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










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38343806

RESUMEN

Adenosine A2A receptors (A2AAR) evoke pleiotropic intracellular signaling events via activation of the stimulatory heterotrimeric G protein, Gs. Here, we used cryoEM to solve the agonist-bound structure of A2AAR in a complex with full-length Gs α and Gß4γ2 (A2AAR-Gs α:ß4γ2). The orthosteric binding site of A2AAR-Gs α:ß4γ2 was similar to other structures of agonist-bound A2AAR, with or without Gs. Unexpectedly, the solvent accessible surface area within the interior of the complex was substantially larger for the complex with Gß4 versus the closest analog, A2AAR-miniGs α:ß1γ2. Consequently, there are fewer interactions between the switch II in Gs α and the Gß4 torus. In reconstitution experiments Gß4γ2 displayed a ten-fold higher efficiency over Gß1γ2 in catalyzing A2AAR dependent GTPγS binding to Gs α. We propose that the less constrained switch II in A2AAR-Gs α:ß4γ2 accounts for this increased efficiency. These results suggest that Gß4 functions as a positive allosteric enhancer versus Gß1.

2.
bioRxiv ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-37961136

RESUMEN

Circulating sexual stages of Plasmodium falciparum (Pf) can be transmitted from humans to mosquitoes, thereby furthering the spread of malaria in the population. It is well established that antibodies (Abs) can efficiently block parasite transmission. In search for naturally acquired Ab targets on sexual stages, we established an efficient method for target-agnostic single B cell activation followed by high-throughput selection of human monoclonal antibodies (mAbs) reactive to sexual stages of Pf in the form of gamete and gametocyte extract. We isolated mAbs reactive against a range of Pf proteins including well-established targets Pfs48/45 and Pfs230. One mAb, B1E11K, was cross-reactive to various proteins containing glutamate-rich repetitive elements expressed at different stages of the parasite life cycle. A crystal structure of two B1E11K Fab domains in complex with its main antigen, RESA, expressed on asexual blood stages, showed binding of B1E11K to a repeating epitope motif in a head-to-head conformation engaging in affinity-matured homotypic interactions. Thus, this mode of recognition of Pf proteins, previously described only for PfCSP, extends to other repeats expressed across various stages. The findings augment our understanding of immune-pathogen interactions to repeating elements of the Plasmodium parasite proteome and underscore the potential of the novel mAb identification method used to provide new insights into the natural humoral immune response against Pf . Impact Statement: A naturally acquired human monoclonal antibody recognizes proteins expressed at different stages of the Plasmodium falciparum lifecycle through affinity-matured homotypic interactions with glutamate-rich repeats.

3.
Sci Transl Med ; 15(697): eadf4549, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37224226

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has been responsible for a global pandemic. Monoclonal antibodies (mAbs) have been used as antiviral therapeutics; however, these therapeutics have been limited in efficacy by viral sequence variability in emerging variants of concern (VOCs) and in deployment by the need for high doses. In this study, we leveraged the multi-specific, multi-affinity antibody (Multabody, MB) platform, derived from the human apoferritin protomer, to enable the multimerization of antibody fragments. MBs were shown to be highly potent, neutralizing SARS-CoV-2 at lower concentrations than their corresponding mAb counterparts. In mice infected with SARS-CoV-2, a tri-specific MB targeting three regions within the SARS-CoV-2 receptor binding domain was protective at a 30-fold lower dose than a cocktail of the corresponding mAbs. Furthermore, we showed in vitro that mono-specific MBs potently neutralize SARS-CoV-2 VOCs by leveraging augmented avidity, even when corresponding mAbs lose their ability to neutralize potently, and that tri-specific MBs expanded the neutralization breadth beyond SARS-CoV-2 to other sarbecoviruses. Our work demonstrates how avidity and multi-specificity combined can be leveraged to confer protection and resilience against viral diversity that exceeds that of traditional monoclonal antibody therapies.


Asunto(s)
COVID-19 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Humanos , Animales , Ratones , SARS-CoV-2 , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antivirales
4.
Cell Rep ; 42(4): 112391, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37053069

RESUMEN

Subunit vaccines typically require co-administration with an adjuvant to elicit protective immunity, adding development hurdles that can impede rapid pandemic responses. To circumvent the need for adjuvant in a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) subunit vaccine, we engineer a thermostable immunotargeting vaccine (ITV) that leverages the pan-HLA-DR monoclonal antibody 44H10 to deliver the viral spike protein receptor-binding domain (RBD) to antigen-presenting cells. X-ray crystallography shows that 44H10 binds to a conserved epitope on HLA-DR, providing the basis for its broad HLA-DR reactivity. Adjuvant-free ITV immunization in rabbits and ferrets induces robust anti-RBD antibody responses that neutralize SARS-CoV-2 variants of concern and protect recipients from SARS-CoV-2 challenge. We demonstrate that the modular nature of the ITV scaffold with respect to helper T cell epitopes and diverse RBD antigens facilitates broad sarbecovirus neutralization. Our findings support anti-HLA-DR immunotargeting as an effective means to induce strong antibody responses to subunit antigens without requiring an adjuvant.


Asunto(s)
COVID-19 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Animales , Humanos , Conejos , SARS-CoV-2 , Vacunas contra la COVID-19 , Anticuerpos Antivirales , Anticuerpos ampliamente neutralizantes , COVID-19/prevención & control , Hurones , Adyuvantes Inmunológicos , Receptores Virales/metabolismo , Antígenos HLA-DR , Vacunas de Subunidad , Anticuerpos Neutralizantes
5.
Nat Struct Mol Biol ; 30(3): 383-390, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36759579

RESUMEN

The HIV-1 capsid is a fullerene cone made of quasi-equivalent hexamers and pentamers of the viral CA protein. Typically, quasi-equivalent assembly of viral capsid subunits is controlled by a molecular switch. Here, we identify a Thr-Val-Gly-Gly motif that modulates CA hexamer/pentamer switching by folding into a 310 helix in the pentamer and random coil in the hexamer. Manipulating the coil/helix configuration of the motif allowed us to control pentamer and hexamer formation in a predictable manner, thus proving its function as a molecular switch. Importantly, the switch also remodels the common binding site for host factors that are critical for viral replication and the new ultra-potent HIV-1 inhibitor lenacapavir. This study reveals that a critical assembly element also modulates the post-assembly and viral replication functions of the HIV-1 capsid and provides new insights on capsid function and inhibition.


Asunto(s)
Cápside , VIH-1 , Cápside/química , VIH-1/química , Proteínas de la Cápside/química
6.
PLoS Pathog ; 18(11): e1010999, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36441829

RESUMEN

Antibodies targeting the human malaria parasite Plasmodium falciparum circumsporozoite protein (PfCSP) can prevent infection and disease. PfCSP contains multiple central repeating NANP motifs; some of the most potent anti-infective antibodies against malaria bind to these repeats. Multiple antibodies can bind the repeating epitopes concurrently by engaging into homotypic Fab-Fab interactions, which results in the ordering of the otherwise largely disordered central repeat into a spiral. Here, we characterize IGHV3-33/IGKV1-5-encoded monoclonal antibody (mAb) 850 elicited by immunization of transgenic mice with human immunoglobulin loci. mAb 850 binds repeating NANP motifs with picomolar affinity, potently inhibits Plasmodium falciparum (Pf) in vitro and, when passively administered in a mouse challenge model, reduces liver burden to a similar extent as some of the most potent anti-PfCSP mAbs yet described. Like other IGHV3-33/IGKV1-5-encoded anti-NANP antibodies, mAb 850 primarily utilizes its HCDR3 and germline-encoded aromatic residues to recognize its core NANP motif. Biophysical and cryo-electron microscopy analyses reveal that up to 19 copies of Fab 850 can bind the PfCSP repeat simultaneously, and extensive homotypic interactions are observed between densely-packed PfCSP-bound Fabs to indirectly improve affinity to the antigen. Together, our study expands on the molecular understanding of repeat-induced homotypic interactions in the B cell response against PfCSP for potently protective mAbs against Pf infection.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Humanos , Ratones , Animales , Plasmodium falciparum , Microscopía por Crioelectrón , Malaria Falciparum/parasitología , Proteínas Protozoarias , Malaria/parasitología , Ratones Transgénicos , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios
7.
Immunity ; 55(9): 1680-1692.e8, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35977542

RESUMEN

Malaria transmission-blocking vaccines (TBVs) aim to elicit human antibodies that inhibit sporogonic development of Plasmodium falciparum in mosquitoes, thereby preventing onward transmission. Pfs48/45 is a leading clinical TBV candidate antigen and is recognized by the most potent transmission-blocking monoclonal antibody (mAb) yet described; still, clinical development of Pfs48/45 antigens has been hindered, largely by its poor biochemical characteristics. Here, we used structure-based computational approaches to design Pfs48/45 antigens stabilized in the conformation recognized by the most potently inhibitory mAb, achieving >25°C higher thermostability compared with the wild-type protein. Antibodies elicited in mice immunized with these engineered antigens displayed on liposome-based or protein nanoparticle-based vaccine platforms exhibited 1-2 orders of magnitude superior transmission-reducing activity, compared with immunogens bearing the wild-type antigen, driven by improved antibody quality. Our data provide the founding principles for using molecular stabilization solely from antibody structure-function information to drive improved immune responses against a parasitic vaccine target.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Animales , Anticuerpos Bloqueadores , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios , Formación de Anticuerpos , Antígenos de Protozoos , Humanos , Malaria Falciparum/prevención & control , Glicoproteínas de Membrana , Ratones , Plasmodium falciparum , Proteínas Protozoarias , Vacunación
8.
Elife ; 112022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35023832

RESUMEN

Malaria is a global health burden, with Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) responsible for the majority of infections worldwide. Circumsporozoite protein (CSP) is the most abundant protein on the surface of Plasmodium sporozoites, and antibodies targeting the central repeat region of CSP can prevent parasite infection. Although much has been uncovered about the molecular basis of antibody recognition of the PfCSP repeats, data remains scarce for PvCSP. Here, we performed molecular dynamics simulations for peptides comprising the PvCSP repeats from strains VK210 and VK247 to reveal how the PvCSP central repeats are highly disordered, with minor propensities to adopt turn conformations. Next, we solved eight crystal structures to unveil the interactions of two inhibitory monoclonal antibodies (mAbs), 2F2 and 2E10.E9, with PvCSP repeats. Both antibodies can accommodate subtle sequence variances in the repeat motifs and recognize largely coiled peptide conformations that also contain isolated turns. Our structural studies uncover various degrees of Fab-Fab homotypic interactions upon recognition of the PvCSP central repeats by these two inhibitory mAbs, similar to potent mAbs against PfCSP. These findings augment our understanding of host-Plasmodium interactions and contribute molecular details of Pv inhibition by mAbs to unlock structure-based engineering of PvCSP-based vaccines.


Asunto(s)
Anticuerpos Antiprotozoarios/metabolismo , Sitios de Unión de Anticuerpos , Simulación de Dinámica Molecular , Plasmodium vivax/inmunología , Plasmodium vivax/metabolismo , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Animales , Anticuerpos Antiprotozoarios/química , Línea Celular , Cristalización , Células HEK293 , Humanos , Ratones , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Esporozoítos/metabolismo
9.
J Biol Chem ; 297(2): 100966, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34273351

RESUMEN

Cluster of differentiation-22 (CD22) belongs to the sialic acid-binding immunoglobulin (Ig)-like lectin family of receptors that is expressed on the surface of B cells. It has been classified as an inhibitory coreceptor for the B-cell receptor because of its function in establishing a baseline level of B-cell inhibition. The restricted expression of CD22 on B cells and its inhibitory function make it an attractive target for B-cell depletion in cases of B-cell malignancies. Genetically modified T cells with chimeric antigen receptors (CARs) derived from the m971 antibody have shown promise when used as an immunotherapeutic agent against B-cell acute lymphoblastic leukemia. A key aspect of the efficacy of this CAR-T was its ability to target a membrane-proximal epitope on the CD22 extracellular domain; however, the molecular details of m971 recognition of CD22 have thus far remained elusive. Here, we report the crystal structure of the m971 fragment antigen-binding in complex with the two most membrane-proximal Ig-like domains of CD22 (CD22d6-d7). The m971 epitope on CD22 resides at the most proximal Ig domain (d7) to the membrane, and the antibody paratope contains electrostatic surfaces compatible with interactions with phospholipid head groups. Together, our data identify molecular details underlying the successful transformation of an antibody epitope on CD22 into an effective CAR immunotherapeutic target.


Asunto(s)
Anticuerpos Monoclonales , Antígenos CD19 , Lectina 2 Similar a Ig de Unión al Ácido Siálico/química , Antígenos CD19/inmunología , Linfocitos B/metabolismo , Dominios Proteicos
10.
Nat Commun ; 12(1): 3661, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135340

RESUMEN

SARS-CoV-2, the virus responsible for COVID-19, has caused a global pandemic. Antibodies can be powerful biotherapeutics to fight viral infections. Here, we use the human apoferritin protomer as a modular subunit to drive oligomerization of antibody fragments and transform antibodies targeting SARS-CoV-2 into exceptionally potent neutralizers. Using this platform, half-maximal inhibitory concentration (IC50) values as low as 9 × 10-14 M are achieved as a result of up to 10,000-fold potency enhancements compared to corresponding IgGs. Combination of three different antibody specificities and the fragment crystallizable (Fc) domain on a single multivalent molecule conferred the ability to overcome viral sequence variability together with outstanding potency and IgG-like bioavailability. The MULTi-specific, multi-Affinity antiBODY (Multabody or MB) platform thus uniquely leverages binding avidity together with multi-specificity to deliver ultrapotent and broad neutralizers against SARS-CoV-2. The modularity of the platform also makes it relevant for rapid evaluation against other infectious diseases of global health importance. Neutralizing antibodies are a promising therapeutic for SARS-CoV-2.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , SARS-CoV-2/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Apoferritinas/química , Disponibilidad Biológica , Mapeo Epitopo , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ingeniería de Proteínas/métodos , Subunidades de Proteína/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Distribución Tisular
11.
Elife ; 92020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33253113

RESUMEN

Plasmodium sporozoites express circumsporozoite protein (CSP) on their surface, an essential protein that contains central repeating motifs. Antibodies targeting this region can neutralize infection, and the partial efficacy of RTS,S/AS01 - the leading malaria vaccine against P. falciparum (Pf) - has been associated with the humoral response against the repeats. Although structural details of antibody recognition of PfCSP have recently emerged, the molecular basis of antibody-mediated inhibition of other Plasmodium species via CSP binding remains unclear. Here, we analyze the structure and molecular interactions of potent monoclonal antibody (mAb) 3D11 binding to P. berghei CSP (PbCSP) using molecular dynamics simulations, X-ray crystallography, and cryoEM. We reveal that mAb 3D11 can accommodate all subtle variances of the PbCSP repeating motifs, and, upon binding, induces structural ordering of PbCSP through homotypic interactions. Together, our findings uncover common mechanisms of antibody evolution in mammals against the CSP repeats of Plasmodium sporozoites.


Malaria is a significant health concern, killing about 400,000 people each year. While antimalarial drugs and insecticides have successfully reduced deaths over the last 20 years, the parasite that causes malaria is starting to gain resistance to these treatments. Vaccines offer an alternative route to preventing the disease. However, the most advanced vaccine currently available provides less than 50% protection. Vaccines work by encouraging the body to develop proteins called antibodies, which can recognize the parasite and trigger an immune response that blocks the infection. These antibodies target a molecule on the parasite's surface called circumsporozoite protein, or CSP for short. Therefore, having a better understanding of how antibodies interact with CSP could help researchers design more effective treatments. A lot of what is known about malaria has come from studying this disease in mice. However, it remained unclear whether antibodies produced in rodents combat the malaria-causing parasite in a similar manner to human antibodies. To answer this question, Kucharska, Thai et al. studied a mouse antibody called 3D11, which targets CSP on the surface of a parasite that causes malaria in rodents. The interaction between CSP and 3D11 was studied using three different techniques in order to better understand how the structure of CSP changes when bound by antibodies. The experiments showed that although CSP has a highly flexible structure, it forms a more stable, spiral-like architecture when bound to multiple copies of 3D11. A similar type of assembly was previously observed in studies investigating how CSP interacts with human antibodies. Further investigation revealed that the molecular connections between 3D11 and CSP share a lot of similarities with how human antibodies recognize CSP. These findings reveal how mammals evolved similar mechanisms for detecting and inhibiting malaria-causing parasites. This highlights the robust features antibodies need to launch an immune response against malaria, which could help develop a more effective vaccine.


Asunto(s)
Plasmodium berghei/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales , Sitios de Unión , Microscopía por Crioelectrón , Regulación de la Expresión Génica , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Proteínas Protozoarias/genética
12.
Cell ; 182(2): 345-356.e16, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32589945

RESUMEN

Pathogenic clostridial species secrete potent toxins that induce severe host tissue damage. Paeniclostridium sordellii lethal toxin (TcsL) causes an almost invariably lethal toxic shock syndrome associated with gynecological infections. TcsL is 87% similar to C. difficile TcdB, which enters host cells via Frizzled receptors in colon epithelium. However, P. sordellii infections target vascular endothelium, suggesting that TcsL exploits another receptor. Here, using CRISPR/Cas9 screening, we establish semaphorins SEMA6A and SEMA6B as TcsL receptors. We demonstrate that recombinant SEMA6A can protect mice from TcsL-induced edema. A 3.3 Å cryo-EM structure shows that TcsL binds SEMA6A with the same region that in TcdB binds structurally unrelated Frizzled. Remarkably, 15 mutations in this evolutionarily divergent surface are sufficient to switch binding specificity of TcsL to that of TcdB. Our findings establish semaphorins as physiologically relevant receptors for TcsL and reveal the molecular basis for the difference in tissue targeting and disease pathogenesis between highly related toxins.


Asunto(s)
Toxinas Bacterianas/metabolismo , Clostridium sordellii/metabolismo , Semaforinas/metabolismo , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Sitios de Unión , Sistemas CRISPR-Cas/genética , Línea Celular , Microscopía por Crioelectrón , Edema/patología , Edema/prevención & control , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/uso terapéutico , Semaforinas/química , Semaforinas/genética
13.
Science ; 368(6492)2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32409444

RESUMEN

De novo protein design has been successful in expanding the natural protein repertoire. However, most de novo proteins lack biological function, presenting a major methodological challenge. In vaccinology, the induction of precise antibody responses remains a cornerstone for next-generation vaccines. Here, we present a protein design algorithm called TopoBuilder, with which we engineered epitope-focused immunogens displaying complex structural motifs. In both mice and nonhuman primates, cocktails of three de novo-designed immunogens induced robust neutralizing responses against the respiratory syncytial virus. Furthermore, the immunogens refocused preexisting antibody responses toward defined neutralization epitopes. Overall, our design approach opens the possibility of targeting specific epitopes for the development of vaccines and therapeutic antibodies and, more generally, will be applicable to the design of de novo proteins displaying complex functional motifs.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Biología Computacional/métodos , Epítopos Inmunodominantes/química , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/química , Vacunas contra Virus Sincitial Respiratorio/química , Virus Sincitial Respiratorio Humano/inmunología , Secuencias de Aminoácidos , Humanos , Epítopos Inmunodominantes/inmunología , Conformación Proteica , Proteínas Recombinantes de Fusión/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología
14.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31801870

RESUMEN

The assembly of an orthoretrovirus such as HIV-1 requires the coordinated functioning of multiple biochemical activities of the viral Gag protein. These activities include membrane targeting, lattice formation, packaging of the RNA genome, and recruitment of cellular cofactors that modulate assembly. In most previous studies, these Gag activities have been investigated individually, which provided somewhat limited insight into how they functionally integrate during the assembly process. Here, we report the development of a biochemical reconstitution system that allowed us to investigate how Gag lattice formation, RNA binding, and the assembly cofactor inositol hexakisphosphate (IP6) synergize to generate immature virus particles in vitro The results identify an important rate-limiting step in assembly and reveal new insights into how RNA and IP6 promote immature Gag lattice formation. The immature virus-like particles can be converted into mature capsid-like particles by the simple addition of viral protease, suggesting that it is possible in principle to fully biochemically reconstitute the sequential processes of HIV-1 assembly and maturation from purified components.IMPORTANCE Assembly and maturation are essential steps in the replication of orthoretroviruses such as HIV-1 and are proven therapeutic targets. These processes require the coordinated functioning of the viral Gag protein's multiple biochemical activities. We describe here the development of an experimental system that allows an integrative analysis of how Gag's multiple functionalities cooperate to generate a retrovirus particle. Our current studies help to illuminate how Gag synergizes the formation of the virus compartment with RNA binding and how these activities are modulated by the small molecule IP6. Further development and use of this system should lead to a more comprehensive understanding of the molecular mechanisms of HIV-1 assembly and maturation and may provide new insights for the development of antiretroviral drugs.


Asunto(s)
VIH-1/genética , VIH-1/fisiología , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Cápside/metabolismo , Humanos , Microscopía Electrónica , Modelos Moleculares , Ácido Fítico , Virión/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
16.
Biophys J ; 112(2): 346-355, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-28122220

RESUMEN

The outer membrane (OM) of Gram-negative bacteria is composed of lipopolysaccharide (LPS) in the outer leaflet and phospholipids in the inner leaflet. The outer membrane protein H (OprH) of Pseudomonas aeruginosa provides an increased stability to the OMs by directly interacting with LPS. Here we report the influence of various P. aeruginosa and, for comparison, Escherichia coli LPS environments on the physical properties of the OMs and OprH using all-atom molecular dynamics simulations. The simulations reveal that although the P. aeruginosa OMs are thinner hydrophobic bilayers than the E. coli OMs, which is expected from the difference in the acyl chain length of their lipid A, this effect is almost imperceptible around OprH due to a dynamically adjusted hydrophobic match between OprH and the OM. The structure and dynamics of the extracellular loops of OprH show distinct behaviors in different LPS environments. Including the O-antigen greatly reduces the flexibility of the OprH loops and increases the interactions between these loops and LPS. Furthermore, our study shows that the interactions between OprH and LPS mainly depend on the secondary structure of OprH and the chemical structure of LPS, resulting in distinctive patterns in different LPS environments.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Lipopolisacáridos/metabolismo , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Membrana Celular/metabolismo , Farmacorresistencia Microbiana , Escherichia coli/citología , Unión Proteica , Conformación Proteica , Pseudomonas aeruginosa/citología , Pseudomonas aeruginosa/efectos de los fármacos , Agua/química
17.
Biochemistry ; 55(36): 5061-72, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27532487

RESUMEN

Pseudomonas aeruginosa is an opportunistic human pathogen causing pneumonias that are particularly severe in cystic fibrosis and immunocompromised patients. The outer membrane (OM) of P. aeruginosa is much less permeable to nutrients and other chemical compounds than that of Escherichia coli. The low permeability of the OM, which also contributes to Pseudomonas' significant antibiotic resistance, is augmented by the presence of the outer membrane protein H (OprH). OprH directly interacts with lipopolysaccharides (LPS) that constitute the outer leaflet of the OM and thus contributes to the structural stability of the OM. In this study, we used solution NMR spectroscopy to characterize the interactions between LPS and OprH in molecular detail. NMR chemical shift perturbations observed upon the addition of LPS to OprH in DHPC micelles indicate that this interaction is predominantly electrostatic and localized to the extracellular loops 2 and 3 and a number of highly conserved basic residues near the extracellular barrel rim of OprH. Single-site mutations of these residues were not enough to completely abolish binding, but OprH with cumulative mutations of Lys70, Arg72, and Lys103 no longer binds LPS. The dissociation constant (∼200 µM) measured by NMR is sufficient to efficiently bind LPS to OprH in the OM. This work highlights that solution NMR is suitable to study specific interactions of lipids with integral membrane proteins and provides a detailed molecular model for the interaction of LPS with OprH; i.e., an interaction that contributes to the integrity of the OM of P. aeruginosa under low divalent cation and antibiotic stress conditions. These methods should thus be useful for screening antibiotics that might disrupt OprH-LPS interactions and thereby increase the permeability of the OM of P. aeruginosa.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Lipopolisacáridos/metabolismo , Resonancia Magnética Nuclear Biomolecular/métodos , Pseudomonas aeruginosa/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Ensayo de Inmunoadsorción Enzimática , Mutagénesis Sitio-Dirigida , Pliegue de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
Sci Rep ; 6: 26847, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27229326

RESUMEN

Protein engineering is an important tool for the design of proteins with novel and desirable features. Templates from the protein databank (PDB) are often used as initial models that can be modified to introduce new properties. We examine whether it is possible to reconnect a protein in a manner that generates a new topology yet preserves its structural integrity. Here, we describe the rethreading of dihydrofolate reductase (DHFR) from E. coli (wtDHFR). The rethreading process involved the removal of three native loops, and the introduction of three new loops with alternate connections. The structure of the rethreaded DHFR (rDHFR-1) was determined to 1.6 Å, demonstrating the success of the rethreading process. Both wtDHFR and rDHFR-1 exhibited similar affinities towards methotrexate. However, rDHFR-1 showed no reducing activity towards dihydrofolate, and exhibited about ~6-fold lower affinity towards NADPH than wtDHFR. This work demonstrates that protein rethreading can be a powerful tool for the design of a large array of proteins with novel structures and topologies, and that by careful rearrangement of a protein sequence, the sequence to structure relationship can be expanded substantially.


Asunto(s)
Proteínas Bacterianas/genética , Ingeniería de Proteínas/métodos , Tetrahidrofolato Deshidrogenasa/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Bases de Datos Factuales , Escherichia coli , Modelos Moleculares , Estructura Terciaria de Proteína , Tetrahidrofolato Deshidrogenasa/química
19.
Structure ; 23(12): 2234-2245, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26655471

RESUMEN

OprG is an outer membrane protein of Pseudomonas aeruginosa whose function as an antibiotic-sensitive porin has been controversial and not well defined. Circumstantial evidence led to the proposal that OprG might transport hydrophobic compounds by using a lateral gate in the barrel wall thought to be lined by three conserved prolines. To test this hypothesis and to find the physiological substrates of OprG, we reconstituted the purified protein into liposomes and found it to facilitate the transport of small amino acids such as glycine, alanine, valine, and serine, which was confirmed by Pseudomonas growth assays. The structures of wild-type and a critical proline mutant were determined by nuclear magnetic resonance in dihexanoyl-phosphatidylcholine micellar solutions. Both proteins formed eight-stranded ß-barrels with flexible extracellular loops. The interfacial prolines did not form a lateral gate in these structures, but loop 3 exhibited restricted motions in the inactive P92A mutant but not in wild-type OprG.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Secuencia Conservada , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína
20.
J Biomol NMR ; 61(3-4): 261-74, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25869397

RESUMEN

Solution NMR spectroscopy has become a robust method to determine structures and explore the dynamics of integral membrane proteins. The vast majority of previous studies on membrane proteins by solution NMR have been conducted in lipid micelles. Contrary to the lipids that form a lipid bilayer in biological membranes, micellar lipids typically contain only a single hydrocarbon chain or two chains that are too short to form a bilayer. Therefore, there is a need to explore alternative more bilayer-like media to mimic the natural environment of membrane proteins. Lipid bicelles and lipid nanodiscs have emerged as two alternative membrane mimetics that are compatible with solution NMR spectroscopy. Here, we have conducted a comprehensive comparison of the physical and spectroscopic behavior of two outer membrane proteins from Pseudomonas aeruginosa, OprG and OprH, in lipid micelles, bicelles, and nanodiscs of five different sizes. Bicelles stabilized with a fraction of negatively charged lipids yielded spectra of almost comparable quality as in the best micellar solutions and the secondary structures were found to be almost indistinguishable in the two environments. Of the five nanodiscs tested, nanodiscs assembled from MSP1D1ΔH5 performed the best with both proteins in terms of sample stability and spectral resolution. Even in these optimal nanodiscs some broad signals from the membrane embedded barrel were severely overlapped with sharp signals from the flexible loops making their assignments difficult. A mutant OprH that had two of the flexible loops truncated yielded very promising spectra for further structural and dynamical analysis in MSP1D1ΔH5 nanodiscs.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Micelas , Resonancia Magnética Nuclear Biomolecular/métodos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Clonación Molecular , Membrana Dobles de Lípidos/química , Modelos Moleculares , Fosfolípidos/química , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...