, with a second class being enhanced by receptor binding. mAbs from both classes display synergistic activity in vitro. In a stringent hamster model of NiV Bangladesh (NiVB) infection, antibodies from both classes reduce morbidity and mortality and achieve synergistic protection in combination. These candidate mAbs might be suitable for use in a cocktail therapeutic approach to achieve synergistic potency and reduce the risk of virus escape.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Antivirales/farmacología , Efrina-B2/antagonistas & inhibidores , Efrina-B3/antagonistas & inhibidores , Infecciones por Henipavirus/prevención & control , Henipavirus/patogenicidad , Receptores Virales/antagonistas & inhibidores , Animales , Especificidad de Anticuerpos , Chlorocebus aethiops , Reacciones Cruzadas , Modelos Animales de Enfermedad , Quimioterapia Combinada , Efrina-B2/inmunología , Efrina-B2/metabolismo , Efrina-B3/inmunología , Efrina-B3/metabolismo , Femenino , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/metabolismo , Infecciones por Henipavirus/virología , Interacciones Huésped-Patógeno , Humanos , Mesocricetus , Receptores Virales/inmunología , Receptores Virales/metabolismo , Células Vero
12.
Cells ; 10(7)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34359995

RESUMEN

Chikungunya virus (CHIKV) is a re-emerging, mosquito-transmitted, enveloped positive stranded RNA virus. Chikungunya fever is characterized by acute and chronic debilitating arthritis. Although multiple host factors have been shown to enhance CHIKV infection, the molecular mechanisms of cell entry and entry factors remain poorly understood. The phosphatidylserine-dependent receptors, T-cell immunoglobulin and mucin domain 1 (TIM-1) and Axl receptor tyrosine kinase (Axl), are transmembrane proteins that can serve as entry factors for enveloped viruses. Previous studies used pseudoviruses to delineate the role of TIM-1 and Axl in CHIKV entry. Conversely, here, we use the authentic CHIKV and cells ectopically expressing TIM-1 or Axl and demonstrate a role for TIM-1 in CHIKV infection. To further characterize TIM-1-dependent CHIKV infection, we generated cells expressing domain mutants of TIM-1. We show that point mutations in the phosphatidylserine binding site of TIM-1 lead to reduced cell binding, entry, and infection of CHIKV. Ectopic expression of TIM-1 renders immortalized keratinocytes permissive to CHIKV, whereas silencing of endogenously expressed TIM-1 in human hepatoma cells reduces CHIKV infection. Altogether, our findings indicate that, unlike Axl, TIM-1 readily promotes the productive entry of authentic CHIKV into target cells.


Asunto(s)
Virus Chikungunya/genética , Receptor Celular 1 del Virus de la Hepatitis A/genética , Interacciones Huésped-Patógeno/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Virales/genética , Internalización del Virus , Animales , Anticuerpos Monoclonales/farmacología , Células CHO , Línea Celular , Línea Celular Tumoral , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/inmunología , Chlorocebus aethiops , Cricetulus , Endosomas/efectos de los fármacos , Endosomas/inmunología , Endosomas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/virología , Fibroblastos/inmunología , Fibroblastos/virología , Expresión Génica , Células HEK293 , Receptor Celular 1 del Virus de la Hepatitis A/antagonistas & inhibidores , Receptor Celular 1 del Virus de la Hepatitis A/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Queratinocitos/inmunología , Queratinocitos/virología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Virales/antagonistas & inhibidores , Receptores Virales/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transgenes , Células Vero , Internalización del Virus/efectos de los fármacos , Tirosina Quinasa del Receptor Axl
13.
Oral Oncol ; 121: 105472, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34333450

RESUMEN

OBJECTIVES: Anti-PD-1/PD-L1 therapy has recently been approved for head and neck squamous cell carcinoma (HNSCC). However, given that large numbers of patients with HNSCC do not respond to PD-1/PD-L1 antibodies, combination strategies for elevating the response rate need to be further investigated. The goal of this study was to explore the possibility of dual-targeting CD155/TIGIT and PD-1/PD-L1 signalling in HNSCC. MATERIALS AND METHODS: Multiplex flow cytometry was performed to determine the co-expression of CD155 and PD-L1 in human HNSCC and transgenic HNSCC mouse models. The combined application of TIGIT mAb and PD-L1 mAb in a mouse model was used to explore the therapeutic effect. RESULTS: CD155 and PD-L1 were highly co-expressed on myeloid-derived suppressor cells (MDSCs) derived from patients with HNSCC and were inversely associated with the percentage of tumour CD3+ T and effector memory T cells. CD155+PD-L1+ MDSCs in the mouse model were gradually enriched in the tumour microenvironment in the middle and late stages of tumour progression. Anti-PD-L1 treatment alone upregulated the expression of CD155 on MDSCs and while anti-TIGIT treatment upregulated the expression of PD-L1 on MDSCs in mice. The combined blockade of TIGIT/CD155 and PD-1/PD-L1 signalling in mice significantly inhibited tumour growth, enhanced the percentages of effector T cells and cytokine secretion and elicited immune memory effects. CONCLUSION: Our study indicated that CD155+PD-L1+ MDSCs are enriched in the tumour microenvironment and blocking TIGIT/CD155 can effectively enhance the response rate of HNSCC to PD-L1 mAb therapy, which provides the clinical potential of co-targeting TIGIT/CD155 and PD-1/PD-L1 signalling.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello , Células Supresoras de Origen Mieloide , Carcinoma de Células Escamosas de Cabeza y Cuello , Animales , Anticuerpos Monoclonales/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Células T de Memoria , Ratones , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Microambiente Tumoral
14.
Biomolecules ; 11(5)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068552

RESUMEN

Strategies boosting both innate and adaptive immunity have great application prospects in cancer immunotherapy. Antibodies dual blocking the innate checkpoint CD47 and adaptive checkpoint PD-L1 or TIGIT could achieve durable anti-tumor effects. However, a small molecule dual blockade of CD47/SIRPα and TIGIT/PVR pathways has not been investigated. Here, an elevated expression of CD47 and PVR was observed in tumor tissues and cell lines analyzed with the GEO datasets and by flow cytometry, respectively. Compounds approved by the FDA were screened with the software MOE by docking to the potential binding pockets of SIRPα and PVR identified with the corresponding structural analysis. The candidate compounds were screened by blocking and MST binding assays. Azelnidipine was found to dual block CD47/SIRPα and TIGIT/PVR pathways by co-targeting SIRPα and PVR. In vitro, azelnidipine could enhance the macrophage phagocytosis when co-cultured with tumor cells. In vivo, azelnidipine alone or combined with irradiation could significantly inhibit the growth of MC38 tumors. Azelnidipine also significantly inhibits the growth of CT26 tumors, by enhancing the infiltration and function of CD8+ T cell in tumor and systematic immune response in the tumor-draining lymph node and spleen in a CD8+ T cell dependent manner. Our research suggests that the anti-hypertensive drug azelnidipine could be repositioned for cancer immunotherapy.


Asunto(s)
Ácido Azetidinocarboxílico/análogos & derivados , Dihidropiridinas/farmacología , Reposicionamiento de Medicamentos/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Ácido Azetidinocarboxílico/farmacología , Antígeno CD47/antagonistas & inhibidores , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular Tumoral , Cricetinae , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos
15.
Nature ; 595(7869): 718-723, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34082438

RESUMEN

Resistance represents a major challenge for antibody-based therapy for COVID-191-4. Here we engineered an immunoglobulin M (IgM) neutralizing antibody (IgM-14) to overcome the resistance encountered by immunoglobulin G (IgG)-based therapeutics. IgM-14 is over 230-fold more potent than its parental IgG-14 in neutralizing SARS-CoV-2. IgM-14 potently neutralizes the resistant virus raised by its corresponding IgG-14, three variants of concern-B.1.1.7 (Alpha, which first emerged in the UK), P.1 (Gamma, which first emerged in Brazil) and B.1.351 (Beta, which first emerged in South Africa)-and 21 other receptor-binding domain mutants, many of which are resistant to the IgG antibodies that have been authorized for emergency use. Although engineering IgG into IgM enhances antibody potency in general, selection of an optimal epitope is critical for identifying the most effective IgM that can overcome resistance. In mice, a single intranasal dose of IgM-14 at 0.044 mg per kg body weight confers prophylactic efficacy and a single dose at 0.4 mg per kg confers therapeutic efficacy against SARS-CoV-2. IgM-14, but not IgG-14, also confers potent therapeutic protection against the P.1 and B.1.351 variants. IgM-14 exhibits desirable pharmacokinetics and safety profiles when administered intranasally in rodents. Our results show that intranasal administration of an engineered IgM can improve efficacy, reduce resistance and simplify the prophylactic and therapeutic treatment of COVID-19.


Asunto(s)
COVID-19/prevención & control , COVID-19/virología , Inmunoglobulina M/administración & dosificación , Inmunoglobulina M/inmunología , SARS-CoV-2/clasificación , SARS-CoV-2/inmunología , Administración Intranasal , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/efectos adversos , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/inmunología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , COVID-19/inmunología , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/efectos adversos , Inmunoglobulina M/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ingeniería de Proteínas , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , SARS-CoV-2/genética , Tratamiento Farmacológico de COVID-19
16.
J Mol Model ; 27(7): 206, 2021 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-34169390

RESUMEN

The interaction between SARS-CoV-2 Spike protein and angiotensin-converting enzyme 2 (ACE2) is essential to viral attachment and the subsequent fusion process. Interfering with this event represents an attractive avenue for the development of therapeutics and vaccine development. Here, a hybrid approach of ligand- and structure-based virtual screening techniques were employed to disclose similar analogues of a reported antiviral phytochemical, glycyrrhizin, targeting the blockade of ACE2 interaction with the SARS-CoV-2 Spike. A ligand-based similarity search using a stringent cut-off revealed 40 FDA-approved compounds in DrugBank. These filtered hits were screened against ACE2 using a blind docking approach to determine the natural binding tendency of the compounds with ACE2. Three compounds, deslanoside, digitoxin, and digoxin, were reported to show strong binding with ACE2. These compounds bind at the H1-H2 binding pocket, in a manner similar to that of glycyrrhizin which was used as a control. To achieve consistency in the docking results, docking calculations were performed via two sets of docking software that predicted binding energy as ACE2-Deslanoside (AutoDock, -10.3 kcal/mol and DockThor, -9.53 kcal/mol), ACE2-Digitoxin (AutoDock, -10.6 kcal/mol and DockThor, -8.84 kcal/mol), and ACE2-Digoxin (AutoDock, -10.6 kcal/mol and DockThor, -8.81 kcal/mol). The docking results were validated by running molecular simulations in aqueous solution that demonstrated the stability of ACE2 with no major conformational changes in the ligand original binding mode (~ 2 Å average RMSD). Binding interactions remained quite stable with an increased potential for getting stronger as the simulation proceeded. MMGB/PBSA binding free energies were also estimated and these supported the high stability of the complexes compared to the control (~ -50 kcal/mol net MMGB/PBSA binding energy versus ~ -30 kcal/mol). Collectively, the data demonstrated that the compounds shortlisted in this study might be subjected to experimental evaluation to uncover their real blockade capacity of SARS-CoV-2 host ACE2 receptor.


Asunto(s)
Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Ácido Glicirrínico/farmacología , Receptores Virales/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/química , Animales , Antivirales/química , Sitios de Unión , COVID-19/enzimología , COVID-19/virología , Descubrimiento de Drogas , Reposicionamiento de Medicamentos , Ácido Glicirrínico/análogos & derivados , Ácido Glicirrínico/química , Interacciones Huésped-Patógeno , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Receptores Virales/química , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Relación Estructura-Actividad
17.
Biomaterials ; 275: 120988, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34186238

RESUMEN

PD-L1/PD-1 blockade therapy shows durable responses to triple-negative breast cancer (TNBC), but the response rate is low. CD155 promotes tumor metastasis intrinsically and modulates the immune response extrinsically as the ligand of DNAM-1 (costimulatory receptor) and TIGIT/CD96 (coinhibitory receptors). Herein, we verified that TNBC cells coexpressed PD-L1 and CD155. By examining the receptors of PD-L1 and CD155 on TNBC tumor-infiltrating lymphocytes (TILs) over time, we observed that PD-1 and DNAM-1 were upregulated early, whereas CD96 and TIGIT were upregulated late in CD8+ TILs. Based on these findings, we developed CD155 siRNA (siCD155)-loaded mPEG-PLGA-PLL (PEAL) nanoparticles (NPs) coated with PD-L1 blocking antibodies (P/PEALsiCD155) to asynchronously block PD-L1 and CD155 in a spatiotemporal manner. P/PEALsiCD155 maximized early-stage CD8+ T cell immune surveillance against 4T1 tumor, whereas reversed inhibition status of the late stage CD8+ T cells to prevent 4T1 tumor immune escape. In addition, the combination of P/PEALsiCD155 and tumor-specific CD8 T cells induced immunogenic cell death (ICD) of 4T1 cells to further boost immune checkpoint therapy. Most importantly, P/PEALsiCD155 displayed excellent TNBC targeting and induced CD8+ TILs-dominant intratumor antitumor immunity to efficiently inhibit TNBC progression and metastasis with excellent safety in a syngeneic 4T1 orthotopic TNBC tumor model.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Nanopartículas , Receptores Virales/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas , Linfocitos T CD8-positivos , Humanos , Linfocitos Infiltrantes de Tumor , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
18.
J Virol ; 95(16): e0001021, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34037420

RESUMEN

Respiratory syncytial virus (RSV) has been reported to use CX3CR1 in vitro as a receptor on cultured primary human airway epithelial cultures. To evaluate CX3CR1 as the receptor for RSV in vivo, we used the cotton rat animal model because of its high permissiveness for RSV infection. Sequencing the cotton rat CX3CR1 gene revealed 91% amino acid similarity to human CX3CR1. Previous work found that RSV binds to CX3CR1 via its attachment glycoprotein (G protein) to infect primary human airway cultures. To determine whether CX3CR1-G protein interaction is necessary for RSV infection, recombinant RSVs containing mutations in the CX3CR1 binding site of the G protein were tested in cotton rats. In contrast to wild-type virus, viral mutants did not grow in the lungs of cotton rats. When RSV was incubated with an antibody blocking the CX3CR1 binding site of G protein and subsequently inoculated intranasally into cotton rats, no virus was found in the lungs 4 days postinfection. In contrast, growth of RSV was not affected after preincubation with heparan sulfate (the receptor for RSV on immortalized cell lines). A reduction in CX3CR1 expression in the cotton rat lung through the use of peptide-conjugated morpholino oligomers led to a 10-fold reduction in RSV titers at day 4 postinfection. In summary, these results indicate that CX3CR1 functions as a receptor for RSV in cotton rats and, in combination with data from human airway epithelial cell cultures, strongly suggest that CX3CR1 is a primary receptor for naturally acquired RSV infection. IMPORTANCE The knowledge about a virus receptor is useful to better understand the uptake of a virus into a cell and potentially develop antivirals directed against either the receptor molecule on the cell or the receptor-binding protein of the virus. Among a number of potential receptor proteins, human CX3CR1 has been demonstrated to act as a receptor for respiratory syncytial virus (RSV) on human epithelial cells in tissue culture. Here, we report that the cotton rat CX3CR1, which is similar to the human molecule, acts as a receptor in vivo. This study strengthens the argument that CX3CR1 is a receptor molecule for RSV.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Receptores Virales/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Anticuerpos Antivirales/farmacología , Sitios de Unión , Receptor 1 de Quimiocinas CX3C/antagonistas & inhibidores , Receptor 1 de Quimiocinas CX3C/química , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/virología , Heparitina Sulfato/metabolismo , Humanos , Mutación , Receptores Virales/antagonistas & inhibidores , Receptores Virales/química , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/crecimiento & desarrollo , Virus Sincitial Respiratorio Humano/metabolismo , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Sigmodontinae , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/genética
19.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33883223

RESUMEN

Long disregarded as junk DNA or genomic dark matter, endogenous retroviruses (ERVs) have turned out to represent important components of the antiviral immune response. These remnants of once-infectious retroviruses not only regulate cellular immune activation, but may even directly target invading viral pathogens. In this Gem, we summarize mechanisms by which retroviral fossils protect us from viral infections. One focus will be on recent advances in the role of ERVs as regulators of antiviral gene expression.


Asunto(s)
Retrovirus Endógenos/fisiología , Retroelementos , Virosis/inmunología , Animales , Retrovirus Endógenos/genética , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Humanos , Inmunidad Celular , Regiones Promotoras Genéticas , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Virión/metabolismo , Virosis/genética , Virosis/virología
20.
Rev Med Virol ; 31(6): e2227, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33763936

RESUMEN

Severe acute respiratory syndrome related coronavirus-2 (SARS-CoV-2) is the cause of Covid-19 which was classified as a global pandemic in March 2020. The increasing global health and economic burden of SARS-CoV-2 has necessitated urgent investigations into the pathogenesis of disease and development of therapeutic and vaccination regimens. Human trials of vaccine and antiviral candidates have been undertaken, but basic pathogenetic studies are still required to inform these trials. Gaps in understanding of cellular infection by, and immunity to, SARS-CoV-2 mean additional models are required to assist in improved design of these therapeutics. Human organoids are three-dimensional models that contain multiple cell types and mimic human organs in ex vivo culture conditions. The SARS-CoV-2 virus has been implicated in causing not only respiratory injury but also injury to other organs such as the brain, liver and kidneys. Consequently, a variety of different organoid models have been employed to investigate the pathogenic mechanisms of disease due to SARS-CoV-2. Data on these models have not been systematically assembled. In this review, we highlight key findings from studies that have utilised different human organoid types to investigate the expression of SARS-CoV-2 receptors, permissiveness, immune response, dysregulation of cellular functions, and potential antiviral therapeutics.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Modelos Biológicos , Organoides/inmunología , Receptores Virales/antagonistas & inhibidores , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Antivirales/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/virología , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Técnicas de Cultivo de Célula , Colon/efectos de los fármacos , Colon/inmunología , Colon/virología , Citocinas/genética , Citocinas/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/virología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Organoides/efectos de los fármacos , Organoides/virología , Receptores Virales/genética , Receptores Virales/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Serina Endopeptidasas/genética , Serina Endopeptidasas/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...