Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(32): e2200879119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35925889

RESUMEN

The value of anti-CTLA-4 antibodies in cancer therapy is well established. However, the broad application of currently available anti-CTLA-4 therapeutic antibodies is hampered by their narrow therapeutic index. It is therefore challenging and attractive to develop the next generation of anti-CTLA-4 therapeutics with improved safety and efficacy. To this end, we generated fully human heavy chain-only antibodies (HCAbs) against CTLA-4. The hIgG1 Fc domain of the top candidate, HCAb 4003-1, was further engineered to enhance its regulatory T (Treg) cell depletion effect and to decrease its half-life, resulting in HCAb 4003-2. We tested these HCAbs in in vitro and in vivo experiments in comparison with ipilimumab and other anti-CTLA4 antibodies. The results show that human HCAb 4003-2 binds human CTLA-4 with high affinity and potently blocks the binding of B7-1 (CD80) and B7-2 (CD86) to CTLA-4. The results also show efficient tumor penetration. HCAb 4003-2 exhibits enhanced antibody-dependent cellular cytotoxicity function, lower serum exposure, and more potent anti-tumor activity than ipilimumab in murine tumor models, which is partly driven by a substantial depletion of intratumoral Tregs. Importantly, the enhanced efficacy combined with the shorter serum half-life and less systemic drug exposure in vivo potentially provides an improved therapeutic window in cynomolgus monkeys and preliminary clinical applications. With its augmented efficacy via Treg depletion and improved safety profile, HCAb 4003-2 is a promising candidate for the development of next generation anti-CTLA-4 therapy.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina , Inmunoterapia , Neoplasias , Linfocitos T Reguladores , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Antígeno CTLA-4/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/farmacología , Ipilimumab/farmacología , Ratones , Neoplasias/patología , Neoplasias/terapia
2.
Cell Mol Life Sci ; 79(2): 82, 2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-35048158

RESUMEN

Senescence, the irreversible cell cycle arrest of damaged cells, is accompanied by a deleterious pro-inflammatory senescence-associated secretory phenotype (SASP). Senescence and the SASP are major factors in aging, cancer, and degenerative diseases, and interfere with the expansion of adult cells in vitro, yet little is known about how to counteract their induction and deleterious effects. Paracrine signals are increasingly recognized as important senescence triggers and understanding their regulation and mode of action may provide novel opportunities to reduce senescence-induced inflammation and improve cell-based therapies. Here, we show that the signalling protein WNT3A counteracts the induction of paracrine senescence in cultured human adult mesenchymal stem cells (MSCs). We find that entry into senescence in a small subpopulation of MSCs triggers a secretome that causes a feed-forward signalling cascade that with increasing speed induces healthy cells into senescence. WNT signals interrupt this cascade by repressing cytokines that mediate this induction of senescence. Inhibition of those mediators by interference with NF-κB or interleukin 6 signalling reduced paracrine senescence in absence of WNT3A and promoted the expansion of MSCs. Our work reveals how WNT signals can antagonize senescence and has relevance not only for expansion of adult cells but can also provide new insights into senescence-associated inflammatory and degenerative diseases.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Fenotipo Secretor Asociado a la Senescencia , Vía de Señalización Wnt , Proliferación Celular , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Proteína Wnt3A/metabolismo
3.
J Infect Dis ; 217(2): 298-309, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29099932

RESUMEN

Background: Carriage of Mycoplasma pneumoniae (Mp) in the nasopharynx is considered a prerequisite for pulmonary infection. It is interesting to note that Mp carriage is also detected after infection. Although B cells are known to be involved in pulmonary Mp clearance, their role in Mp carriage is unknown. Methods: In this study, we show in a mouse model that Mp persists in the nose after pulmonary infection, similar to humans. Results: Infection of mice enhanced Mp-specific immunoglobulin (Ig) M and IgG levels in serum and bronchoalveolar lavage fluid. However, nasal washes only contained elevated Mp-specific IgA. These differences in Ig compartmentalization correlated with differences in Mp-specific B cell responses between nose- and lung-draining lymphoid tissues. Moreover, transferred Mp-specific serum Igs had no effect on nasal carriage in B cell-deficient µMT mice, whereas this enabled µMT mice to clear pulmonary Mp infection. Conclusions: We report the first evidence that humoral immunity is limited in clearing Mp from the upper respiratory tract.


Asunto(s)
Linfocitos B/inmunología , Portador Sano/inmunología , Mycoplasma pneumoniae/inmunología , Nasofaringe/inmunología , Nasofaringe/microbiología , Neumonía por Mycoplasma/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Ratones Endogámicos C57BL , Mucosa Nasal/inmunología
4.
J Mol Cell Cardiol ; 88: 145-54, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26436984

RESUMEN

Nitric oxide (NO) produced by endothelial NO synthase (eNOS) exerts beneficial effects in a variety of cardiovascular disease states. Studies on the benefit of eNOS activity in pressure-overload cardiac hypertrophy and dysfunction produced by aortic stenosis are equivocal, which may be due to different expression levels of eNOS or different severities of pressure-overload. Consequently, we investigated the effects of eNOS-expression level on cardiac hypertrophy and dysfunction produced by mild or severe pressure-overload. To unravel the impact of eNOS on pressure-overload cardiac dysfunction we subjected eNOS deficient, wildtype and eNOS overexpressing transgenic (eNOS-Tg) mice to 8weeks of mild or severe transverse aortic constriction (TAC) and studied cardiac geometry and function at the whole organ and tissue level. In both mild and severe TAC, lack of eNOS ameliorated, whereas eNOS overexpression aggravated, TAC-induced cardiac remodeling and dysfunction. Moreover, the detrimental effects of eNOS in severe TAC were associated with aggravation of TAC-induced NOS-dependent oxidative stress and by further elevation of eNOS monomer levels, consistent with enhanced eNOS uncoupling. In the presence of TAC, scavenging of reactive oxygen species with N-acetylcysteine reduced eNOS S-glutathionylation, eNOS monomer and NOS-dependent superoxide levels in eNOS-Tg mice to wildtype levels. Accordingly, N-acetylcysteine improved cardiac function in eNOS-Tg but not in wildtype mice with TAC. In conclusion, independent of the severity of TAC, eNOS aggravates cardiac remodeling and dysfunction, which appears due to TAC-induced eNOS uncoupling and superoxide production.


Asunto(s)
Cardiomegalia/enzimología , Cardiomegalia/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico/metabolismo , Remodelación Ventricular , Acetilcisteína/farmacología , Animales , Aorta/cirugía , Cardiomegalia/etiología , Cardiomegalia/patología , Constricción Patológica/complicaciones , Constricción Patológica/cirugía , Activación Enzimática , Femenino , Depuradores de Radicales Libres/farmacología , Eliminación de Gen , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Índice de Severidad de la Enfermedad , Superóxidos/antagonistas & inhibidores , Superóxidos/metabolismo
5.
Lab Invest ; 95(10): 1092-104, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26214584

RESUMEN

The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKß) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Suplementos Dietéticos , Endotelio Vascular/metabolismo , Hipoglucemiantes/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Animales , Aorta/citología , Aorta/metabolismo , Aorta/fisiopatología , Arginina/metabolismo , Arginina/uso terapéutico , Bovinos , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Endotelio Vascular/citología , Endotelio Vascular/fisiopatología , Femenino , Heterocigoto , Humanos , Hipoglucemiantes/metabolismo , Insulina/genética , Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Inhibidores de Proteínas Quinasas/metabolismo , Pterinas/metabolismo , Pterinas/uso terapéutico , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Salicilatos/metabolismo , Salicilatos/uso terapéutico , Destete
6.
Clin Lab ; 60(10): 1659-62, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25651711

RESUMEN

BACKGROUND: Phospholipid transfer protein (PLTP) is an emerging cardiometabolic risk factor. Plasma PLTP is elevated in humans with end-stage kidney disease and glomerular proteinuria, but the contribution of systemic PLTP elevation to the development of renal damage is unknown. We tested whether human PLTP expression in ApoE deficient mice (an atherosclerosis-prone model) results in renal insufficiency, albuminuria, or glomerulosclerosis. METHODS: Serum creatinine, albuminuria, as well as kidney and aortic arch histology were determined in 6 male huPLTPtgApoE-/- mice and 8 similarly aged male wild type mice fed a regular chow diet. RESULTS: huPLTPtgApoE-/- mice (2- to 3-fold elevated PLTP activity) showed marked aortic atherosclerosis. However, serum creatinine (p = 0.11) and albuminuria (p = 0.87) were not increased, whereas renal arteriolar atherosclerosis and glomerulosclerosis were not evident in this PLTP transgenic model. CONCLUSIONS: High systemic PLTP expression does not contribute significantly to a renal phenotype despite being implicated in systemic atherosclerosis.


Asunto(s)
Aorta Torácica/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Glomerulonefritis/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteinuria/metabolismo , Insuficiencia Renal/metabolismo , Animales , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Genotipo , Glomerulonefritis/genética , Glomerulonefritis/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteínas de Transferencia de Fosfolípidos/genética , Proteinuria/genética , Proteinuria/patología , Insuficiencia Renal/genética , Insuficiencia Renal/patología , Factores de Riesgo , Índice de Severidad de la Enfermedad , Regulación hacia Arriba
7.
Proc Natl Acad Sci U S A ; 108(39): 16404-9, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21930905

RESUMEN

Panton-Valentine leukocidin (PVL) is a pore-forming toxin associated with current outbreaks of community-associated methicillin-resistant strains and implicated directly in the pathophysiology of Staphylococcus aureus-related diseases. Humanized heavy chain-only antibodies (HCAb) were generated against S. aureus PVL from immunized transgenic mice to neutralize toxin activity. The active form of PVL consists of the two components, LukS-PV and LukF-PV, which induce osmotic lysis following pore formation in host defense cells. One anti-LukS-PV HCAb, three anti-LukF-PV HCAbs with affinities in the nanomolar range, and one engineered tetravalent bispecific HCAb were tested in vitro and in vivo, and all prevented toxin binding and pore formation. Anti-LukS-PV HCAb also binds to γ-hemolysin C (HlgC) and inhibits HlgC/HlgB pore formation. Experiments in vivo in a toxin-induced rabbit endophthalmitis model showed that these HCAbs inhibit inflammatory reactions and tissue destruction, with the tetravalent bispecific HCAb performing best. Our findings show the therapeutic potential of HCAbs, and in particular, bispecific antibodies.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Neutralizantes/inmunología , Exotoxinas/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Neutralizantes/química , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Ensayo de Inmunoadsorción Enzimática , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Datos de Secuencia Molecular , Conejos , Homología de Secuencia de Aminoácido
8.
Nat Commun ; 15(1): 5330, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909062

RESUMEN

Porcine deltacoronavirus (PDCoV) is an emerging enteric pathogen that has recently been detected in humans. Despite this zoonotic concern, the antigenic structure of PDCoV remains unknown. The virus relies on its spike (S) protein for cell entry, making it a prime target for neutralizing antibodies. Here, we generate and characterize a set of neutralizing antibodies targeting the S protein, shedding light on PDCoV S interdomain crosstalk and its vulnerable sites. Among the four identified antibodies, one targets the S1A domain, causing local and long-range conformational changes, resulting in partial exposure of the S1B domain. The other antibodies bind the S1B domain, disrupting binding to aminopeptidase N (APN), the entry receptor for PDCoV. Notably, the epitopes of these S1B-targeting antibodies are concealed in the prefusion S trimer conformation, highlighting the necessity for conformational changes for effective antibody binding. The binding footprint of one S1B binder entirely overlaps with APN-interacting residues and thus targets a highly conserved epitope. These findings provide structural insights into the humoral immune response against the PDCoV S protein, potentially guiding vaccine and therapeutic development for this zoonotic pathogen.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Deltacoronavirus , Epítopos , Glicoproteína de la Espiga del Coronavirus , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Animales , Anticuerpos Neutralizantes/inmunología , Porcinos , Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Humanos , Deltacoronavirus/inmunología , Deltacoronavirus/metabolismo , Antígenos CD13/metabolismo , Antígenos CD13/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Dominios Proteicos , Unión Proteica , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/inmunología , Células HEK293
9.
Toxicon X ; 21: 100185, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38425752

RESUMEN

Snakebite envenoming is a priority Neglected Tropical Disease that causes an estimated 81,000-135,000 fatalities each year. The development of a new generation of safer, affordable, and accessible antivenom therapies is urgently needed. With this goal in mind, rigorous characterisation of the specific toxins in snake venom is key to generating novel therapies for snakebite. Monoclonal antibodies directed against venom toxins are emerging as potentially strong candidates in the development of new snakebite diagnostics and treatment. Venoms comprise many different toxins of which several are responsible for their pathological effects. Due to the large variability of venoms within and between species, formulations of combinations of human antibodies are proposed as the next generation antivenoms. Here a high-throughput screening method employing antibody-based ligand fishing of venom toxins in 384 filter-well plate format has been developed to determine the antibody target/s The approach uses Protein G beads for antibody capture followed by exposure to a full venom or purified toxins to bind their respective ligand toxin(s). This is followed by a washing/centrifugation step to remove non-binding toxins and an in-well tryptic digest. Finally, peptides from each well are analysed by nanoLC-MS/MS and subsequent Mascot database searching to identify the bound toxin/s for each antibody under investigation. The approach was successfully validated to rapidly screen antibodies sourced from hybridomas, derived from venom-immunised mice expressing either regular human antibodies or heavy-chain-only human antibodies (HCAbs).

10.
Front Immunol ; 14: 1111385, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36895554

RESUMEN

Emerging SARS-CoV-2 variants have accrued mutations within the spike protein rendering most therapeutic monoclonal antibodies against COVID-19 ineffective. Hence there is an unmet need for broad-spectrum mAb treatments for COVID-19 that are more resistant to antigenically drifted SARS-CoV-2 variants. Here we describe the design of a biparatopic heavy-chain-only antibody consisting of six antigen binding sites recognizing two distinct epitopes in the spike protein NTD and RBD. The hexavalent antibody showed potent neutralizing activity against SARS-CoV-2 and variants of concern, including the Omicron sub-lineages BA.1, BA.2, BA.4 and BA.5, whereas the parental components had lost Omicron neutralization potency. We demonstrate that the tethered design mitigates the substantial decrease in spike trimer affinity seen for escape mutations for the hexamer components. The hexavalent antibody protected against SARS-CoV-2 infection in a hamster model. This work provides a framework for designing therapeutic antibodies to overcome antibody neutralization escape of emerging SARS-CoV-2 variants.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , Humanos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Cadenas Pesadas de Inmunoglobulina/genética , Anticuerpos Monoclonales
11.
Am J Pathol ; 178(1): 55-60, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224043

RESUMEN

Bone marrow-derived mononuclear cells (BMMNCs) enhance postischemic neovascularization, and their therapeutic use is currently under clinical investigation. However, cardiovascular risk factors, including diabetes mellitus and hypercholesterolemia, lead to the abrogation of BMMNCs proangiogenic potential. NO has been shown to be critical for the proangiogenic function of BMMNCs, and increased endothelial NO synthase (eNOS) activity promotes vessel growth in ischemic conditions. We therefore hypothesized that eNOS overexpression could restore both the impaired neovascularization response and decreased proangiogenic function of BMMNCs in clinically relevant models of diabetes and hypercholesterolemia. Transgenic eNOS overexpression in diabetic, atherosclerotic, and wild-type mice induced a 1.5- to 2.3-fold increase in postischemic neovascularization compared with control. eNOS overexpression in diabetic or atherosclerotic BMMNCs restored their reduced proangiogenic potential in ischemic hind limb. This effect was associated with an increase in BMMNC ability to differentiate into cells with endothelial phenotype in vitro and in vivo and an increase in BMMNCs paracrine function, including vascular endothelial growth factor A release and NO-dependent vasodilation. Moreover, although wild-type BMMNCs treatment resulted in significant progression of atherosclerotic plaque in ischemic mice, eNOS transgenic atherosclerotic BMMNCs treatment even had antiatherogenic effects. Cell-based eNOS gene therapy has both proangiogenic and antiatherogenic effects and should be further investigated for the development of efficient therapeutic neovascularization designed to treat ischemic cardiovascular disease.


Asunto(s)
Aterosclerosis/terapia , Terapia Genética/métodos , Isquemia/terapia , Monocitos/enzimología , Monocitos/trasplante , Neovascularización Fisiológica/genética , Óxido Nítrico Sintasa de Tipo III/genética , Animales , Apolipoproteínas E/genética , Aterosclerosis/fisiopatología , Miembro Posterior/irrigación sanguínea , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos
12.
Methods Mol Biol ; 2446: 121-141, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35157271

RESUMEN

The antibody repertoires of transgenic mice expressing human heavy chain only antibodies (HCAbs) can be retrieved from immune cells after antigen challenge. Compared with genetically modified rodents expressing conventional human antibodies (tetramers consisting of two heavy chains paired with two light chains), there is no chain pairing problem, since each antibody consists of a heavy chain dimer which is solely responsible for antigen binding. HCAbs can be obtained by classical hybridoma fusion, or the generation of phage libraries or eukaryotic cell libraries displaying or secreting HCAbs. Combined transcriptomic/serum proteomic approaches can also be used to determine the repertoire of antibodies, as well as single cell technologies such as the Beacon system that enable capture of immune cells of interest, analysis, and sequencing of antibodies in a short period of time. Here, we describe a protocol for obtaining monoclonal HCAbs from immunized Harbour transgenic mice through the generation and screening of HEK cell libraries of secreted antibodies. The method can be used routinely and is fast and affordable for everyone. Selected VH regions (single domains) are sequenced and individual HCAbs can be produced and purified from the same expression vector that is used for library generation (hIgG1 Fc). They can also be cloned into other expression plasmids and reformatted to equip them with a particular effector function, modify lifespan in serum, or optimize valency and avidity depending on the specific aim.


Asunto(s)
Anticuerpos de Dominio Único , Animales , Humanos , Inmunoglobulina G , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Ratones Transgénicos , Proteómica
13.
Nat Commun ; 13(1): 2921, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35614127

RESUMEN

Human coronavirus OC43 is a globally circulating common cold virus sustained by recurrent reinfections. How it persists in the population and defies existing herd immunity is unknown. Here we focus on viral glycoprotein S, the target for neutralizing antibodies, and provide an in-depth analysis of its antigenic structure. Neutralizing antibodies are directed to the sialoglycan-receptor binding site in S1A domain, but, remarkably, also to S1B. The latter block infection yet do not prevent sialoglycan binding. While two distinct neutralizing S1B epitopes are readily accessible in the prefusion S trimer, other sites are occluded such that their accessibility must be subject to conformational changes in S during cell-entry. While non-neutralizing antibodies were broadly reactive against a collection of natural OC43 variants, neutralizing antibodies generally displayed restricted binding breadth. Our data provide a structure-based understanding of protective immunity and adaptive evolution for this endemic coronavirus which emerged in humans long before SARS-CoV-2.


Asunto(s)
COVID-19 , Coronavirus Humano OC43 , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Coronavirus Humano OC43/metabolismo , Epítopos , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus
14.
Sci Immunol ; 7(73): eabp9312, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35471062

RESUMEN

The ongoing evolution of SARS-CoV-2 has resulted in the emergence of Omicron, which displays notable immune escape potential through mutations at key antigenic sites on the spike protein. Many of these mutations localize to the spike protein ACE2 receptor binding domain, annulling the neutralizing activity of therapeutic antibodies that were effective against other variants of concern (VOCs) earlier in the pandemic. Here, we identified a receptor-blocking human monoclonal antibody, 87G7, that retained potent in vitro neutralizing activity against SARS-CoV-2 variants including the Alpha, Beta, Gamma, Delta, and Omicron (BA.1/BA.2) VOCs. Using cryo-electron microscopy and site-directed mutagenesis experiments, we showed that 87G7 targets a patch of hydrophobic residues in the ACE2-binding site that are highly conserved in SARS-CoV-2 variants, explaining its broad neutralization capacity. 87G7 protected mice and hamsters prophylactically against challenge with all current SARS-CoV-2 VOCs and showed therapeutic activity against SARS-CoV-2 challenge in both animal models. Our findings demonstrate that 87G7 holds promise as a prophylactic or therapeutic agent for COVID-19 that is more resilient to SARS-CoV-2 antigenic diversity.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Anticuerpos Neutralizantes , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Microscopía por Crioelectrón , Humanos , Glicoproteínas de Membrana , Ratones , Pruebas de Neutralización , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas del Envoltorio Viral
15.
Nat Commun ; 12(1): 1715, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731724

RESUMEN

The coronavirus spike glycoprotein, located on the virion surface, is the key mediator of cell entry and the focus for development of protective antibodies and vaccines. Structural studies show exposed sites on the spike trimer that might be targeted by antibodies with cross-species specificity. Here we isolated two human monoclonal antibodies from immunized humanized mice that display a remarkable cross-reactivity against distinct spike proteins of betacoronaviruses including SARS-CoV, SARS-CoV-2, MERS-CoV and the endemic human coronavirus HCoV-OC43. Both cross-reactive antibodies target the stem helix in the spike S2 fusion subunit which, in the prefusion conformation of trimeric spike, forms a surface exposed membrane-proximal helical bundle. Both antibodies block MERS-CoV infection in cells and provide protection to mice from lethal MERS-CoV challenge in prophylactic and/or therapeutic models. Our work highlights an immunogenic and vulnerable site on the betacoronavirus spike protein enabling elicitation of antibodies with unusual binding breadth.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Betacoronavirus/inmunología , Epítopos/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/inmunología , Betacoronavirus/clasificación , Camelus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Reacciones Cruzadas , Epítopos/química , Epítopos/genética , Humanos , Ratones , Conformación Proteica , Subunidades de Proteína , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
16.
J Mol Cell Cardiol ; 48(6): 1041-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20153335

RESUMEN

Exercise training attenuates left ventricular (LV) dysfunction after myocardial infarction (MI). It could be speculated that these effects of exercise are mediated by increased endothelial NO synthase (eNOS) activity. In the present study we tested the hypothesis that eNOS plays a critical role in the exercise-induced amelioration of LV dysfunction after MI. MI or sham was induced in eNOS(-/-), eNOS(+/-) and eNOS(+/+) mice. After 8 weeks of voluntary wheel running (approximately 7 km/day in all groups) or sedentary housing, global cardiac function was determined in vivo and (immuno)histochemistry was performed to assess cardiomyocyte size, fibrosis, capillary density and apoptosis in remote myocardium. At baseline eNOS(-/-) mice had higher mean aortic pressure compared to eNOS(+/-) and eNOS(+/+) mice, but had normal global cardiac function. MI resulted in marked LV remodeling, including cardiomyocyte hypertrophy and a reduction in capillary density, increased fibrosis and apoptosis, as well as LV systolic and diastolic dysfunction to the same extent in all genotypes. In eNOS(+/+) MI mice exercise abolished fibrosis and apoptosis in the remote myocardium, attenuated LV systolic dysfunction and ameliorated pulmonary congestion. These beneficial effects were lost in eNOS(+/-) and eNOS(-/-) mice, while LV systolic dysfunction and pulmonary congestion in eNOS(+/-) mice were exacerbated by exercise. In conclusion, the beneficial effects of exercise after MI on LV remodeling and dysfunction depend critically on endogenous eNOS. The observation that the lack of one eNOS allele is sufficient to negate all beneficial effects of exercise, strongly suggests that exercise depends on full eNOS expression.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Infarto del Miocardio/rehabilitación , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Condicionamiento Físico Animal , Animales , Apoptosis , Capilares/patología , Colágeno/química , Femenino , Ventrículos Cardíacos/patología , Humanos , Hipertrofia , Masculino , Ratones , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno
17.
Biochim Biophys Acta ; 1791(8): 790-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19422933

RESUMEN

Phospholipid transfer protein (PLTP) is associated with HDL particles in plasma, where it transfers phospholipids between lipoproteins and remodels HDL particles. Tangier disease patients, with a mutated ABCA1 transporter, have extremely low plasma HDL concentration and reduced PLTP activity levels, a phenotype that is also observed in mice lacking ABCA1. We investigated whether low HDL levels and low PLTP activity are mechanistically related. Firstly, we studied PLTP expression and distribution among lipoproteins in mice lacking ABCA1 (ABCA1(-/-)). Parallel to the strong reduction in PLTP activity in plasma of ABCA1(-/-) mice, decreased PLTP protein levels were observed. Neither PLTP synthesis in liver or macrophages nor the ability of the macrophages to secrete PLTP were impaired in ABCA1(-/-) mice. However, the PLTP activity level in the medium of cultured macrophages was determined by HDL levels in the medium. PLTP was associated with HDL particles in wild type mice, whereas in ABCA1(-/-) mice, PLTP was associated with VLDL and LDL particles. Secondly, we treated different mouse models with varying plasma HDL and PLTP levels (wild type, ABCA1(-/-), apoE(-/-) and PLTPtg mice, overexpressing human PLTP) with a synthetic LXR ligand, and investigated the relationship between LXR-mediated PLTP induction and HDL levels in plasma. Plasma PLTP activity in wild type mice was induced 5.6-fold after LXR activation, whereas in ABCA1(-/-), apoE(-/-) and PLTPtg mice, all having reduced HDL levels, induction of PLTP activity was 2.4- , 3.2- and 2.0-fold, respectively. The less pronounced PLTP induction in these mice compared to wild type mice was not caused by a decreased PLTP gene expression in the liver or macrophages. Our findings indicate that the extent of LXR-mediated PLTP induction depends on plasma HDL levels. In conclusion, we demonstrate that ABCA1 deficiency in mice affects plasma PLTP level and distribution through an indirect effect on HDL metabolism. In addition, we show that the extent of LXR-mediated PLTP induction is HDL-dependent. These findings indicate that plasma HDL level is an important regulator of plasma PLTP and might play a role in the stabilization of PLTP in plasma.


Asunto(s)
Lipoproteínas HDL/sangre , Proteínas de Transferencia de Fosfolípidos/sangre , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Humanos , Receptores X del Hígado , Macrófagos/metabolismo , Ratones , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/metabolismo
18.
Biochim Biophys Acta ; 1791(10): 1031-6, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19524061

RESUMEN

OBJECTIVE: Elevated plasma phospholipid transfer protein (PLTP) expression may increase atherosclerosis in mice by reducing plasma HDL and increasing hepatic VLDL secretion. Hepatic lipase (HL) is a lipolytic enzyme involved in several aspects of the same pathways of lipoprotein metabolism. We investigated whether the effects of elevated PLTP activity are compromised by HL deficiency. METHODS AND RESULTS: HL deficient mice were crossbred with PLTP transgenic (PLTPtg) mice and studied in the fasted state. Plasma triglycerides were decreased in HL deficiency, explained by reduced hepatic triglyceride secretion. In PLTPtg mice, a redistribution of HL activity between plasma and tissue was evident and plasma triglycerides were also decreased. HL deficiency mitigated or even abolished the stimulatory effect of elevated PLTP activity on hepatic triglyceride secretion. HL deficiency had a modest incremental effect on plasma HDL, which remained present in PLTP transgenic/HL(-/-) mice, thereby partially compensating the decrease in HDL caused by elevation of PLTP activity. HDL decay experiments showed that the fractional turnover rate of HDL cholesteryl esters was delayed in HL deficient mice, increased in PLTPtg mice and intermediate in PLTPtg mice in an HL(-/-) background. CONCLUSIONS: HL affects hepatic VLDL. Elevated PLTP activity lowers plasma HDL-cholesterol by stimulating the plasma turnover and hepatic uptake of HDL cholesteryl esters. HL is not required for the increase in hepatic triglyceride secretion or for the lowering of HDL-cholesterol induced by PLTP overexpression.


Asunto(s)
HDL-Colesterol/metabolismo , VLDL-Colesterol/metabolismo , Lipasa/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Animales , HDL-Colesterol/sangre , VLDL-Colesterol/sangre , Lipasa/sangre , Lipoproteína Lipasa/sangre , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Transferencia de Fosfolípidos/sangre
19.
J Clin Invest ; 117(3): 616-26, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17304353

RESUMEN

We previously found that low shear stress (LSS) induces atherosclerotic plaques in mice with increased lipid and matrix metalloproteinase content and decreased vascular smooth muscle and collagen content. Here, we evaluated the role of chemokines in this process, using an extravascular device inducing regions of LSS, high shear stress, and oscillatory shear stress (OSS) in the carotid artery. One week of shear stress alterations induced expression of IFN-gamma-inducible protein-10 (IP-10) exclusively in the LSS region, whereas monocyte chemoattractant protein-1 (MCP-1) and the mouse homolog of growth-regulated oncogene alpha (GRO-alpha) were equally upregulated in both LSS and OSS regions. After 3 weeks, GRO-alpha and IP-10 were specifically upregulated in LSS regions. After 9 weeks, lesions with thinner fibrous caps and larger necrotic cores were found in the LSS region compared with the OSS region. Equal levels of MCP-1 expression were observed in both regions, while expression of fractalkine was found in the LSS region only. Blockage of fractalkine inhibited plaque growth and resulted in striking differences in plaque composition in the LSS region. We conclude that LSS or OSS triggers expression of chemokines involved in atherogenesis. Fractalkine upregulation is critically important for the composition of LSS-induced atherosclerotic lesions.


Asunto(s)
Aterosclerosis/etiología , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/etiología , Quimiocinas/fisiología , Resistencia al Corte , Animales , Apolipoproteínas E/genética , Aterosclerosis/patología , Receptor 1 de Quimiocinas CX3C , Arterias Carótidas/química , Enfermedades de las Arterias Carótidas/patología , Quimiocinas/genética , Expresión Génica , Ratones , Ratones Mutantes , Receptores de Citocinas/análisis , Receptores del VIH/análisis , Estrés Mecánico
20.
Nat Commun ; 11(1): 2251, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366817

RESUMEN

The emergence of the novel human coronavirus SARS-CoV-2 in Wuhan, China has caused a worldwide epidemic of respiratory disease (COVID-19). Vaccines and targeted therapeutics for treatment of this disease are currently lacking. Here we report a human monoclonal antibody that neutralizes SARS-CoV-2 (and SARS-CoV) in cell culture. This cross-neutralizing antibody targets a communal epitope on these viruses and may offer potential for prevention and treatment of COVID-19.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , Afinidad de Anticuerpos/inmunología , Betacoronavirus/química , Betacoronavirus/efectos de los fármacos , COVID-19 , Chlorocebus aethiops , Secuencia Conservada , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Reacciones Cruzadas/inmunología , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Modelos Moleculares , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Unión Proteica/efectos de los fármacos , Dominios Proteicos/inmunología , Receptores Virales/química , Receptores Virales/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA