Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Transplant Direct ; 9(9): e1526, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37654682

ABSTRACT

Background: Delayed graft function (DGF) among deceased donor kidney transplant recipients (DDKTRs) is a well-known risk factor for allograft rejection, decreased graft survival, and increased cost. Although DGF is associated with an increased risk of rejection, it is unclear whether it also increases the risk of infection. Methods: We reviewed all adult DDKTRs at our center between 2010 and 2018. The primary outcomes of interest were BK viremia, cytomegalovirus viremia, pneumonia, and urinary tract infection (UTI) within the first year of transplant. Additional analysis was made with censoring follow-up at the time of allograft rejection. Results: A total of 1512 DDKTRs were included, of whom 468 (31%) had DGF. As expected, several recipient, donor, and baseline immunological characteristics differed by DGF status. After adjustment, DGF was significantly associated with an increased risk of BK viremia (hazard ratio: 1.34; 95% confidence interval, 1.0-1.81; P = 0.049) and UTI (hazard ratio: 1.70; 95% confidence interval, 1.31-2.19; P < 0.001) but not cytomegalovirus viremia or pneumonia. Associations were similar in models censored at the time of rejection. Conclusions: DGF is associated with an increased risk of early infectious complications, mainly UTI and BK viremia. Close monitoring and appropriate management are warranted for better outcomes in this unique population.

2.
NPJ Vaccines ; 7(1): 74, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35773301

ABSTRACT

Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.

3.
Transplant Direct ; 7(10): e764, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34514119

ABSTRACT

The clinical significance and appropriate management of graft pyelonephritis diagnosed by biopsy are poorly understood. METHODS: We analyzed data from all patients with pyelonephritis on transplant kidney biopsy from January 1998 to December 2019. Patients were divided into 2 groups: those whose urinalysis was positive for urinary tract infection (UA+) and those whose urinalysis was negative (UA-). RESULTS: There were a total of 101 patients with the diagnosis of pyelonephritis by biopsy during the study period. The mean time from transplant to pyelonephritis diagnosis was 3.3 ± 4 y. Thirty-six (35.6%) of the patients with pyelonephritis on biopsy had a negative UA. Out of 65 patients in the UA+ group, 54 (83%) received antibiotics. Only 12 of the UA- patients (33%) received antibiotics. The use of antibiotics in both the UA+ (P = 0.03) and UA- groups (P = 0.02) compared with no use of antibiotics was associated with better death-censored graft survival. On multivariate analysis, the use of antibiotics (hazard ratio = 0.22, P = 0.001, 95% confidence interval, 0.12-0.61) was associated with improved graft survival. CONCLUSION: The finding of pyelonephritis on a transplant kidney biopsy is almost always a surprise but is an important finding. Treatment with antibiotics, regardless of signs or symptoms of urinary tract infection, is associated with improved graft survival.

4.
Virology ; 550: 21-26, 2020 11.
Article in English | MEDLINE | ID: mdl-32866728

ABSTRACT

Human respiratory syncytial virus (RSV) is a major cause of serious respiratory tract infections in infants and the elderly. Recently it was shown that the RSV G glycoprotein mediates attachment to cells using CX3CR1 as a receptor, and that G-specific neutralizing antibodies can be detected using human airway epithelial (HAE) cell cultures. To investigate the contributions of G-specific antibodies to RSV neutralization, we performed HAE neutralization assays on sera from RSV G-immunized mice or RSV-infected infants. We confirmed that G-specific neutralization using serum from mice or humans could only be detected on HAE cultures. We also found that RSV G-specific antibodies in infants were either subgroup specific or cross-neutralizing. Altogether, our results suggest that G is an important target for generating neutralizing antibodies and would be beneficial to include in an RSV vaccine. Further, inclusion of G antigens from both RSV subgroups may enhance the vaccine cross protection potency.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/immunology , CX3C Chemokine Receptor 1/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , CX3C Chemokine Receptor 1/genetics , Chlorocebus aethiops , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Gene Expression , Humans , Immune Sera/chemistry , Immunization , Mice , Mice, Inbred BALB C , Models, Biological , Neutralization Tests , Protein Binding , Receptors, Virus/genetics , Receptors, Virus/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/pathogenicity , Vero Cells , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics
5.
Sci Immunol ; 5(47)2020 05 01.
Article in English | MEDLINE | ID: mdl-32358170

ABSTRACT

A stabilized form of the respiratory syncytial virus (RSV) fusion (F) protein has been explored as a vaccine to prevent viral infection because it presents several potent neutralizing epitopes. Here, we used a structure-based rational design to optimize antigen presentation and focus antibody (Ab) responses to key epitopes on the pre-fusion (pre-F) protein. This protein was fused to ferritin nanoparticles (pre-F-NP) and modified with glycans to mask nonneutralizing or poorly neutralizing epitopes to further focus the Ab response. The multimeric pre-F-NP elicited durable pre-F-specific Abs in nonhuman primates (NHPs) after >150 days and elicited potent neutralizing Ab (NAb) responses in mice and NHPs in vivo, as well as in human cells evaluated in the in vitro MIMIC system. This optimized pre-F-NP stimulated a more potent Ab response than a representative pre-F trimer, DS-Cav1. Collectively, this pre-F vaccine increased the generation of NAbs targeting the desired pre-F conformation, an attribute that facilitates the development of an effective RSV vaccine.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Nanoparticles/chemistry , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/chemistry , Viral Fusion Proteins/immunology , Animals , Antibody Formation , Antigens, Viral/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/chemistry
6.
NPJ Vaccines ; 5(1): 33, 2020.
Article in English | MEDLINE | ID: mdl-32377398

ABSTRACT

A growing global health concern, Lyme disease has become the most common tick-borne disease in the United States and Europe. Caused by the bacterial spirochete Borrelia burgdorferi sensu lato (sl), this disease can be debilitating if not treated promptly. Because diagnosis is challenging, prevention remains a priority; however, a previously licensed vaccine is no longer available to the public. Here, we designed a six component vaccine that elicits antibody (Ab) responses against all Borrelia strains that commonly cause Lyme disease in humans. The outer surface protein A (OspA) of Borrelia was fused to a bacterial ferritin to generate self-assembling nanoparticles. OspA-ferritin nanoparticles elicited durable high titer Ab responses to the seven major serotypes in mice and non-human primates at titers higher than a previously licensed vaccine. This response was durable in rhesus macaques for more than 6 months. Vaccination with adjuvanted OspA-ferritin nanoparticles stimulated protective immunity from both B. burgdorferi and B. afzelii infection in a tick-fed murine challenge model. This multivalent Lyme vaccine offers the potential to limit the spread of Lyme disease.

7.
PLoS One ; 14(12): e0226245, 2019.
Article in English | MEDLINE | ID: mdl-31887144

ABSTRACT

Antibody therapies for Alzheimer's Disease (AD) hold promise but have been limited by the inability of these proteins to migrate efficiently across the blood brain barrier (BBB). Central nervous system (CNS) gene transfer by vectors like adeno-associated virus (AAV) overcome this barrier by allowing the bodies' own cells to produce the therapeutic protein, but previous studies using this method to target amyloid-ß have shown success only with truncated single chain antibodies (Abs) lacking an Fc domain. The Fc region mediates effector function and enhances antigen clearance from the brain by neonatal Fc receptor (FcRn)-mediated reverse transcytosis and is therefore desirable to include for such treatments. Here, we show that single chain Abs fused to an Fc domain retaining FcRn binding, but lacking Fc gamma receptor (FcγR) binding, termed a silent scFv-IgG, can be expressed and released into the CNS following gene transfer with AAV. While expression of canonical IgG in the brain led to signs of neurotoxicity, this modified Ab was efficiently secreted from neuronal cells and retained target specificity. Steady state levels in the brain exceeded peak levels obtained by intravenous injection of IgG. AAV-mediated expression of this scFv-IgG reduced cortical and hippocampal plaque load in a transgenic mouse model of progressive ß-amyloid plaque accumulation. These findings suggest that CNS gene delivery of a silent anti-Aß scFv-IgG was well-tolerated, durably expressed and functional in a relevant disease model, demonstrating the potential of this modality for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/therapy , Central Nervous System/metabolism , Genetic Vectors/administration & dosage , Immunoglobulin Fc Fragments/genetics , Single-Chain Antibodies/genetics , Alzheimer Disease/genetics , Animals , Blood-Brain Barrier , Cell Line , Dependovirus/genetics , Disease Models, Animal , Disease Progression , Genetic Therapy , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/metabolism , Mice , Mice, Transgenic , Protein Domains , Receptors, Fc/metabolism , Receptors, IgG/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism
8.
Vaccine ; 37(42): 6208-6220, 2019 09 30.
Article in English | MEDLINE | ID: mdl-31493950

ABSTRACT

Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Neutralizing/immunology , Influenza Vaccines/immunology , Adjuvants, Immunologic/chemistry , Animals , Female , HEK293 Cells , Hemagglutination Inhibition Tests , Hemagglutinins , Humans , Macaca mulatta , Mice, Inbred BALB C , Nanoparticles , Toll-Like Receptors/agonists
9.
J Virol ; 88(20): 11802-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25078705

ABSTRACT

Respiratory syncytial virus (RSV) is the leading infectious cause of severe respiratory disease in infants and a major cause of respiratory illness in the elderly. There remains an unmet vaccine need despite decades of research. Insufficient potency, homogeneity, and stability of previous RSV fusion protein (F) subunit vaccine candidates have hampered vaccine development. RSV F and related parainfluenza virus (PIV) F proteins are cleaved by furin during intracellular maturation, producing disulfide-linked F1 and F2 fragments. During cell entry, the cleaved Fs rearrange from prefusion trimers to postfusion trimers. Using RSV F constructs with mutated furin cleavage sites, we isolated an uncleaved RSV F ectodomain that is predominantly monomeric and requires specific cleavage between F1 and F2 for self-association and rearrangement into stable postfusion trimers. The uncleaved RSV F monomer is folded and homogenous and displays at least two key RSV-neutralizing epitopes shared between the prefusion and postfusion conformations. Unlike the cleaved trimer, the uncleaved monomer binds the prefusion-specific monoclonal antibody D25 and human neutralizing immunoglobulins that do not bind to postfusion F. These observations suggest that the uncleaved RSV F monomer has a prefusion-like conformation and is a potential prefusion subunit vaccine candidate. Importance: RSV is the leading infectious cause of severe respiratory disease in infants and a major cause of respiratory illness in the elderly. Development of an RSV vaccine was stymied when a clinical trial using a formalin-inactivated RSV virus made disease, following RSV infection, more severe. Recent studies have defined the structures that the RSV F envelope glycoprotein adopts before and after virus entry (prefusion and postfusion conformations, respectively). Key neutralization epitopes of prefusion and postfusion RSV F have been identified, and a number of current vaccine development efforts are focused on generating easily produced subunit antigens that retain these epitopes. Here we show that a simple modification in the F ectodomain results in a homogeneous protein that retains critical prefusion neutralizing epitopes. These results improve our understanding of RSV F protein folding and structure and can guide further vaccine design efforts.


Subject(s)
Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Epitopes/immunology , Respiratory Syncytial Viruses/immunology , Humans , Proteolysis
10.
Vaccine ; 31(2): 306-12, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23153444

ABSTRACT

In the 1960s, infant immunization with a formalin-inactivated respiratory syncytial virus (FI-RSV) vaccine candidate caused enhanced respiratory disease (ERD) following natural RSV infection. Because of this tragedy, intensive effort has been made to understand the root causes of how the FI-RSV vaccine induced a pathogenic response to subsequent RSV infection in vaccinees. A well-established cotton rat model of FI-RSV vaccine-enhanced disease has been used by numerous researchers to study the mechanisms of ERD. Here, we have dissected the model and found it to have significant limitations for understanding FI-RSV ERD. This view is shaped by our finding that a major driver of lung pathology is cell-culture contaminants, although FI-RSV immunization and RSV challenge serve as co-factors to exacerbate disease. Specifically, non-viral products from the vaccine and challenge preparations that are devoid of RSV give rise to alveolitis, which is considered a hallmark of FI-RSV ERD in the cotton rat model. Although FI-RSV immunization and RSV challenge promote more severe alveolitis, they also drive stronger cellular immune responses to non-viral antigens. The severity of alveolitis is associated with T cells specific for non-viral antigens more than with T cells specific for RSV. These results highlight the limitations of the cotton rat ERD model and the need for an improved animal model to evaluate the safety of RSV vaccine candidates.


Subject(s)
Antigens/immunology , Lung Diseases/immunology , Lung Diseases/prevention & control , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/immunology , Vaccines/immunology , Animals , Antibodies/immunology , Female , Immunity, Cellular/immunology , Immunization/methods , Lung/immunology , Lung/pathology , Rats , Sigmodontinae
11.
Proc Natl Acad Sci U S A ; 108(36): 14920-5, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21873198

ABSTRACT

The paramyxovirus hemagglutinin-neuraminidase (HN) protein plays multiple roles in viral entry and egress, including binding to sialic acid receptors, activating the fusion (F) protein to activate membrane fusion and viral entry, and cleaving sialic acid from carbohydrate chains. HN is an oligomeric integral membrane protein consisting of an N-terminal transmembrane domain, a stalk region, and an enzymatically active neuraminidase (NA) domain. Structures of the HN NA domains have been solved previously; however, the structure of the stalk region has remained elusive. The stalk region contains specificity determinants for F interactions and activation, underlying the requirement for homotypic F and HN interactions in viral entry. Mutations of the Newcastle disease virus HN stalk region have been shown to affect both F activation and NA activities, but a structural basis for understanding these dual affects on HN functions has been lacking. Here, we report the structure of the Newcastle disease virus HN ectodomain, revealing dimers of NA domain dimers flanking the N-terminal stalk domain. The stalk forms a parallel tetrameric coiled-coil bundle (4HB) that allows classification of extensive mutational data, providing insight into the functional roles of the stalk region. Mutations that affect both F activation and NA activities map predominantly to the 4HB hydrophobic core, whereas mutations that affect only F-protein activation map primarily to the 4HB surface. Two of four NA domains interact with the 4HB stalk, and residues at this interface in both the stalk and NA domain have been implicated in HN function.


Subject(s)
HN Protein/chemistry , Newcastle disease virus/enzymology , Crystallography, X-Ray , HN Protein/genetics , Mutation , Newcastle disease virus/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary
12.
Proc Natl Acad Sci U S A ; 108(23): 9619-24, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21586636

ABSTRACT

Respiratory syncytial virus (RSV), the main cause of infant bronchiolitis, remains a major unmet vaccine need despite more than 40 years of vaccine research. Vaccine candidates based on a chief RSV neutralization antigen, the fusion (F) glycoprotein, have foundered due to problems with stability, purity, reproducibility, and potency. Crystal structures of related parainfluenza F glycoproteins have revealed a large conformational change between the prefusion and postfusion states, suggesting that postfusion F antigens might not efficiently elicit neutralizing antibodies. We have generated a homogeneous, stable, and reproducible postfusion RSV F immunogen that elicits high titers of neutralizing antibodies in immunized animals. The 3.2-Å X-ray crystal structure of this substantially complete RSV F reveals important differences from homology-based structural models. Specifically, the RSV F crystal structure demonstrates the exposure of key neutralizing antibody binding sites on the surface of the postfusion RSV F trimer. This unanticipated structural feature explains the engineered RSV F antigen's efficiency as an immunogen. This work illustrates how structural-based antigen design can guide the rational optimization of candidate vaccine antigens.


Subject(s)
Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Viral Fusion Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Binding Sites, Antibody , Circular Dichroism , Crystallography, X-Ray , Humans , Immunization , Infant , Mice , Mice, Inbred BALB C , Microscopy, Electron , Models, Molecular , Molecular Sequence Data , Palivizumab , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/metabolism , Sequence Homology, Amino Acid , Sigmodontinae , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/ultrastructure
13.
Virology ; 402(2): 372-9, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20439109

ABSTRACT

The paramyxovirus F protein is a class I viral membrane fusion protein which undergoes a significant refolding transition during virus entry. Previous studies of the Newcastle disease virus, human parainfluenza virus 3 and parainfluenza virus 5 F proteins revealed differences in the pre- and post-fusion structures. The NDV Queensland (Q) F structure lacked structural elements observed in the other two structures, which are key to the refolding and fusogenic activity of F. Here we present the NDV Australia-Victoria (AV) F protein post-fusion structure and provide EM evidence for its folding to a pre-fusion form. The NDV AV F structure contains heptad repeat elements missing in the previous NDV Q F structure, forming a post-fusion six-helix bundle (6HB) similar to the post-fusion hPIV3 F structure. Electrostatic and temperature factor analysis of the F structures points to regions of these proteins that may be functionally important in their membrane fusion activity.


Subject(s)
Newcastle disease virus/chemistry , Protein Folding , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Newcastle disease virus/ultrastructure , Protein Conformation , Protein Structure, Tertiary , Viral Fusion Proteins/ultrastructure
14.
J Mol Biol ; 375(5): 1444-56, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18089292

ABSTRACT

The recruitment of chromatin-modifying coregulator complexes by transcription factors to specific sites of the genome constitutes an important step in many eukaryotic transcriptional regulatory pathways. The histone deacetylase-associated Sin3 corepressor complex is recruited by a large and diverse array of transcription factors through direct interactions with the N-terminal PAH domains of Sin3. Here, we describe the solution structures of the mSin3A PAH1 domain in the apo form and when bound to SAP25, a component of the corepressor complex. Unlike the apo-mSin3A PAH2 domain, the apo-PAH1 domain is conformationally pure and is largely, but not completely, folded. Portions of the interacting segments of both mSin3A PAH1 and SAP25 undergo folding upon complex formation. SAP25 binds through an amphipathic helix to a predominantly hydrophobic cleft on the surface of PAH1. Remarkably, the orientation of the helix is reversed compared to that adopted by NRSF, a transcription factor unrelated to SAP25, upon binding to the mSin3B PAH1 domain. The reversal in helical orientations is correlated with a reversal in the underlying PAH1-interaction motifs, echoing a theme previously described for the mSin3A PAH2 domain. The definition of these so-called type I and type II PAH1-interaction motifs has allowed us to predict the precise location of these motifs within previously experimentally characterized PAH1 binders. Finally, we explore the specificity determinants of protein-protein interactions involving the PAH1 and PAH2 domains. These studies reveal that even conservative replacements of PAH2 residues with equivalent PAH1 residues are sufficient to alter the affinity and specificity of these protein-protein interactions dramatically.


Subject(s)
Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Transcription Factors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites , Calorimetry , Conserved Sequence , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Mice , Models, Chemical , Models, Molecular , Molecular Sequence Data , Nitrogen Isotopes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sequence Homology, Amino Acid , Sin3 Histone Deacetylase and Corepressor Complex , Static Electricity , Titrimetry , Transcription Factors/metabolism
15.
J Mol Biol ; 358(3): 713-24, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16563434

ABSTRACT

Monoubiquitination is a general mechanism for downregulating the activity of cell surface receptors by consigning these proteins for lysosome-mediated degradation through the endocytic pathway. The yeast Ede1 protein functions at the internalization step of endocytosis and binds monoubiquitinated proteins through a ubiquitin associated (UBA) domain. UBA domains are found in a broad range of cellular proteins but previous studies have suggested that the mode of ubiquitin recognition might not be universally conserved. Here we present the solution structure of the Ede1 UBA domain in complex with monoubiquitin. The Ede1 UBA domain forms a three-helix bundle structure and binds ubiquitin through a largely hydrophobic surface in a manner reminiscent of the Dsk2 UBA and the remotely homologous Cue2 CUE domains, for which high-resolution structures have been described. However, the interaction is dissimilar to the molecular models proposed for the hHR23A UBA domains bound to either monoubiquitin or Lys48-linked diubiquitin. Our mutational analyses of the Ede1 UBA domain-ubiquitin interaction reveal several key affinity determinants and, unexpectedly, a negative affinity determinant in the wild-type Ede1 protein, implying that high-affinity interactions may not be the sole criterion for optimal function of monoubiquitin-binding endocytic proteins.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Ubiquitin/genetics
16.
Methods Enzymol ; 399: 135-57, 2005.
Article in English | MEDLINE | ID: mdl-16338353

ABSTRACT

To receive and transmit the information carried by ubiquitin signals, cells have evolved an array of modular ubiquitin-binding domains. These domains bind directly and noncovalently to monoubiquitin and polyubiquitin chains and are found within proteins that function in diverse biological processes. Ubiquitin-binding domains characterized thus far are generally small and structurally diverse, yet they all interact with the same hydrophobic patch on the surface of ubiquitin. The rapid identification and characterization of ubiquitin-binding domains has been accomplished through the extensive use of bioinformatics, biochemistry, molecular biology, and biophysics. Here, we discuss the strategies and tools that have been most successful in the identification and characterization of ubiquitin-binding domains.


Subject(s)
Ubiquitin/metabolism , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Two-Hybrid System Techniques , Ubiquitin/chemistry
17.
Nat Struct Mol Biol ; 11(8): 738-46, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15235594

ABSTRACT

Recruitment of the histone deacetylase (HDAC)-associated Sin3 corepressor is an obligatory step in many eukaryotic gene silencing pathways. Here we show that HBP1, a cell cycle inhibitor and regulator of differentiation, represses transcription in a HDAC/Sin3-dependent manner by targeting the mammalian Sin3A (mSin3A) PAH2 domain. HBP1 is unrelated to the Mad1 repressor for which high-resolution structures in complex with PAH2 have been described. We show that like Mad1, the HBP1 transrepression domain binds through a helical structure to the hydrophobic cleft of mSin3A PAH2. Notably, the HBP1 helix binds PAH2 in a reversed orientation relative to Mad1 and, equally unexpectedly, this is correlated with a chain reversal of the minimal Sin3 interaction motifs. These results not only provide insights into how multiple, unrelated transcription factors recruit the same coregulator, but also have implications for how sequence similarity searches are conducted.


Subject(s)
High Mobility Group Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Calorimetry , Cell Cycle Proteins , Cell Differentiation , Cell Line , Glutathione Transferase/metabolism , Histone Deacetylases , Humans , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Nuclear Proteins , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Stereoisomerism , Transcription, Genetic , Transfection , Two-Hybrid System Techniques
18.
EMBO J ; 22(18): 4597-606, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12970172

ABSTRACT

Monoubiquitylation is a well-characterized signal for the internalization and sorting of integral membrane proteins to distinct cellular organelles. Recognition and transmission of monoubiquitin signals is mediated by a variety of ubiquitin-binding motifs such as UIM, UBA, UEV, VHS and CUE in endocytic proteins. The yeast Vps27 protein requires two UIMs for efficient interactions with ubiquitin and for sorting cargo into multivesicular bodies. Here we show that the individual UIMs of Vps27 exist as autonomously folded alpha-helices that bind ubiquitin independently, non-cooperatively and with modest affinity. The Vps27 N-terminal UIM engages the Leu8-Ile44-Val70 hydrophobic patch of ubiquitin through a helical surface conserved in UIMs of diverse proteins, including that of the S5a proteasomal regulatory subunit. The Leu8-Ile44-Val70 ubiquitin surface is also the site of interaction for CUE and UBA domains in endocytic proteins, consistent with the view that ubiquitin-binding endocytic proteins act serially on the same monoubiquitylated cargo during transport from cell surface to the lysosome.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Vesicular Transport Proteins , Amino Acid Sequence , Animals , Binding Sites , Endosomal Sorting Complexes Required for Transport , Humans , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...