Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1143, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36854761

ABSTRACT

The protein phosphatase 2A (PP2A) heterotrimer PP2A-B56α is a human tumour suppressor. However, the molecular mechanisms inhibiting PP2A-B56α in cancer are poorly understood. Here, we report molecular level details and structural mechanisms of PP2A-B56α inhibition by an oncoprotein CIP2A. Upon direct binding to PP2A-B56α trimer, CIP2A displaces the PP2A-A subunit and thereby hijacks both the B56α, and the catalytic PP2Ac subunit to form a CIP2A-B56α-PP2Ac pseudotrimer. Further, CIP2A competes with B56α substrate binding by blocking the LxxIxE-motif substrate binding pocket on B56α. Relevant to oncogenic activity of CIP2A across human cancers, the N-terminal head domain-mediated interaction with B56α stabilizes CIP2A protein. Functionally, CRISPR/Cas9-mediated single amino acid mutagenesis of the head domain blunted MYC expression and MEK phosphorylation, and abrogated triple-negative breast cancer in vivo tumour growth. Collectively, we discover a unique multi-step hijack and mute protein complex regulation mechanism resulting in tumour suppressor PP2A-B56α inhibition. Further, the results unfold a structural determinant for the oncogenic activity of CIP2A, potentially facilitating therapeutic modulation of CIP2A in cancer and other diseases.


Subject(s)
Carcinogenesis , Protein Phosphatase 2 , Protein Processing, Post-Translational , Triple Negative Breast Neoplasms , Humans , Amino Acids , Carcinogenesis/genetics , Carcinogenesis/metabolism , Catalytic Domain , Phosphorylation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/ultrastructure , Triple Negative Breast Neoplasms/metabolism
2.
PLoS One ; 17(4): e0267343, 2022.
Article in English | MEDLINE | ID: mdl-35442992

ABSTRACT

An association between genetic variants in the genes HFE, HJV, BMP4 and arterial hypertension has been shown earlier. Proteins encoded by these genes participate in the signalling routes leading eventually to the production of the peptide hormone hepcidin. Mutations in these genes have been associated with the abnormal production of hepcidin in the body. This finding led to studies exploring the possible role of hepcidin in regulating the activity of blood pressure related renin-angiotensin system enzymes. We used molecular modelling to find out if it is possible for hepcidin to bind to the active site of the renin-angiotensin system enzymes, especially renin. Fluorometric assays were used to evaluate the inhibitory effect of hepcidin on renin as well as angiotensin converting enzymes 1 and 2. Finally, bio-layer interferometry technique was used to study hepcidin binding to renin. The molecular modelling showed that hepcidin seems to have similar binding properties to the renin active site as angiotensinogen does. Based on fluorometric enzyme activity assay, hepcidin has an inhibitory effect on renin in vitro, too. However, angiotensin converting enzymes 1 and 2 were not inhibited remarkably by hepcidin-25. In bio-layer interferometry analysis hepcidin-renin binding was concentration dependent. Our results suggest that hepcidin could act as an inhibitor to the renin. Nowadays, there is no known biological inhibitor for renin in vivo and our finding may thus have important clinical implications.


Subject(s)
Hypertension , Renin , Angiotensinogen/genetics , Blood Pressure , Hepcidins/genetics , Hepcidins/pharmacology , Humans , Renin-Angiotensin System
3.
Proc Natl Acad Sci U S A ; 117(51): 32402-32412, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288722

ABSTRACT

Binding of the intracellular adapter proteins talin and its cofactor, kindlin, to the integrin receptors induces integrin activation and clustering. These processes are essential for cell adhesion, migration, and organ development. Although the talin head, the integrin-binding segment in talin, possesses a typical FERM-domain sequence, a truncated form has been crystallized in an unexpected, elongated form. This form, however, lacks a C-terminal fragment and possesses reduced ß3-integrin binding. Here, we present a crystal structure of a full-length talin head in complex with the ß3-integrin tail. The structure reveals a compact FERM-like conformation and a tightly associated N-P-L-Y motif of ß3-integrin. A critical C-terminal poly-lysine motif mediates FERM interdomain contacts and assures the tight association with the ß3-integrin cytoplasmic segment. Removal of the poly-lysine motif or disrupting the FERM-folded configuration of the talin head significantly impairs integrin activation and clustering. Therefore, structural characterization of the FERM-folded active talin head provides fundamental understanding of the regulatory mechanism of integrin function.


Subject(s)
Integrin beta3/metabolism , Talin/chemistry , Talin/metabolism , Amino Acid Motifs , Animals , Binding Sites , Humans , Integrin beta3/chemistry , Leucine/metabolism , Mice , Microscopy, Electron, Transmission , Models, Molecular , Mutagenesis , Polylysine/chemistry , Protein Domains , Protein Folding , Talin/genetics
4.
J Cell Sci ; 133(19)2020 10 12.
Article in English | MEDLINE | ID: mdl-33046605

ABSTRACT

Integrin activation and clustering by talin are early steps of cell adhesion. Membrane-bound talin head domain and kindlin bind to the ß integrin cytoplasmic tail, cooperating to activate the heterodimeric integrin, and the talin head domain induces integrin clustering in the presence of Mn2+ Here we show that kindlin-1 can replace Mn2+ to mediate ß3 integrin clustering induced by the talin head, but not that induced by the F2-F3 fragment of talin. Integrin clustering mediated by kindlin-1 and the talin head was lost upon deletion of the flexible loop within the talin head F1 subdomain. Further mutagenesis identified hydrophobic and acidic motifs in the F1 loop responsible for ß3 integrin clustering. Modeling, computational and cysteine crosslinking studies showed direct and catalytic interactions of the acidic F1 loop motif with the juxtamembrane domains of α- and ß3-integrins, in order to activate the ß3 integrin heterodimer, further detailing the mechanism by which the talin-kindlin complex activates and clusters integrins. Moreover, the F1 loop interaction with the ß3 integrin tail required the newly identified compact FERM fold of the talin head, which positions the F1 loop next to the inner membrane clasp of the talin-bound integrin heterodimer.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Integrin beta3 , Talin , Cell Adhesion , Cluster Analysis , Integrin beta3/metabolism , Protein Binding , Protein Structure, Tertiary , Talin/genetics , Talin/metabolism
5.
J Pathol ; 252(2): 138-150, 2020 10.
Article in English | MEDLINE | ID: mdl-32666594

ABSTRACT

Sorsby fundus dystrophy (SFD) is a rare autosomal dominant disease of the macula that leads to bilateral loss of central vision and is caused by mutations in the TIMP3 gene. However, the mechanisms by which TIMP3 mutations cause SFD are poorly understood. Here, we generated human induced pluripotent stem cell-derived retinal pigmented epithelial (hiPSC-RPE) cells from three SFD patients carrying TIMP3 p.(Ser204Cys) and three non-affected controls to study disease-related structural and functional differences in the RPE. SFD-hiPSC-RPE exhibited characteristic RPE structure and physiology but showed significantly reduced transepithelial electrical resistance associated with enriched expression of cytoskeletal remodelling proteins. SFD-hiPSC-RPE exhibited basolateral accumulation of TIMP3 monomers, despite no change in TIMP3 gene expression. TIMP3 dimers were observed in both SFD and control hiPSC-RPE, suggesting that mutant TIMP3 dimerisation does not drive SFD pathology. Furthermore, mutant TIMP3 retained matrix metalloproteinase activity. Proteomic profiling showed increased expression of ECM proteins, endothelial cell interactions and angiogenesis-related pathways in SFD-hiPSC-RPE. By contrast, there were no changes in VEGF secretion. However, SFD-hiPSC-RPE secreted higher levels of monocyte chemoattractant protein 1, PDGF and angiogenin. Our findings provide a proof-of-concept that SFD patient-derived hiPSC-RPE mimic mature RPE cells and support the hypothesis that excess accumulation of mutant TIMP3, rather than an absence or deficiency of functional TIMP3, drives ECM and angiogenesis-related changes in SFD. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Macular Degeneration/pathology , Retinal Pigment Epithelium/pathology , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Adult , Cells, Cultured , Female , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells , Macular Degeneration/genetics , Macular Degeneration/metabolism , Middle Aged , Mutation , Proof of Concept Study , Retinal Pigment Epithelium/metabolism
6.
Viruses ; 11(12)2019 11 28.
Article in English | MEDLINE | ID: mdl-31795245

ABSTRACT

Enteroviruses are small RNA viruses that cause diseases with various symptoms ranging from mild to severe. Enterovirus proteins are translated as a single polyprotein, which is cleaved by viral proteases to release capsid and nonstructural proteins. Here, we show that also cellular calpains have a potential role in the processing of the enteroviral polyprotein. Using purified calpains 1 and 2 in an in vitro assay, we show that addition of calpains leads to an increase in the release of VP1 and VP3 capsid proteins from P1 of enterovirus B species, detected by western blotting. This was prevented with a calpain inhibitor and was dependent on optimal calcium concentration, especially for calpain 2. In addition, calpain cleavage at the VP3-VP1 interface was supported by a competition assay using a peptide containing the VP3-VP1 cleavage site. Moreover, a mass spectrometry analysis showed that calpains can cleave this same peptide at the VP3-VP1 interface, the cutting site being two amino acids aside from 3C's cutting site. Furthermore, we show that calpains cannot cleave between P1 and 2A. In conclusion, we show that cellular proteases, calpains, can cleave structural proteins from enterovirus polyprotein in vitro. Whether they assist polyprotein processing in infected cells remains to be shown.


Subject(s)
Calpain/metabolism , Capsid Proteins/metabolism , Enterovirus Infections/virology , Enterovirus/metabolism , Polyproteins/metabolism , Animals , Capsid/metabolism , Cells, Cultured , Glycoproteins/pharmacology , Humans , Mass Spectrometry , Peptides/metabolism , Proteolysis , Rats , Viral Proteins/metabolism
7.
PLoS One ; 14(2): e0212339, 2019.
Article in English | MEDLINE | ID: mdl-30785944

ABSTRACT

Chicken avidin (Avd) and streptavidin from Streptomyces avidinii are extensively used in bionanotechnology due to their extremely tight binding to biotin (Kd ~ 10-15 M for chicken Avd). We previously reported engineered Avds known as antidins, which have micro- to nanomolar affinities for steroids, non-natural ligands of Avd. Here, we report the 2.8 Å X-ray structure of the sbAvd-2 (I117Y) antidin co-crystallized with progesterone. We describe the creation of new synthetic phage display libraries and report the experimental as well as computational binding analysis of progesterone-binding antidins. We introduce a next-generation antidin with 5 nM binding affinity for progesterone, and demonstrate the use of antidins for measuring progesterone in serum samples. Our data give insights on how to engineer and alter the binding preferences of Avds and to develop better molecular tools for modern bionanotechnological applications.


Subject(s)
Avidin/metabolism , Biotin/metabolism , Progesterone/blood , Progesterone/metabolism , Animals , Avidin/chemistry , Binding Sites , Biological Assay , Biotin/chemistry , Dogs , Ligands , Models, Molecular , Progesterone/chemistry , Protein Binding
8.
Sci Rep ; 9(1): 995, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700796

ABSTRACT

Tuberculosis is a multifactorial bacterial disease, which can be modeled in the zebrafish (Danio rerio). Abdominal cavity infection with Mycobacterium marinum, a close relative of Mycobacterium tuberculosis, leads to a granulomatous disease in adult zebrafish, which replicates the different phases of human tuberculosis, including primary infection, latency and spontaneous reactivation. Here, we have carried out a transcriptional analysis of zebrafish challenged with low-dose of M. marinum, and identified intelectin 3 (itln3) among the highly up-regulated genes. In order to clarify the in vivo significance of Itln3 in immunity, we created nonsense itln3 mutant zebrafish by CRISPR/Cas9 mutagenesis and analyzed the outcome of M. marinum infection in both zebrafish embryos and adult fish. The lack of functional itln3 did not affect survival or the mycobacterial burden in the zebrafish. Furthermore, embryonic survival was not affected when another mycobacterial challenge responsive intelectin, itln1, was silenced using morpholinos either in the WT or itln3 mutant fish. In addition, M. marinum infection in dexamethasone-treated adult zebrafish, which have lowered lymphocyte counts, resulted in similar bacterial burden in both WT fish and homozygous itln3 mutants. Collectively, although itln3 expression is induced upon M. marinum infection in zebrafish, it is dispensable for protective mycobacterial immune response.


Subject(s)
Cytokines/metabolism , Lectins/metabolism , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/physiology , Zebrafish Proteins/metabolism , Zebrafish/microbiology , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Codon, Nonsense/genetics , Cytokines/genetics , Dexamethasone/pharmacology , Disease Resistance/immunology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/microbiology , Gene Expression Regulation/drug effects , Genome , Lectins/genetics , Lymphocyte Depletion , Morpholinos/pharmacology , Mutation/genetics , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/drug effects , Survival Analysis , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
J Virol Methods ; 255: 29-37, 2018 05.
Article in English | MEDLINE | ID: mdl-29425680

ABSTRACT

Enteroviruses (EVs), such as the Coxsackie B-viruses (CVBs), are common human pathogens, which can cause severe diseases including meningitis, myocarditis and neonatal sepsis. EVs encode two proteases (2Apro and 3Cpro), which perform the proteolytic cleavage of the CVB polyprotein and also cleave host cell proteins to facilitate viral replication. The 2Apro cause direct damage to the infected heart and tools to investigate 2Apro and 3Cpro expression may contribute new knowledge on virus-induced pathologies. Here, we developed new antibodies to CVB-encoded 2Apro and 3Cpro; Two monoclonal 2Apro antibodies and one 3Cpro antibody were produced. Using cells infected with selected viruses belonging to the EV A, B and C species and immunocytochemistry, we demonstrate that the 3Cpro antibody detects all of the EV species B (EV-B) viruses tested and that the 2Apro antibody detects all EV-B viruses apart from Echovirus 9. We furthermore show that the new antibodies work in Western blotting, immunocyto- and immunohistochemistry, and flow cytometry to detect CVBs. Confocal microscopy demonstrated the expression kinetics of 2Apro and 3Cpro, and revealed a preferential cytosolic localization of the proteases in CVB3 infected cells. In summary, the new antibodies detect proteases that belong to EV species B in cells and tissue using multiple applications.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Cysteine Endopeptidases/immunology , Enterovirus B, Human/immunology , Enterovirus Infections/diagnosis , Enterovirus Infections/immunology , Viral Proteins/immunology , 3C Viral Proteases , Animals , Antigens, Viral/genetics , Cells, Cultured , Cloning, Molecular , Cysteine Endopeptidases/genetics , Enterovirus B, Human/enzymology , Enterovirus B, Human/genetics , Enterovirus Infections/virology , Gene Expression , HeLa Cells , Humans , Immunohistochemistry , Mice , Serogroup , Viral Proteins/genetics
10.
Vaccine ; 35(30): 3718-3725, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28579231

ABSTRACT

Coxsackie B viruses are among the most common enteroviruses, causing a wide range of diseases. Recent studies have also suggested that they may contribute to the development of type 1 diabetes. Vaccination would provide an effective way to prevent CVB infections, and the objective of this study was to develop an efficient vaccine production protocol for the generation of novel CVB vaccines. Various steps in the production of a formalin-inactivated Coxsackievirus B1 (CVB1) vaccine were optimized including the Multiplicity Of Infection (MOI) used for virus amplification, virus cultivation time, type of cell growth medium, virus purification method and formulation of the purified virus. Safety and immunogenicity of the formalin inactivated CVB1 vaccine was characterized in a mouse model. Two of the developed methods were found to be optimal for virus purification: the first employed PEG-precipitation followed by gelatin-chromatography and sucrose cushion pelleting (three-step protocol), yielding 19-fold increase in virus concentration (0.06µg/cm2) as compared to gold standard method. The second method utilized tandem sucrose pelleting without a PEG precipitation step, yielding 83-fold increase in virus concentration (0.24µg/cm2), but it was more labor-intensive and cannot be efficiently scaled up. Both protocols provide radically higher virus yields compared with traditional virus purification protocols involving PEG-precipitation and sucrose gradient ultracentrifugation. Formalin inactivation of CVB1 produced a vaccine that induced a strong, virus-neutralizing antibody response in vaccinated mice, which protected against challenge with CVB1 virus. Altogether, these results provide valuable information for the development of new enterovirus vaccines.


Subject(s)
Coxsackievirus Infections/prevention & control , Enterovirus A, Human/immunology , Immunogenicity, Vaccine , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chlorocebus aethiops , Coxsackievirus Infections/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Enterovirus A, Human/growth & development , Enterovirus A, Human/isolation & purification , Female , Formaldehyde/pharmacology , Mice , Polysorbates/pharmacology , Vaccination , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/isolation & purification , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/isolation & purification , Virus Cultivation
11.
PLoS One ; 12(4): e0176086, 2017.
Article in English | MEDLINE | ID: mdl-28426764

ABSTRACT

Bradavidin is a tetrameric biotin-binding protein similar to chicken avidin and bacterial streptavidin, and was originally cloned from the nitrogen-fixing bacteria Bradyrhizobium diazoefficiens. We have previously reported the crystal structure of the full-length, wild-type (wt) bradavidin with 138 amino acids, where the C-terminal residues Gly129-Lys138 ("Brad-tag") act as an intrinsic ligand (i.e. Gly129-Lys138 bind into the biotin-binding site of an adjacent subunit within the same tetramer) and has potential as an affinity tag for biotechnological purposes. Here, the X-ray structure of core-bradavidin lacking the C-terminal residues Gly114-Lys138, and hence missing the Brad-tag, was crystallized in complex with biotin at 1.60 Å resolution [PDB:4BBO]. We also report a homology model of rhodavidin, an avidin-like protein from Rhodopseudomonas palustris, and of an avidin-like protein from Bradyrhizobium sp. Ai1a-2, both of which have the Brad-tag sequence at their C-terminus. Moreover, core-bradavidin V1, an engineered variant of the original core-bradavidin, was also expressed at high levels in E. coli, as well as a double mutant (Cys39Ala and Cys69Ala) of core-bradavidin (CC mutant). Our data help us to further engineer the core-bradavidin-Brad-tag pair for biotechnological assays and chemical biology applications, and provide deeper insight into the biotin-binding mode of bradavidin.


Subject(s)
Biotin/chemistry , Carrier Proteins/chemistry , Affinity Labels , Binding Sites , Crystallography, X-Ray , Models, Molecular , Protein Conformation
12.
Amino Acids ; 49(3): 529-540, 2017 03.
Article in English | MEDLINE | ID: mdl-27503559

ABSTRACT

Coeliac disease is hallmarked by an abnormal immune reaction against ingested wheat-, rye- and barley-derived gluten and the presence of transglutaminase 2 (TG2)-targeted autoantibodies. The small-bowel mucosal damage characteristic of the disorder develops gradually from normal villus morphology to inflammation and finally to villus atrophy with crypt hyperplasia. Patients with early-stage coeliac disease have TG2-autoantibodies present in serum and small-intestinal mucosa and they may already suffer from abdominal symptoms before the development of villus atrophy. Previously, we have shown that intraperitoneal injections of coeliac patient-derived sera or purified immunoglobulin fraction into mice induce a condition mimicking early-stage coeliac disease. In the current study, we sought to establish whether recombinantly produced patient-derived TG2-targeted autoantibodies are by themselves sufficient for the development of such an experimentally induced condition in immune-compromised mice. Interestingly, mice injected with coeliac patient TG2-antibodies had altered small-intestinal mucosal morphology, increased lamina propria cellular infiltration and disease-specific autoantibodies deposited in the small bowel, but did not evince clinical features of the disease. Thus, coeliac patient-derived TG2-specific autoantibodies seem to be sufficient for the induction of subtle small-bowel mucosal alterations in mice, but the development of clinical features probably requires additional factors such as other antibody populations relevant in coeliac disease.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Autoantibodies/biosynthesis , Celiac Disease/immunology , GTP-Binding Proteins/immunology , Immunocompromised Host , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Transglutaminases/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , CHO Cells , Celiac Disease/genetics , Celiac Disease/pathology , Cricetulus , Female , GTP-Binding Proteins/genetics , Gene Expression , Glutens/chemistry , Glutens/immunology , Humans , Immunoglobulin A/biosynthesis , Immunohistochemistry , Inflammation , Injections, Intraperitoneal , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Mice , Mice, Nude , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transglutaminases/genetics
13.
J Virol ; 89(15): 7593-603, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972558

ABSTRACT

UNLABELLED: Thermus thermophilus bacteriophage P23-77 is the type member of a new virus family of icosahedral, tailless, inner-membrane-containing double-stranded DNA (dsDNA) viruses infecting thermophilic bacteria and halophilic archaea. The viruses have a unique capsid architecture consisting of two major capsid proteins assembled in various building blocks. We analyzed the function of the minor capsid protein VP11, which is the third known capsid component in bacteriophage P23-77. Our findings show that VP11 is a dynamically elongated dimer with a predominantly α-helical secondary structure and high thermal stability. The high proportion of basic amino acids in the protein enables electrostatic interaction with negatively charged molecules, including nucleic acid and large unilamellar lipid vesicles (LUVs). The plausible biological function of VP11 is elucidated by demonstrating the interactions of VP11 with Thermus-derived LUVs and with the major capsid proteins by means of the dynamic-light-scattering technique. In particular, the major capsid protein VP17 was able to link VP11-complexed LUVs into larger particles, whereas the other P23-77 major capsid protein, VP16, was unable to link VP11-comlexed LUVs. Our results rule out a previously suggested penton function for VP11. Instead, the electrostatic membrane association of VP11 triggers the binding of the major capsid protein VP17, thus facilitating a controlled incorporation of the two different major protein species into the assembling capsid. IMPORTANCE: The study of thermophilic viruses with inner membranes provides valuable insights into the mechanisms used for stabilization and assembly of protein-lipid systems at high temperatures. Our results reveal a novel way by which an internal membrane and outer capsid shell are linked in a virus that uses two different major protein species for capsid assembly. We show that a positive protein charge is important in order to form electrostatic interactions with the lipid surface, thereby facilitating the incorporation of other capsid proteins on the membrane surface. This implies an alternative function for basic proteins present in the virions of other lipid-containing thermophilic viruses, whose proposed role in genome packaging is based on their capability to bind DNA. The unique minor capsid protein of bacteriophage P23-77 resembles in its characteristics the scaffolding proteins of tailed phages, though it constitutes a substantial part of the mature virion.


Subject(s)
Bacteriophages/metabolism , Capsid Proteins/metabolism , Lipids/chemistry , Thermus/metabolism , Virus Assembly , Amino Acid Sequence , Bacteriophages/chemistry , Bacteriophages/genetics , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Lipid Metabolism , Models, Molecular , Molecular Sequence Data , Static Electricity , Thermus/chemistry , Thermus/virology , Virion/chemistry , Virion/genetics , Virion/metabolism
14.
Bioconjug Chem ; 25(12): 2233-43, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25405260

ABSTRACT

Switchavidin is a chicken avidin mutant displaying reversible binding to biotin, an improved binding affinity toward conjugated biotin, and low nonspecific binding due to reduced surface charge. These properties make switchavidin an optimal tool in biosensor applications for the reversible immobilization of biotinylated proteins on biotinylated sensor surfaces. Furthermore, switchavidin opens novel possibilities for patterning, purification, and labeling.


Subject(s)
Avidin/chemistry , Avidin/metabolism , Biosensing Techniques , Biotin/chemistry , 3T3 Cells , Animals , Avidin/genetics , Binding Sites , Biotinylation , Calorimetry, Differential Scanning , Chickens , Mice , Mutation , Surface Plasmon Resonance
15.
Mol Biosyst ; 10(12): 3217-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25277990

ABSTRACT

The major mechanical function of talin is to couple the ß-integrin cytoplasmic tails to actin filaments. A variety of ß-integrin tails contain conserved binding motifs for talin, and recent research shows that ß-integrins differ both in affinity to talin and preferences for other cytoplasmic adaptor proteins. While talin predominantly links ß3 integrins to actin filaments within the peripheral cell adhesion sites, talin can become replaced by other integrin adaptor proteins through their overlapping binding sites on integrin tails. Although the NPxY motif in the ß-integrin tail is important for talin recognition, our simulations suggest considerably smaller contribution of the NPxY motif in the force resistance of the talin-integrin complex than for the residues upstream of the NPxY. It might thus be possible for the NPxY motif to detach from talin and interact with other integrin binding proteins while the ß-integrin still remains bound to talin. The epithelial integrin ß6 reportedly activates latent TGFß1, and we propose that its function may involve direct interaction with talin.


Subject(s)
Integrins/chemistry , Stress, Mechanical , Talin/chemistry , Amino Acid Sequence , Biosensing Techniques , Glutamine/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry
16.
PLoS One ; 9(6): e100564, 2014.
Article in English | MEDLINE | ID: mdl-24959850

ABSTRACT

Chimeric avidin (ChiAVD) is a product of rational protein engineering remarkably resistant to heat and harsh conditions. In quest of the fundamentals behind factors affecting stability we have elucidated the solution NMR spectroscopic structure of the biotin-bound form of ChiAVD and characterized the protein dynamics through 15N relaxation and hydrogen/deuterium (H/D) exchange of this and the biotin-free form. To surmount the challenges arising from the very large size of the protein for NMR spectroscopy, we took advantage of its high thermostability. Conventional triple resonance experiments for fully protonated proteins combined with methyl-detection optimized experiments acquired at 58°C were adequate for the structure determination of this 56 kDa protein. The model-free parameters derived from the 15N relaxation data reveal a remarkably rigid protein at 58°C in both the biotin-bound and the free forms. The H/D exchange experiments indicate a notable increase in hydrogen protection upon biotin binding.


Subject(s)
Avidin/chemistry , Avidin/metabolism , Biotin/chemistry , Biotin/metabolism , Models, Molecular , Molecular Weight , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Protein Engineering , Protein Multimerization , Thermodynamics
17.
FEBS Lett ; 588(9): 1523-8, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24681097

ABSTRACT

The modulation of protein deacetylase SIRT1 has a vast therapeutic potential in treatment of several aging-associated diseases. Active regulator of SIRT1 (AROS) is a small endogenous protein which was originally reported to activate SIRT1 through a direct interaction in cancer cells. We show that the interaction between the two proteins is weak and does not alter the activity of SIRT1 in non-cancerous human cells. The results of different in vitro SIRT1 activity assays disclosed AROS as an inhibitor of SIRT1. The functional relationship between AROS and SIRT1 proved to be dependent on the biological context and experimental setting.


Subject(s)
Nuclear Proteins/metabolism , Sirtuin 1/metabolism , Transcription Factors/metabolism , Acetylation , Epithelial Cells/enzymology , HEK293 Cells , Histones/chemistry , Humans , NAD/chemistry , Nuclear Proteins/chemistry , Protein Binding , Protein Processing, Post-Translational , Retinal Pigment Epithelium/cytology , Sirtuin 1/chemistry , Transcription Factors/chemistry , Tumor Suppressor Protein p53/metabolism
18.
J Cell Biol ; 205(2): 265-81, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24778313

ABSTRACT

Integrin-dependent cell adhesion and spreading are critical for morphogenesis, tissue regeneration, and immune defense but also tumor growth. However, the mechanisms that induce integrin-mediated cell spreading and provide mechanosensing on different extracellular matrix conditions are not fully understood. By expressing ß3-GFP-integrins with enhanced talin-binding affinity, we experimentally uncoupled integrin activation, clustering, and substrate binding from its function in cell spreading. Mutational analysis revealed Tyr747, located in the first cytoplasmic NPLY(747) motif, to induce spreading and paxillin adapter recruitment to substrate- and talin-bound integrins. In addition, integrin-mediated spreading, but not focal adhesion localization, was affected by mutating adjacent sequence motifs known to be involved in kindlin binding. On soft, spreading-repellent fibronectin substrates, high-affinity talin-binding integrins formed adhesions, but normal spreading was only possible with integrins competent to recruit the signaling adapter protein paxillin. This proposes that integrin-dependent cell-matrix adhesion and cell spreading are independently controlled, offering new therapeutic strategies to modify cell behavior in normal and pathological conditions.


Subject(s)
Integrin beta3/metabolism , Mechanotransduction, Cellular/physiology , Talin/metabolism , Amino Acid Motifs , Animals , COS Cells , Cell Adhesion/physiology , Chlorocebus aethiops , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Integrin beta3/genetics , Mice , NIH 3T3 Cells , Paxillin/genetics , Paxillin/metabolism , Talin/genetics
19.
PLoS One ; 8(10): e77207, 2013.
Article in English | MEDLINE | ID: mdl-24204770

ABSTRACT

The avidin protein family members are well known for their high affinity towards D-biotin and high structural stability. These properties make avidins valuable tools for a wide range of biotechnology applications. We have identified a new member of the avidin family in the zebrafish (Danio rerio) genome, hereafter called zebavidin. The protein is highly expressed in the gonads of both male and female zebrafish and in the gills of male fish, but our data suggest that zebavidin is not crucial for the developing embryo. Biophysical and structural characterisation of zebavidin revealed distinct properties not found in any previously characterised avidins. Gel filtration chromatography and native mass spectrometry suggest that the protein forms dimers in the absence of biotin at low ionic strength, but assembles into tetramers upon binding biotin. Ligand binding was analysed using radioactive and fluorescently labelled biotin and isothermal titration calorimetry. Moreover, the crystal structure of zebavidin in complex with biotin was solved at 2.4 Å resolution and unveiled unique ligand binding and subunit interface architectures; the atomic-level details support our physicochemical observations.


Subject(s)
Avidin/chemistry , Fish Proteins/chemistry , Genome , Glycoproteins/chemistry , Zebrafish Proteins/chemistry , Zebrafish/genetics , Amino Acid Sequence , Animals , Avidin/genetics , Avidin/metabolism , Biotin/chemistry , Biotin/metabolism , Crystallography, X-Ray , Embryo, Nonmammalian , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression , Gills/embryology , Gills/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Gonads/embryology , Gonads/metabolism , Male , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Biochim Biophys Acta ; 1834(10): 1988-97, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856547

ABSTRACT

T-cell protein tyrosine phosphatase (TCPTP) is a ubiquitously expressed non-receptor protein tyrosine phosphatase. It is involved in the negative regulation of many cellular signaling pathways. Thus, activation of TCPTP could have important therapeutic applications in diseases such as cancer and inflammation. We have previously shown that the α-cytoplasmic tail of integrin α1ß1 directly binds and activates TCPTP. In addition, we have identified in a large-scale high-throughput screen six small molecules that activate TCPTP. These small molecule activators include mitoxantrone and spermidine. In this study, we have investigated the molecular mechanism behind agonist-induced TCPTP activation. By combining several molecular modeling and biochemical techniques, we demonstrate that α1-peptide and mitoxantrone activate TCPTP via direct binding to the catalytic domain, whereas spermidine does not interact with the catalytic domain of TCPTP in vitro. Furthermore, we have identified a hydrophobic groove surrounded by negatively charged residues on the surface of TCPTP as a putative binding site for the α1-peptide and mitoxantrone. Importantly, these data have allowed us to identify a new molecule that binds to TCPTP, but interestingly cannot activate its phosphatase activity. Accordingly, we describe here mechanism of TCPTP activation by mitoxantrone, the cytoplasmic tail of α1-integrin, and a mitoxantrone-like molecule at the atomic level. These data provide invaluable insight into the development of novel TCPTP activators, and may facilitate the rational discovery of small-molecule cancer therapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Integrin alpha1beta1/chemistry , Mitoxantrone/chemistry , Peptides/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 2/chemistry , Small Molecule Libraries/chemistry , Spermidine/chemistry , Databases, Protein , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Static Electricity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...