Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 1073-1082, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38816615

RESUMEN

A key barrier to the development of vaccines that induce broadly neutralizing antibodies (bnAbs) against human immunodeficiency virus (HIV) and other viruses of high antigenic diversity is the design of priming immunogens that induce rare bnAb-precursor B cells. The high neutralization breadth of the HIV bnAb 10E8 makes elicitation of 10E8-class bnAbs desirable; however, the recessed epitope within gp41 makes envelope trimers poor priming immunogens and requires that 10E8-class bnAbs possess a long heavy chain complementarity determining region 3 (HCDR3) with a specific binding motif. We developed germline-targeting epitope scaffolds with affinity for 10E8-class precursors and engineered nanoparticles for multivalent display. Scaffolds exhibited epitope structural mimicry and bound bnAb-precursor human naive B cells in ex vivo screens, protein nanoparticles induced bnAb-precursor responses in stringent mouse models and rhesus macaques, and mRNA-encoded nanoparticles triggered similar responses in mice. Thus, germline-targeting epitope scaffold nanoparticles can elicit rare bnAb-precursor B cells with predefined binding specificities and HCDR3 features.


Asunto(s)
Vacunas contra el SIDA , Anticuerpos Neutralizantes , Anticuerpos Anti-VIH , Proteína gp41 de Envoltorio del VIH , Infecciones por VIH , VIH-1 , Macaca mulatta , Animales , Humanos , Proteína gp41 de Envoltorio del VIH/inmunología , Anticuerpos Anti-VIH/inmunología , Ratones , Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/inmunología , VIH-1/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Vacunación , Anticuerpos ampliamente neutralizantes/inmunología , Linfocitos B/inmunología , Nanopartículas/química , Femenino , Regiones Determinantes de Complementariedad/inmunología , Epítopos/inmunología
2.
Immunity ; 57(5): 1141-1159.e11, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38670113

RESUMEN

Broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem of influenza A viruses (IAVs) tend to be effective against either group 1 or group 2 viral diversity. In rarer cases, intergroup protective bnAbs can be generated by human antibody paratopes that accommodate the conserved glycan differences between the group 1 and group 2 stems. We applied germline-engaging nanoparticle immunogens to elicit a class of cross-group bnAbs from physiological precursor frequency within a humanized mouse model. Cross-group protection depended on the presence of the human bnAb precursors within the B cell repertoire, and the vaccine-expanded antibodies enriched for an N55T substitution in the CDRH2 loop, a hallmark of the bnAb class. Structurally, this single mutation introduced a flexible fulcrum to accommodate glycosylation differences and could alone enable cross-group protection. Thus, broad IAV immunity can be expanded from the germline repertoire via minimal antigenic input and an exceptionally simple antibody development pathway.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Vacunación , Animales , Ratones , Humanos , Anticuerpos Antivirales/inmunología , Vacunas contra la Influenza/inmunología , Virus de la Influenza A/inmunología , Anticuerpos Neutralizantes/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Sustitución de Aminoácidos , Linfocitos B/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Anticuerpos ampliamente neutralizantes/inmunología
4.
Proc Natl Acad Sci U S A ; 119(25): e2203326119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696580

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of morbidity and hospitalization among children worldwide, however, no vaccines or therapeutics are currently available for hMPV disease prevention and treatment. The hMPV fusion (F) protein is the sole target of neutralizing antibodies. To map the immunodominant epitopes on the hMPV F protein, we isolated a panel of human monoclonal antibodies (mAbs), and the mAbs were assessed for binding avidity, neutralization potency, and epitope specificity. We found the majority of the mAbs target diverse epitopes on the hMPV F protein, and we discovered multiple mAb binding approaches for antigenic site III. The most potent mAb, MPV467, which had picomolar potency, was examined in prophylactic and therapeutic mouse challenge studies, and MPV467 limited virus replication in mouse lungs when administered 24 h before or 72 h after viral infection. We determined the structure of MPV467 in complex with the hMPV F protein using cryo-electron microscopy to a resolution of 3.3 Å, which revealed a complex novel prefusion-specific epitope overlapping antigenic sites II and V on a single protomer. Overall, our data reveal insights into the immunodominant antigenic epitopes on the hMPV F protein, identify a mAb therapy for hMPV F disease prevention and treatment, and provide the discovery of a prefusion-specific epitope on the hMPV F protein.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Antígenos Virales , Metapneumovirus , Infecciones por Paramyxoviridae , Proteínas Virales de Fusión , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/aislamiento & purificación , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/química , Antígenos Virales/inmunología , Microscopía por Crioelectrón , Epítopos/inmunología , Humanos , Metapneumovirus/inmunología , Ratones , Infecciones por Paramyxoviridae/prevención & control , Prevención Primaria , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología
5.
J Virol ; 95(18): e0059321, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34160259

RESUMEN

Human metapneumovirus (hMPV) is an important cause of acute viral respiratory infection. As the only target of neutralizing antibodies, the hMPV fusion (F) protein has been a major focus for vaccine development and targeting by drugs and monoclonal antibodies (MAbs). While X-ray structures of trimeric prefusion and postfusion hMPV F proteins from genotype A, and monomeric prefusion hMPV F protein from genotype B have been determined, structural data for the postfusion conformation for genotype B is lacking. We determined the crystal structure of this protein and compared the structural differences of postfusion hMPV F between hMPV A and B genotypes. We also assessed the receptor binding properties of the hMPV F protein to heparin and heparan sulfate (HS). A library of HS oligomers was used to verify the HS binding activity of hMPV F, and several compounds showed binding to predominantly prefusion hMPV F, but had limited binding to postfusion hMPV F. Furthermore, MAbs to antigenic sites III and the 66-87 intratrimeric epitope block heparin binding. In addition, we evaluated the efficacy of postfusion hMPV B2 F protein as a vaccine candidate in BALB/c mice. Mice immunized with hMPV B2 postfusion F protein showed a balanced Th1/Th2 immune response and generated neutralizing antibodies against both subgroup A2 and B2 hMPV strains, which protected the mice from hMPV challenge. Antibody competition analysis revealed the antibodies generated by immunization target two known antigenic sites (III and IV) on the hMPV F protein. Overall, this study provides new characteristics of the hMPV F protein, which may be informative for vaccine and therapy development. IMPORTANCE Human metapneumovirus (hMPV) is an important cause of viral respiratory disease. In this paper, we report the X-ray crystal structure of the hMPV fusion (F) protein in the postfusion conformation from genotype B. We also assessed binding of the hMPV F protein to heparin and heparan sulfate, a previously reported receptor for the hMPV F protein. Furthermore, we determined the immunogenicity and protective efficacy of postfusion hMPV B2 F protein, which is the first study using a homogenous conformation of the protein. Antibodies generated in response to vaccination give a balanced Th1/Th2 response and target two previously discovered neutralizing epitopes.


Asunto(s)
Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Heparina/metabolismo , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Femenino , Heparina/análogos & derivados , Humanos , Inmunización , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Paramyxoviridae/virología , Unión Proteica , Conformación Proteica , Proteoglicanos/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Proteínas Virales de Fusión/metabolismo
6.
PLoS Pathog ; 16(10): e1008942, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33035266

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of viral respiratory infection in children, and can cause severe lower respiratory tract infection in infants, the elderly, and immunocompromised patients. However, there remain no licensed vaccines or specific treatments for hMPV infection. Although the hMPV fusion (F) protein is the sole target of neutralizing antibodies, the immunological properties of hMPV F remain poorly understood. To further define the humoral immune response to the hMPV F protein, we isolated two new human monoclonal antibodies (mAbs), MPV458 and MPV465. Both mAbs are neutralizing in vitro and were determined to target a unique antigenic site using competitive biolayer interferometry. We determined both MPV458 and MPV465 have higher affinity for monomeric hMPV F than trimeric hMPV F. MPV458 was co-crystallized with hMPV F, and the mAb primarily interacts with an alpha helix on the F2 region of the hMPV F protein. Surprisingly, the major epitope for MPV458 lies within the trimeric interface of the hMPV F protein, suggesting significant breathing of the hMPV F protein must occur for host immune recognition of the novel epitope. In addition, significant glycan interactions were observed with a somatically mutated light chain framework residue. The data presented identifies a novel epitope on the hMPV F protein for epitope-based vaccine design, and illustrates a new mechanism for human antibody neutralization of viral glycoproteins.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Pneumovirus/inmunología , Anticuerpos Neutralizantes/farmacología , Epítopos/inmunología , Humanos , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/virología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología
7.
Infect Immun ; 89(5)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33649050

RESUMEN

Streptococcus pneumoniae remains a leading cause of bacterial pneumonia despite the widespread use of vaccines. While vaccines are effective at reducing the incidence of most serotypes included in vaccines, a rise in infection due to nonvaccine serotypes and moderate efficacy against some vaccine serotypes have contributed to high disease incidence. Additionally, numerous isolates of S. pneumoniae are antibiotic or multidrug resistant. Several conserved pneumococcal proteins prevalent in the majority of serotypes have been examined for their potential as vaccines in preclinical and clinical trials. An additional, yet-unexplored tool for disease prevention and treatment is the use of human monoclonal antibodies (MAbs) targeting conserved pneumococcal proteins. Here, we isolated the first human MAbs (PhtD3, PhtD6, PhtD7, PhtD8, and PspA16) against the pneumococcal histidine triad protein (PhtD) and the pneumococcal surface protein A (PspA), two conserved and protective antigens. MAbs to PhtD target diverse epitopes on PhtD, and MAb PspA16 targets the N-terminal segment of PspA. The PhtD-specific MAbs bind to multiple serotypes, while MAb PspA16 serotype breadth is limited. MAbs PhtD3 and PhtD8 prolong the survival of mice infected with pneumococcal serotype 3. Furthermore, MAb PhtD3 prolongs the survival of mice in intranasal and intravenous infection models with pneumococcal serotype 4 and in mice infected with pneumococcal serotype 3 when administered 24 h after pneumococcal infection. All PhtD and PspA MAbs demonstrate opsonophagocytic activity, suggesting a potential mechanism of protection. Our results identify new human MAbs for pneumococcal disease prevention and treatment and identify epitopes on PhtD and PspA recognized by human B cells.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Interacciones Huésped-Patógeno/inmunología , Hidrolasas/antagonistas & inhibidores , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/inmunología , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Relación Dosis-Respuesta Inmunológica , Epítopos/inmunología , Humanos , Hidrolasas/inmunología , Unión Proteica , Serogrupo
8.
J Virol ; 93(19)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31292250

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of viral lower respiratory tract infection in children. The sole target of neutralizing antibodies targeting hMPV is the fusion (F) protein, a class I viral fusion protein mediating virus-cell membrane fusion. There have been several monoclonal antibodies (mAbs) isolated that neutralize hMPV; however, determining the antigenic sites on the hMPV F protein mediating such neutralizing antibody generation would assist efforts for effective vaccine design. In this report, the isolation and characterization of four new human mAbs, termed MPV196, MPV201, MPV314, and MPV364, are described. Among the four mAbs, MPV364 was found to be the most potent neutralizing mAb in vitro Binding studies with monomeric and trimeric hMPV F revealed that MPV364 had the weakest binding affinity for monomeric hMPV F compared to the other three mAbs, yet binding experiments with trimeric hMPV F showed limited differences in binding affinity, suggesting that MPV364 targets an antigenic site incorporating two protomers. Epitope binning studies showed that MPV364 targets antigenic site III on the hMPV F protein and competes for binding with previously discovered mAbs MPE8 and 25P13, both of which cross-react with the respiratory syncytial virus (RSV) F protein. However, MPV364 does not cross-react with the RSV F protein, and the competition profile suggests that it binds to the hMPV F protein in a binding pose slightly shifted from mAbs MPE8 and 25P13. MPV364 was further assessed in vivo and was shown to substantially reduce viral replication in the lungs of BALB/c mice. Overall, these data reveal a new binding region near antigenic site III of the hMPV F protein that elicits potent neutralizing hMPV F-specific mAbs and provide a new panel of neutralizing mAbs that are candidates for therapeutic development.IMPORTANCE Recent progress in understanding the human immune response to respiratory syncytial virus has paved the way for new vaccine antigens and therapeutics to prevent and treat disease. Progress toward understanding the immune response to human metapneumovirus (hMPV) has lagged behind, although hMPV is a leading cause of lower respiratory tract infection in children. In this report, we advanced the field by isolating a panel of human mAbs to the hMPV F protein. One potent neutralizing mAb, MPV364, targets antigenic site III on the hMPV F protein and incorporates two protomers into its epitope yet is unique from previously discovered site III mAbs, as it does not cross-react with the RSV F protein. We further examined MPV364 in vivo and found that it limits viral replication in BALB/c mice. Altogether, these data provide new mAb candidates for therapeutic development and provide insights into hMPV vaccine development.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Epítopos de Linfocito B/inmunología , Metapneumovirus/inmunología , Proteínas Virales de Fusión/inmunología , Sitios de Unión , Mapeo Epitopo , Humanos , Unión Proteica
9.
Angew Chem Int Ed Engl ; 59(34): 14449-14457, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32472603

RESUMEN

A distannylated electron-deficient bithiophene imide (BTI-Tin) monomer was synthesized and polymerized with imide-functionalized co-units to afford homopolymer PBTI and copolymer P(BTI-BTI2), both featuring an acceptor-acceptor backbone with high molecular weight. Both polymers exhibited excellent unipolar n-type character in transistors with electron mobility up to 2.60 cm2 V-1 s-1 . When applied as acceptor materials in all-polymer solar cells, PBTI and P(BTI-BTI2) achieved high power-conversion efficiency (PCE) of 6.67 % and 8.61 %, respectively. The PCE (6.67 %) of polymer PBTI, synthesized from the distannylated monomer, is much higher than that (0.14 %) of the same polymer PBTI*, synthesized from typical dibrominated monomer. The 8.61 % PCE of copolymer P(BTI-BTI2) is also higher than those (<1 %) of homopolymers synthesized from dibrominated monomers. The results demonstrate the success of BTI-Tin for accessing n-type polymers with greatly improved device performance.

10.
Appl Environ Microbiol ; 84(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29802193

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) strains producing heat-labile toxin (LT) and/or heat-stable toxin (STa) are a top cause of children's diarrhea and travelers' diarrhea. Holotoxin-structured GM1-binding LT is a strong immunogen and an effective adjuvant, and can serve a carrier or a platform for multivalent vaccine development. However, the significance of peptide domains or epitopes of LT particularly enzymatic LTA subunit in association with LT enterotoxicity and immunogenicity has not been characterized. In this study, we identified B-cell epitopes in silico from LTA subunit and examined epitopes for immunogenicity and association with LT enterotoxicity. Epitopes identified from LTA subunit were individually fused to a modified chicken ovalbumin carrier protein, and each epitope-ovalbumin fusion was used to immunize mice. Data showed all 11 LTA epitopes were immunogenic; epitope 7 (105SPHPYEQEVSA115) induced greater titers of anti-LT antibodies which neutralized LT enterotoxicity more effectively. To examine these epitopes for the significance in LT enterotoxicity, we constructed LT mutants by substituting each of 10 epitopes at the toxic A1 domain of LTA subunit with a foreign epitope and examined LT mutants for enterotoxicity and GM1-binding activity. Data showed that LT mutants exhibited no enterotoxicity but retained GM1-binding activity. The results from this study indicated that while not all immunodominant LTA epitopes were neutralizing, LT mutants with an individual epitope substituted lost enterotoxicity but retained GM1-binding activity. These results provided additional information to understand LT immunogenicity and enterotoxicity and suggested the potential application of LT platform for multivalent vaccines against ETEC diarrhea and other diseases.IMPORTANCE No vaccine is licensed for enterotoxigenic Escherichia coli (ETEC) strains, which remain a leading cause of diarrhea in children from developing countries and international travelers. GM1-binding heat-labile toxin (LT) which is a key virulence factor of ETEC diarrhea is a strong vaccine antigen and a self-adjuvant. LT can also serve a backbone or platform for MEFA (multiepitope fusion antigen), a newly developed structural vaccinology technology, to present heterogeneous epitopes (by replacing LT epitopes) and to mimic epitope antigenicity for development of broadly protective vaccines. Data from this study identified neutralizing LT epitopes and demonstrated that substitution of LT epitopes eliminated LT enterotoxicity without altering GM1-binding activity, suggesting LT is potentially a versatile MEFA platform to present heterogeneous epitopes for multivalent vaccines against ETEC and other pathogens.


Asunto(s)
Toxinas Bacterianas/inmunología , Escherichia coli Enterotoxigénica/inmunología , Enterotoxinas/inmunología , Epítopos de Linfocito B/inmunología , Proteínas de Escherichia coli/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Pollos , Diarrea/inmunología , Diarrea/microbiología , Diarrea/prevención & control , Escherichia coli Enterotoxigénica/química , Escherichia coli Enterotoxigénica/genética , Enterotoxinas/administración & dosificación , Enterotoxinas/química , Enterotoxinas/genética , Epítopos de Linfocito B/química , Epítopos de Linfocito B/genética , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/administración & dosificación , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Vacunas contra Escherichia coli/administración & dosificación , Vacunas contra Escherichia coli/química , Vacunas contra Escherichia coli/genética , Vacunas contra Escherichia coli/inmunología , Femenino , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Ovalbúmina/genética , Ovalbúmina/inmunología
11.
Appl Environ Microbiol ; 84(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29079628

RESUMEN

Heat-stable toxin (STa)-producing enterotoxigenic Escherichia coli (ETEC) strains are a top cause of moderate-to-severe diarrhea in children from developing countries and a common cause of travelers' diarrhea. Recent progress in using STa toxoids and toxoid fusions to induce neutralizing anti-STa antibodies has accelerated ETEC vaccine development. However, concern remains regarding whether the derived anti-STa antibodies cross-react with STa-like guanylin and uroguanylin, two guanylate cyclase C (GC-C) ligands regulating fluid and electrolyte transportation in human intestinal and renal epithelial cells. To further divert STa from guanylin and uroguanylin structurally and antigenically and to eliminate anti-STa antibody cross-reactivity with guanylin and uroguanylin, we mutated STa at the 9th (leucine), 12th (asparagine), and 14th (alanine) residues for the double and triple mutants STaL9A/N12S, STaL9A/A14H, STaN12S/A14T, and STaL9A/N12S/A14H We then fused each STa mutant (three copies) to a monomeric heat-labile toxin (LT) mutant (mnLTR192G/L211A) for the toxoid fusions 3×STaL9A/N12S-mnLTR192G/L211A, 3×STaL9A/A14H-mnLTR192G/L211A, 3×STaN12S/A14T-mnLTR192G/L211A, and 3×STaL9A/N12S/A14H-mnLTR192G/L211A; examined each fusion for anti-STa immunogenicity; and assessed the derived antibodies for in vitro neutralization activity against STa toxicity and for cross-reactivity with guanylin and uroguanylin. Mice subcutaneously immunized with each fusion protein developed anti-STa antibodies, and the antibodies derived from 3×STaN12S-mnLTR192G/L211A, 3×STaL9A/N12S-mnLTR192G/L211A, or 3×STaN12S/A14T-mnLTR192G/L211A prevented STa from the stimulation of intracellular cGMP in T-84 cells. Competitive enzyme-linked immunosorbent assays (ELISAs) showed that guanylin and uroguanylin hardly blocked the binding of anti-STa antibodies to the coated STa-ovalbumin conjugate. These results indicated that antibodies derived from 3×STaN12S-mnLTR192G/L211A, 3×STaL9A/N12S-mnLTR192G/L211A, or 3×STaN12S/A14T-mnLTR192G/L211A neutralized STa and had little cross-reactivity with guanylin and uroguanylin, suggesting that these toxoid fusions are suitable antigens for ETEC vaccines.IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) strains are a leading cause of children's diarrhea and travelers' diarrhea. Currently, there is no licensed vaccine against ETEC diarrhea. One key challenge is to identify safe antigens to induce antibodies neutralizing the key STa without cross-reacting with guanylin and uroguanylin, two important ligands controlling homeostasis in human intestinal and renal epithelial cells. In this study, we generated nontoxic fusion antigens that induced antibodies that neutralize STa enterotoxicity in vitro and do not cross-react with guanylin or uroguanylin. These fusions have become the preferred antigens for the development of ETEC vaccines to potentially prevent the deaths of hundreds of thousands of young children and hundreds of millions of diarrheal cases each year.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Escherichia coli Enterotoxigénica/inmunología , Hormonas Gastrointestinales/inmunología , Péptidos Natriuréticos/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Niño , Reacciones Cruzadas , Escherichia coli Enterotoxigénica/genética , Enterotoxinas/genética , Enterotoxinas/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Femenino , Calor , Humanos , Inmunización , Ratones , Mutación , Toxoides/inmunología
12.
J Am Chem Soc ; 139(45): 16092-16095, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29112393

RESUMEN

We report a novel small molecule acceptor (SMA) named FTTB-PDI4 obtained via ring-fusion between the thiophene and perylene diimide (PDI) units of a PDI-tetramer with a tetrathienylbezene (TTB) core. A small voltage loss of 0.53 V and a high power conversion efficiency of 10.58% were achieved, which is the highest value reported for PDI-based devices to date. By comparing the fused and nonfused SMAs, we show that the ring-fusion introduces several beneficial effects on the properties and performances of the acceptor material, including more favorable energy levels, enhanced light absorption and stronger intermolecular packing. Interestingly, morphology data reveal that the fused molecule yields higher domain purity and thus can better maintain its molecular packing and electron mobility in the blend. Theoretical calculations also demonstrate that FTTB-PDI4 exhibits a "double-decker" geometry with two pairs of mostly parallel PDI units, which is distinctively different from reported PDI-tetramers with highly twisted geometries and can explain the better performance of the material. This work highlights the promising design of PDI-based acceptors by the ring-fusion strategy.

13.
Nat Commun ; 15(1): 1335, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38351001

RESUMEN

Many pathogenic viruses rely on class I fusion proteins to fuse their viral membrane with the host cell membrane. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more stable postfusion state. Mounting evidence underscores that antibodies targeting the prefusion conformation are the most potent, making it a compelling vaccine candidate. Here, we establish a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. With this protocol, we stabilize the fusion proteins of the RSV, hMPV, and SARS-CoV-2 viruses, testing fewer than a handful of designs. The solved structures of these designed proteins from all three viruses evidence the atomic accuracy of our approach. Furthermore, the humoral response of the redesigned RSV F protein compares to that of the recently approved vaccine in a mouse model. While the parallel design of two conformations allows the identification of energetically sub-optimal positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.


Asunto(s)
Vacunas , Proteínas Virales de Fusión , Animales , Ratones , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Conformación Proteica
14.
bioRxiv ; 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36993551

RESUMEN

Many pathogenic viruses, including influenza virus, Ebola virus, coronaviruses, and Pneumoviruses, rely on class I fusion proteins to fuse viral and cellular membranes. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more favorable and stable postfusion state. An increasing amount of evidence exists highlighting that antibodies targeting the prefusion conformation are the most potent. However, many mutations have to be evaluated before identifying prefusion-stabilizing substitutions. We therefore established a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. As a proof of concept, we applied this principle to the fusion protein of the RSV, hMPV, and SARS-CoV-2 viruses. For each protein, we tested less than a handful of designs to identify stable versions. Solved structures of designed proteins from the three different viruses evidenced the atomic accuracy of our approach. Furthermore, the immunological response of the RSV F design compared to a current clinical candidate in a mouse model. While the parallel design of two conformations allows identifying and selectively modifying energetically less optimized positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. We recaptured many approaches previously introduced manually for the stabilization of viral surface proteins, such as cavity-filling, optimization of polar interactions, as well as postfusion-disruptive strategies. Using our approach, it is possible to focus on the most impacting mutations and potentially preserve the immunogen as closely as possible to its native version. The latter is important as sequence re-design can cause perturbations to B and T cell epitopes. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.

15.
Phys Med Biol ; 68(17)2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37578025

RESUMEN

Objective.In clinical proton therapy, the spread-out Bragg peak (SOBP) is commonly used to fit the target shape. Dose depositions at microscopic sites vary, even with a consistent absorbed dose (D) in SOBP. In the present study, monolayer mesh-type cell population models were developed for microdosimetric assessment at different SOBP depths.Approach.Normal human bronchial epithelial (BEAS-2B) and hepatocytes (L-O2) mesh-type cell models were constructed based on fluorescence tomography images of normal human cells. Particle transport simulation in cell populations was performed coupled with Monte Carlo software PHITS. The relationship between microdosimetry and macrodosimetry of SOBP at different depths was described by analyzing the microdosimetric indicators such as specific energyz,specific energy distributionfz,D,and relative standard deviationσz/z¯within cells. Additionally, the microdosimetric distributions characteristics and their contributing factors were also discussed.Main results.The microscopic dose distribution is strongly influenced by cellular size, shape, and material. The mean specific energyz¯of nucleus and cytoplasm in the cell population is greater than the overall absorbed dose of the cell population model (Dp), with a maximumz¯/Dpof 1.1. The cellular dose distribution is different between the BEAS-2B mesh-type model and its concentric ellipsoid geometry-type model, which difference inz¯is about 10.3% for the nucleus and about 7.5% for the cytoplasm with the SOBP depth of 15 cm. WhenD= 2 Gy, the maximumzof L-O2 nucleus reaches 2.8 Gy andσz/z¯is 5.1% at the mid-depth SOBP (16-18 cm); while the maximumzof the BEAS-2B nucleus reaches 2.2 Gy with only 2.7% ofσz/z¯.Significance.The significant variation of microdosimetric distributions of SOBP different depths indicates the necessity to use mesh-type cell population models, which have the potential to be compared with biological results and build the bio-physical model.


Asunto(s)
Terapia de Protones , Protones , Humanos , Radiometría/métodos , Simulación por Computador , Programas Informáticos , Método de Montecarlo , Efectividad Biológica Relativa
16.
Front Immunol ; 13: 941865, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36003370

RESUMEN

Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are two leading causes of severe respiratory infections in children, the elderly, and immunocompromised patients. The fusion (F) protein is the major target of neutralizing antibodies. Recent developments in stabilizing the pre-fusion conformation of the F proteins, and identifying immunodominant epitopes that elicit potent neutralizing antibodies have led to the testing of numerous pre-fusion RSV F-based vaccines in clinical trials. We designed and tested the immunogenicity and protective efficacy of a chimeric fusion protein that contains immunodominant epitopes of RSV F and hMPV F (RHMS-1). RHMS-1 has several advantages over vaccination with pre-fusion RSV F or hMPV F, including a focus on recalling B cells to the most important protective epitopes and the ability to induce protection against two viruses with a single antigen. RHMS-1 was generated as a trimeric recombinant protein, and analysis by negative-stain electron microscopy demonstrated the protein resembles the pre-fusion conformation. Probing of RHMS-1 antigenicity using a panel of RSV and hMPV F-specific monoclonal antibodies (mAbs) revealed the protein retains features of both viruses, including the pre-fusion site Ø epitope of RSV F. Mice immunized with RHMS-1 generated neutralizing antibodies to both viruses and were completely protected from RSV or hMPV challenge. Overall, this study demonstrates protection against two viruses with a single antigen and supports testing of RHMS-1 in additional pre-clinical animal models.


Asunto(s)
Metapneumovirus , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Vacunas Virales , Anciano , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Niño , Epítopos , Humanos , Epítopos Inmunodominantes/genética , Metapneumovirus/genética , Ratones , Proteínas Recombinantes , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales de Fusión/genética , Vacunas Virales/genética
17.
ACS Nano ; 16(8): 11902-11911, 2022 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-35866886

RESUMEN

Charge carrier nonradiative recombination (NRR) caused by interface defects and nonoptimal energy level alignment is the primary factor restricting the performance improvement of perovskite solar cells (PSCs). Interfacial modification is a vital strategy to restrain NRR and enable high-performance PSCs. We report here two interfacial materials, PhI-TPA and BTZI-TPA, consisting of phthalimide and a 2,1,3-benzothiadiazole-5,6-dicarboxylicimide core, respectively. The difunctionalized BTZI-TPA with imide and thiadiazole shows higher hole mobility, better aligned energy levels, and stronger interaction with uncoordinated Pb2+ on the perovskite surface, suppressing NRR and carrier accumulation at the interface of perovskite/spiro-OMeTAD and yielding enhanced open-circuit voltage and fill factor. Consequently, the PSC based on BTZI-TPA delivers a high efficiency of 24.06% with an excellent fill factor of 83.10%, superior to that (21.47%) of the reference cell without an interfacial layer, and 21.45% efficiency for the device with a scaled-up area (1.00 cm2). These results underscore the potential of imide and thiadiazole groups in developing interfacial layers with strong passivation capability, effective charge transport property, and fine-tuned energetics for stable and efficient PSCs.

18.
bioRxiv ; 2022 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-35702147

RESUMEN

Immunization with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines has greatly reduced coronavirus disease 2019 (COVID-19)-related deaths and hospitalizations, but waning immunity and the emergence of variants capable of immune escape indicate the need for novel SARS-CoV-2 vaccines. An intranasal parainfluenza virus 5 (PIV5)-vectored COVID-19 vaccine CVXGA1 has been proven efficacious in animal models and blocks contact transmission of SARS-CoV-2 in ferrets. CVXGA1 vaccine is currently in human clinical trials in the United States. This work investigates the immunogenicity and efficacy of CVXGA1 and other PIV5-vectored vaccines expressing additional antigen SARS-CoV-2 nucleoprotein (N) or SARS-CoV-2 variant spike (S) proteins of beta, delta, gamma, and omicron variants against homologous and heterologous challenges in hamsters. A single intranasal dose of CVXGA1 induces neutralizing antibodies against SARS-CoV-2 WA1 (ancestral), delta variant, and omicron variant and protects against both homologous and heterologous virus challenges. Compared to mRNA COVID-19 vaccine, neutralizing antibody titers induced by CVXGA1 were well-maintained over time. When administered as a boost following two doses of a mRNA COVID-19 vaccine, PIV5-vectored vaccines expressing the S protein from WA1 (CVXGA1), delta, or omicron variants generate higher levels of cross-reactive neutralizing antibodies compared to three doses of a mRNA vaccine. In addition to the S protein, the N protein provides added protection as assessed by the highest body weight gain post-challenge infection. Our data indicates that PIV5-vectored COVID-19 vaccines, such as CVXGA1, can serve as booster vaccines against emerging variants.

19.
Sci Total Environ ; 780: 146632, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34030314

RESUMEN

Membrane fouling is an intractable issue in wastewater treatment by nanofiltration (NF) membrane, and alkaline cleaning is the most effective approach to remove organic fouling on NF membrane. However, it was found that pore swelling of NF membrane induced by alkaline cleaning might reduce cleaning efficiency, and it is never quantified and its effect on membrane fouling behavior is still mysterious. In this work, membrane pore swelling effect (~9.7%, increment of effective pore size) induced by alkaline cleaning (pH 11) is confirmed and its effect on fouling behavior of the polyamide NF membrane is investigated based on experimental and modelling results. It is found that the alkali-induced pore swelling phenomenon would disappear after water filtration at neutral pH for 30 min, and if such cleaned membrane is faced by the small foulants during this pore shrinkage period, the concentration polarization and membrane fouling would be severer, and the subsequent alkaline cleaning is less effective because more foulants enter the enlarged pores and are tightly embedded in the membrane. Thus, the irreversible fouling of the NF membrane increases from 20% to 40% while its permeability recovery declines from 100% to 67% after six fouling/cleaning cycles. When an anionic surfactant sodium dodecyl sulfate (SDS, 10 mM) is added in the alkaline cleaning solution, the adsorption of SDS in/on the membrane can not only improve its hydrophilicity and negative charge, but also quickly eliminate the alkali-induced pore swelling effect and avoid the accumulation of foulants in the pores, thereby enhancing the antifouling performance of the NF membrane. Using the alkaline SDS cleaning, the irreversible fouling of the NF membrane maintains below 10% while its permeability recovery keeps above 100% in six continuous fouling/cleaning cycles.

20.
Pain Physician ; 24(8): 495-506, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34793634

RESUMEN

BACKGROUND: Chronic musculoskeletal pain (CMP) management is a major global public health goal owing to increased social and economic burdens. However, the risk of CMP in smokers compared with nonsmokers remains uncertain. OBJECTIVES: This study aims to determine the magnitude and importance of the relationship between cigarette smoking and risk of CMP. STUDY DESIGN: A meta-analysis of the CMP risk of cigarette smokers. METHODS: We systematically searched PubMed, Embase, and Cochrane library databases from inception to August 2020. Data extraction and quality assessment were performed by 2 independent reviewers using a standardized extraction checklist. Data were pooled using a random-effects model. RESULTS: In this meta-analysis of 32 studies involving 296,109 participants, current smoking was associated with increased CMP risk (OR: 1.23, 95% CI: 1.09-1.40), whereas ever and past smoking did not show such an association (OR: 1.14, 95% CI: 0.95-1.37; OR: 1.06, 95% CI: 0.83-1.35, respectively). Stratified analyses showed that there was a marked significance in almost all strata of current smokers compared with non-smokers, except for mean age (>= 50 years), location of pain (neck pain, sacral pain, and knee pain), smoking frequency (occasionally), study design (cross-sectional), mean follow-up (< 10 years), and adjustment for confounding factors (>= 6). Interestingly, there was statistically negative association between cigarette smoking and knee pain risk in current smokers, ever smokers, and past smokers. LIMITATIONS: The major limitation of this meta-analysis relates to the heterogeneities across included studies. CONCLUSIONS: Cigarette smoking was associated with increased risk of CMP. In view of the high prevalence of smoking in many countries and the increasing number of CMP patients worldwide, reducing tobacco use should be an important public health strategy to prevent and control the global epidemic of CMP. Future research should attempt to establish whether this association is causal and clarify its mechanisms.


Asunto(s)
Fumar Cigarrillos , Sistemas Electrónicos de Liberación de Nicotina , Dolor Musculoesquelético , Fumar Cigarrillos/efectos adversos , Estudios Transversales , Humanos , Persona de Mediana Edad , Dolor Musculoesquelético/epidemiología , Dolor Musculoesquelético/etiología , Factores de Riesgo , Fumar/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA