Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Nat Prod ; 87(6): 1628-1634, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38869194

ABSTRACT

The unfolded protein response (UPR) is a key component of fungal virulence. The prenylated xanthone γ-mangostin isolated from Garcinia mangostana (Clusiaceae) fruit pericarp, has recently been described to inhibit this fungal adaptative pathway. Considering that Calophyllum caledonicum (Calophyllaceae) is known for its high prenylated xanthone content, its stem bark extract was fractionated using a bioassay-guided procedure based on the cell-based anti-UPR assay. Four previously undescribed xanthone derivatives were isolated, caledonixanthones N-Q (3, 4, 8, and 12), among which compounds 3 and 8 showed promising anti-UPR activities with IC50 values of 11.7 ± 0.9 and 7.9 ± 0.3 µM, respectively.


Subject(s)
Calophyllum , Unfolded Protein Response , Xanthones , Xanthones/pharmacology , Xanthones/chemistry , Xanthones/isolation & purification , Unfolded Protein Response/drug effects , Calophyllum/chemistry , Molecular Structure , Humans , Plant Bark/chemistry
2.
J Agric Food Chem ; 71(37): 13706-13716, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37697453

ABSTRACT

Discovering new solutions for crop protection is a major challenge for the next decades as a result of the ecotoxicological impact of classical fungicides, the emergence of fungicide resistances, and the consequence of climate change on pathogen distribution. Previous work on fungal mutants deficient in the unfolded protein response (UPR) supported that targeting this pathway is a promising plant disease control strategy. In particular, we showed that the UPR is involved in fungal virulence by altering cell protection against host defense compounds, such as phytoalexins and phytoanticipins. In this study, we evaluated natural products targeting fungal IRE1 protein (UPR effector) and consequently increasing fungal susceptibility to plant defenses. Developing an in vitro cell-based screening assay allowed for the identification of seven potential IRE1 inhibitors with a focus on polyhydroxylated prenylated xanthones. Inhibition of hac1 mRNA splicing, which is mediated by IRE1, was then validated for the most active compound, namely, γ-mangostin 3. To study the mode of interaction between the binding site of IRE1 and active xanthones, molecular docking was also undertaken, revealing similar and novel interactions between the known inhibitor and the binding site. Eventually, active xanthones applied at subtoxic doses induced a significant reduction in necrosis size for leaves of Brassica oleracea inoculated with Alternaria brassicicola and Botrytis cinerea.


Subject(s)
Biological Products , Fungicides, Industrial , Crop Protection , Molecular Docking Simulation , Binding Sites , Fungal Proteins/genetics , Fungicides, Industrial/pharmacology , Protein Serine-Threonine Kinases
3.
Int J Parasitol Parasites Wildl ; 22: 69-74, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37720360

ABSTRACT

Toxoplasma gondii is an intracellular protozoon found worldwide, which completes its life cycle between felids (its definitive host) and other warm-blooded animals. While the infection rarely leads to severe complications in humans, many animal species are very susceptible to this infection, for example the squirrel monkey (Saimiri sciureus) which is the subject of this study. Toxoplasmosis is lethal for 80% of cases in this species, and fatal outbreaks are frequently reported in zoological parks. No efficient treatment exists, but a new vaccine prepared with maltodextrin nanoparticles containing killed T. gondii antigens has been tested recently in French zoos. The animals were immunized through heterologous administrations, with two nasal doses at one-month interval, followed by nasal/subcutaneous boosts thereafter. No death has been reported since the beginning of this vaccination campaign, but we felt the protocol could be simplified. Here, an improved and less-invasive immunization protocol was evaluated on 6 Saimiri sciureus in the French zoo La Palmyre. It consisted of two nasal administrations at one-month interval, followed by a nasal boost at 6 months. A specific memory T-cell immunity was observed by ELISPOT after two administrations in all the animals, without humoral responses. The results suggest that 2 nasal administrations induce a protective immune response against T. gondii infection and might be sufficient to induce a strong Tcell memory, further improving immunity.

4.
Plants (Basel) ; 11(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35050032

ABSTRACT

Concentrated bud macerates (CBMs) are obtained from meristematic tissues such as buds and young shoots by maceration in a solvent composed of glycerin, water and ethanol (1/1/1/, v/v). Their traditional utilization in gemmotherapy has gained interest in the past years, and the knowledge of their chemical characterization can provide commercial arguments, particularly to secure their quality control. Therefore, an optimized method for phytochemical analysis including glycerol removal by a preliminary solid phase extraction (SPE) followed by compound identification using high performance liquid chromatography coupled with ultra-violet and tandem mass detectors (HPLC-UV-MS2) was developed. This method was applied on 5 CBMs obtained from Alnus glutinosa, Ribesnigrum, Rosmarinus officinalis, Rosa canina and Tilia tomentosa in order to determinate their chemical composition. Their antioxidant effects were also investigated by radical scavenging activity assays (DPPH and ORAC). Glycerol removal improved the resolution of HPLC chemical profiles and allowed us to perform TLC antioxidant screening. Our approach permitted the identification of 57 compounds distributed in eight major classes, three of them being common to all macerates including nucleosides, phenolic acids and glycosylated flavonoids. Quantification of the later class as a rutin equivalent (RE) showed a great disparity between Rosa canina macerate (809 mg RE/L), and the other ones (from 175 to 470 mg RE/L). DPPH and ORAC assays confirmed the great activity of Rosa canina (4857 and 6479 µmol TE/g of dry matter, respectively). Finally, phytochemical and antioxidant analysis of CBMs strengthened their phytomedicinal interest in the gemmotherapy field.

5.
J Vet Intern Med ; 34(6): 2365-2373, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33140893

ABSTRACT

BACKGROUND: The dog erythrocyte antigen (DEA) 1 blood group is considered as the most immunogenic and clinically important in dogs. Little is known in nondomesticated canids. OBJECTIVES: To type DEA 1 in nondomesticated captive canids and to evaluate potential interspecific blood transfusions between domestic and nondomestic canids. ANIMALS: One hundred forty captive nondomesticated canids belonging to 13 species from 19 French zoos, and 63 domestic dogs. METHODS: Prospective study. Blood samples were typed for DEA 1 using immunochromatographic and flow cytometric techniques. A neutral gel column test was used for crossmatching. RESULTS: Of 140 nondomesticated canids, 72.9% were DEA 1+ and 27.1% were DEA 1- using immunochromatographic technique and 74.3% were DEA 1+ and 25.7% were DEA 1- by flow cytometric technique. Crossmatch (XM) between 140 nondomesticated canid red blood cells (RBCs) and plasma from a previously DEA 1+ sensitized DEA 1- dog revealed 112 incompatibilities (80%). Crossmatches between 130 nondomesticated canid serum and 1 or up to 8 donor dogs' RBCs revealed 99 of 130 (76%) compatibilities. Crossmatches between 115 nondomesticated canid RBCs and donor dogs' serum revealed 59 of 115 (51%) compatibilities. CONCLUSIONS AND CLINICAL IMPORTANCE: Dog erythrocyte antigen 1 blood type is present in nondomesticated canids with variable prevalence depending on species. The majority of tested nondomesticated canids appear to have no naturally occurring alloantibodies against domestic dogs' RBCs. Therefore xenotransfusion of blood from domestic dogs can be considered when species specific blood is not available. Cross matching is essential before xenotransfusion.


Subject(s)
Blood Group Antigens , Animals , Blood Grouping and Crossmatching/veterinary , Blood Transfusion/veterinary , Dogs , Erythrocytes , Prospective Studies
6.
Mol Cancer Res ; 17(4): 963-973, 2019 04.
Article in English | MEDLINE | ID: mdl-30567972

ABSTRACT

Uveal melanoma is the most common intraocular tumor in adults and often metastasizes to the liver, leaving patients with few options. Recurrent activating mutations in the G proteins, Gαq and Gα11, are observed in approximately 93% of all uveal melanomas. Although therapeutic intervention of downstream Gαq/11 targets has been unsuccessful in treating uveal melanoma, we have found that the Gαq/11 inhibitor, FR900359 (FR), effectively inhibits oncogenic Gαq/11 signaling in uveal melanoma cells expressing either mutant Gαq or Gα11. Inhibition of oncogenic Gαq/11 by FR results in cell-cycle arrest and induction of apoptosis. Furthermore, colony formation is prevented by FR treatment of uveal melanoma cells in 3D-cell culture, providing promise for future in vivo studies. This suggests direct inhibition of activating Gαq/11 mutants may be a potential means of treating uveal melanoma. IMPLICATIONS: Oncogenic Gαq/11 inhibition by FR900359 may be a potential treatment option for those with uveal melanoma.


Subject(s)
Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , Melanoma/drug therapy , Uveal Neoplasms/drug therapy , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/isolation & purification , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Insecta/cytology , MAP Kinase Signaling System , Melanoma/metabolism , Melanoma/pathology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology
7.
J Biol Chem ; 291(49): 25608-25616, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27742837

ABSTRACT

In contrast to G protein-coupled receptors, for which chemical and peptidic inhibitors have been extensively explored, few compounds are available that directly modulate heterotrimeric G proteins. Active Gαq binds its two major classes of effectors, the phospholipase C (PLC)-ß isozymes and Rho guanine nucleotide exchange factors (RhoGEFs) related to Trio, in a strikingly similar fashion: a continuous helix-turn-helix of the effectors engages Gαq within its canonical binding site consisting of a groove formed between switch II and helix α3. This information was exploited to synthesize peptides that bound active Gαq in vitro with affinities similar to full-length effectors and directly competed with effectors for engagement of Gαq A representative peptide was specific for active Gαq because it did not bind inactive Gαq or other classes of active Gα subunits and did not inhibit the activation of PLC-ß3 by Gß1γ2 In contrast, the peptide robustly prevented activation of PLC-ß3 or p63RhoGEF by Gαq; it also prevented G protein-coupled receptor-promoted neuronal depolarization downstream of Gαq in the mouse prefrontal cortex. Moreover, a genetically encoded form of this peptide flanked by fluorescent proteins inhibited Gαq-dependent activation of PLC-ß3 at least as effectively as a dominant-negative form of full-length PLC-ß3. These attributes suggest that related, cell-penetrating peptides should effectively inhibit active Gαq in cells and that these and genetically encoded sequences may find application as molecular probes, drug leads, and biosensors to monitor the spatiotemporal activation of Gαq in cells.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Prefrontal Cortex/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , HEK293 Cells , Humans , Mice , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Protein Structure, Secondary
8.
J Med Chem ; 59(2): 592-608, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26727270

ABSTRACT

The drug pentamidine inhibits calcium-dependent complex formation with p53 ((Ca)S100B·p53) in malignant melanoma (MM) and restores p53 tumor suppressor activity in vivo. However, off-target effects associated with this drug were problematic in MM patients. Structure-activity relationship (SAR) studies were therefore completed here with 23 pentamidine analogues, and X-ray structures of (Ca)S100B·inhibitor complexes revealed that the C-terminus of S100B adopts two different conformations, with location of Phe87 and Phe88 being the distinguishing feature and termed the "FF-gate". For symmetric pentamidine analogues ((Ca)S100B·5a, (Ca)S100B·6b) a channel between sites 1 and 2 on S100B was occluded by residue Phe88, but for an asymmetric pentamidine analogue ((Ca)S100B·17), this same channel was open. The (Ca)S100B·17 structure illustrates, for the first time, a pentamidine analog capable of binding the "open" form of the "FF-gate" and provides a means to block all three "hot spots" on (Ca)S100B, which will impact next generation (Ca)S100B·p53 inhibitor design.


Subject(s)
S100 Calcium Binding Protein beta Subunit/antagonists & inhibitors , S100 Calcium Binding Protein beta Subunit/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cattle , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Pentamidine/analogs & derivatives , Pentamidine/chemistry , Pentamidine/pharmacology , Protein Conformation , Rats , Small Molecule Libraries , Structure-Activity Relationship , Tumor Suppressor Protein p53/drug effects
9.
Nat Commun ; 6: 10156, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26658454

ABSTRACT

Despite the discovery of heterotrimeric αßγ G proteins ∼25 years ago, their selective perturbation by cell-permeable inhibitors remains a fundamental challenge. Here we report that the plant-derived depsipeptide FR900359 (FR) is ideally suited to this task. Using a multifaceted approach we systematically characterize FR as a selective inhibitor of Gq/11/14 over all other mammalian Gα isoforms and elaborate its molecular mechanism of action. We also use FR to investigate whether inhibition of Gq proteins is an effective post-receptor strategy to target oncogenic signalling, using melanoma as a model system. FR suppresses many of the hallmark features that are central to the malignancy of melanoma cells, thereby providing new opportunities for therapeutic intervention. Just as pertussis toxin is used extensively to probe and inhibit the signalling of Gi/o proteins, we anticipate that FR will at least be its equivalent for investigating the biological relevance of Gq.


Subject(s)
Depsipeptides/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Animals , Ardisia/chemistry , Cell Line, Tumor , Depsipeptides/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Melanoma/metabolism , Mice , Models, Molecular , Molecular Structure , Protein Conformation , Protein Isoforms , Signal Transduction , Tail/blood supply , Vasoconstriction/drug effects
10.
J Biol Chem ; 289(43): 29545-57, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25193662

ABSTRACT

All peripheral membrane proteins must negotiate unique constraints intrinsic to the biological interface of lipid bilayers and the cytosol. Phospholipase C-ß (PLC-ß) isozymes hydrolyze the membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) to propagate diverse intracellular responses that underlie the physiological action of many hormones, neurotransmitters, and growth factors. PLC-ß isozymes are autoinhibited, and several proteins, including Gαq, Gßγ, and Rac1, directly engage distinct regions of these phospholipases to release autoinhibition. To understand this process, we used a novel, soluble analog of PIP2 that increases in fluorescence upon cleavage to monitor phospholipase activity in real time in the absence of membranes or detergents. High concentrations of Gαq or Gß1γ2 did not activate purified PLC-ß3 under these conditions despite their robust capacity to activate PLC-ß3 at membranes. In addition, mutants of PLC-ß3 with crippled autoinhibition dramatically accelerated the hydrolysis of PIP2 in membranes without an equivalent acceleration in the hydrolysis of the soluble analog. Our results illustrate that membranes are integral for the activation of PLC-ß isozymes by diverse modulators, and we propose a model describing membrane-mediated allosterism within PLC-ß isozymes.


Subject(s)
Cell Membrane/enzymology , Phospholipase C beta/metabolism , Allosteric Regulation , Animals , Biocatalysis , COS Cells , Chlorocebus aethiops , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Genes, Reporter , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Hydrolysis , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Models, Biological , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/chemistry , Phospholipase C beta/isolation & purification , Protein Structure, Secondary , Protein Structure, Tertiary , Solubility
11.
Chem Biol ; 21(7): 890-902, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036778

ABSTRACT

In spite of the crucial role of heterotrimeric G proteins as molecular switches transmitting signals from G protein-coupled receptors, their selective manipulation with small molecule, cell-permeable inhibitors still remains an unmet challenge. Here, we report that the small molecule BIM-46187, previously classified as pan-G protein inhibitor, preferentially silences Gαq signaling in a cellular context-dependent manner. Investigations into its mode of action reveal that BIM traps Gαq in the empty pocket conformation by permitting GDP exit but interdicting GTP entry, a molecular mechanism not yet assigned to any other small molecule Gα inhibitor to date. Our data show that Gα proteins may be "frozen" pharmacologically in an intermediate conformation along their activation pathway and propose a pharmacological strategy to specifically silence Gα subclasses with cell-permeable inhibitors.


Subject(s)
Cyclohexanes/metabolism , Cyclohexanes/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Pyrazines/metabolism , Pyrazines/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cyclohexanes/chemistry , Dimerization , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Models, Molecular , Permeability , Protein Conformation/drug effects , Pyrazines/chemistry , Signal Transduction/drug effects
12.
Biochemistry ; 52(28): 4810-9, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23777354

ABSTRACT

Multiple extracellular stimuli, such as growth factors and antigens, initiate signaling cascades through tyrosine phosphorylation and activation of phospholipase C-γ (PLC-γ) isozymes. Like most other PLCs, PLC-γ1 is basally autoinhibited by its X-Y linker, which separates the X- and Y-boxes of the catalytic core. The C-terminal SH2 (cSH2) domain within the X-Y linker is the critical determinant for autoinhibition of phospholipase activity. Release of autoinhibition requires an intramolecular interaction between the cSH2 domain and a phosphorylated tyrosine, Tyr783, also located within the X-Y linker. The molecular mechanisms that mediate autoinhibition and phosphorylation-induced activation have not been defined. Here, we describe structures of the cSH2 domain both alone and bound to a PLC-γ1 peptide encompassing phosphorylated Tyr783. The cSH2 domain remains largely unaltered by peptide engagement. Point mutations in the cSH2 domain located at the interface with the peptide were sufficient to constitutively activate PLC-γ1, suggesting that peptide engagement directly interferes with the capacity of the cSH2 domain to block the lipase active site. This idea is supported by mutations in a complementary surface of the catalytic core that also enhanced phospholipase activity.


Subject(s)
Isoenzymes/metabolism , Phospholipase C gamma/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Enzyme Activation , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Models, Molecular , Molecular Sequence Data , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/chemistry , Phosphorylation , Sequence Homology, Amino Acid , src Homology Domains
13.
Int J High Throughput Screen ; 2010(1): 109-126, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-21132089

ABSTRACT

S100B is highly over-expressed in many cancers, including malignant melanoma. In such cancers, S100B binds wild-type p53 in a calcium-dependent manner, sequestering it, and promoting its degradation, resulting in the loss of p53-dependent tumor suppression activities. Therefore, S100B inhibitors may be able to restore wild-type p53 levels in certain cancers and provide a useful therapeutic strategy. In this regard, an automated and sensitive fluorescence polarization competition assay (FPCA) was developed and optimized to screen rapidly for lead compounds that bind Ca(2+)-loaded S100B and inhibit S100B target complex formation. A screen of 2000 compounds led to the identification of 26 putative S100B low molecular weight inhibitors. The binding of these small molecules to S100B was confirmed by nuclear magnetic resonance spectroscopy, and additional structural information was provided by x-ray crystal structures of several compounds in complexes with S100B. Notably, many of the identified inhibitors function by chemically modifying Cys84 in protein. These results validate the use of high-throughput FPCA to facilitate the identification of compounds that inhibit S100B. These lead compounds will be the subject of future optimization studies with the ultimate goal of developing a drug with therapeutic activity for the treatment of malignant melanoma and/or other cancers with elevated S100B.

14.
J Mol Biol ; 396(5): 1227-43, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20053360

ABSTRACT

Structure-based drug design is underway to inhibit the S100B-p53 interaction as a strategy for treating malignant melanoma. X-ray crystallography was used here to characterize an interaction between Ca(2)(+)-S100B and TRTK-12, a target that binds to the p53-binding site on S100B. The structures of Ca(2+)-S100B (1.5-A resolution) and S100B-Ca(2)(+)-TRTK-12 (2.0-A resolution) determined here indicate that the S100B-Ca(2+)-TRTK-12 complex is dominated by an interaction between Trp7 of TRTK-12 and a hydrophobic binding pocket exposed on Ca(2+)-S100B involving residues in helices 2 and 3 and loop 2. As with an S100B-Ca(2)(+)-p53 peptide complex, TRTK-12 binding to Ca(2+)-S100B was found to increase the protein's Ca(2)(+)-binding affinity. One explanation for this effect was that peptide binding introduced a structural change that increased the number of Ca(2+) ligands and/or improved the Ca(2+) coordination geometry of S100B. This possibility was ruled out when the structures of S100B-Ca(2+)-TRTK-12 and S100B-Ca(2+) were compared and calcium ion coordination by the protein was found to be nearly identical in both EF-hand calcium-binding domains (RMSD=0.19). On the other hand, B-factors for residues in EF2 of Ca(2+)-S100B were found to be significantly lowered with TRTK-12 bound. This result is consistent with NMR (15)N relaxation studies that showed that TRTK-12 binding eliminated dynamic properties observed in Ca(2+)-S100B. Such a loss of protein motion may also provide an explanation for how calcium-ion-binding affinity is increased upon binding a target. Lastly, it follows that any small-molecule inhibitor bound to Ca(2+)-S100B would also have to cause an increase in calcium-ion-binding affinity to be effective therapeutically inside a cell, so these data need to be considered in future drug design studies involving S100B.


Subject(s)
Calcium/metabolism , Nerve Growth Factors/chemistry , Nerve Growth Factors/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , S100 Proteins/chemistry , S100 Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , CapZ Actin Capping Protein , Cattle , Crystallography, X-Ray , Fluorescence Polarization , Humans , In Vitro Techniques , Kinetics , Models, Molecular , Multiprotein Complexes , Mutagenesis, Site-Directed , Nerve Growth Factors/genetics , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/genetics , Peptide Fragments , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Thermodynamics
15.
Biochemistry ; 48(26): 6202-12, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19469484

ABSTRACT

Structural studies are part of a rational drug design program aimed at inhibiting the S100B-p53 interaction and restoring wild-type p53 function in malignant melanoma. To this end, structures of three compounds (SBi132, SBi1279, and SBi523) bound to Ca(2+)-S100B were determined by X-ray crystallography at 2.10 A (R(free) = 0.257), 1.98 A (R(free) = 0.281), and 1.90 A (R(free) = 0.228) resolution, respectively. Upon comparison, SBi132, SBi279, and SBi523 were found to bind in distinct locations and orientations within the hydrophobic target binding pocket of Ca(2+)-S100B with minimal structural changes observed for the protein upon complex formation with each compound. Specifically, SBi132 binds nearby residues in loop 2 (His-42, Phe-43, and Leu-44) and helix 4 (Phe-76, Met-79, Ile-80, Ala-83, Cys-84, Phe-87, and Phe-88), whereas SBi523 interacts with a separate site defined by residues within loop 2 (Ser-41, His-42, Phe-43, Leu-44, Glu-45, and Glu-46) and one residue on helix 4 (Phe-87). The SBi279 binding site on Ca(2+)-S100B overlaps the SBi132 and SBi523 sites and contacts residues in both loop 2 (Ser-41, His-42, Phe-43, Leu-44, and Glu-45) and helix 4 (Ile-80, Ala-83, Cys-84, Phe-87, and Phe-88). NMR data, including saturation transfer difference (STD) and (15)N backbone and (13)C side chain chemical shift perturbations, were consistent with the X-ray crystal structures and demonstrated the relevance of all three small molecule-S100B complexes in solution. The discovery that SBi132, SBi279, and SBi523 bind to proximal sites on Ca(2+)-S100B could be useful for the development of a new class of molecule(s) that interacts with one or more of these binding sites simultaneously, thereby yielding novel tight binding inhibitors specific for blocking protein-protein interactions involving S100B.


Subject(s)
Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/chemistry , Nuclear Magnetic Resonance, Biomolecular , S100 Proteins/antagonists & inhibitors , S100 Proteins/chemistry , Animals , Binding Sites , Cattle , Crystallography, X-Ray , Drug Design , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Structure , Nerve Growth Factors/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
16.
J Mol Biol ; 382(1): 56-73, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18602402

ABSTRACT

As part of an effort to inhibit S100B, structures of pentamidine (Pnt) bound to Ca(2+)-loaded and Zn(2+),Ca(2+)-loaded S100B were determined by X-ray crystallography at 2.15 A (R(free)=0.266) and 1.85 A (R(free)=0.243) resolution, respectively. These data were compared to X-ray structures solved in the absence of Pnt, including Ca(2+)-loaded S100B and Zn(2+),Ca(2+)-loaded S100B determined here (1.88 A; R(free)=0.267). In the presence and absence of Zn(2+), electron density corresponding to two Pnt molecules per S100B subunit was mapped for both drug-bound structures. One Pnt binding site (site 1) was adjacent to a p53 peptide binding site on S100B (+/-Zn(2+)), and the second Pnt molecule was mapped to the dimer interface (site 2; +/-Zn(2+)) and in a pocket near residues that define the Zn(2+) binding site on S100B. In addition, a conformational change in S100B was observed upon the addition of Zn(2+) to Ca(2+)-S100B, which changed the conformation and orientation of Pnt bound to sites 1 and 2 of Pnt-Zn(2+),Ca(2+)-S100B when compared to Pnt-Ca(2+)-S100B. That Pnt can adapt to this Zn(2+)-dependent conformational change was unexpected and provides a new mode for S100B inhibition by this drug. These data will be useful for developing novel inhibitors of both Ca(2+)- and Ca(2+),Zn(2+)-bound S100B.


Subject(s)
Calcium/metabolism , Nerve Growth Factors/metabolism , Pentamidine/metabolism , S100 Proteins/metabolism , Zinc/metabolism , Animals , Cattle , Crystallography, X-Ray , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Nerve Growth Factors/chemistry , Pentamidine/chemistry , Protein Structure, Secondary , Rats , S100 Calcium Binding Protein beta Subunit , S100 Proteins/chemistry , Thermodynamics
17.
Biochemistry ; 47(18): 5111-26, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18410126

ABSTRACT

S100A4, also known as mts1, is a member of the S100 family of Ca2+-binding proteins that is directly involved in tumor invasion and metastasis via interactions with specific protein targets, including nonmuscle myosin-IIA (MIIA). Human S100A4 binds two Ca2+ ions with the typical EF-hand exhibiting an affinity that is nearly 1 order of magnitude tighter than that of the pseudo-EF-hand. To examine how Ca2+ modifies the overall organization and structure of the protein, we determined the 1.7 A crystal structure of the human Ca2+-S100A4. Ca2+ binding induces a large reorientation of helix 3 in the typical EF-hand. This reorganization exposes a hydrophobic cleft that is comprised of residues from the hinge region,helix 3, and helix 4, which afford specific target recognition and binding. The Ca2+-dependent conformational change is required for S100A4 to bind peptide sequences derived from the C-terminal portion of the MIIA rod with submicromolar affinity. In addition, the level of binding of Ca2+ to both EF-hands increases by 1 order of magnitude in the presence of MIIA. NMR spectroscopy studies demonstrate that following titration with a MIIA peptide, the largest chemical shift perturbations and exchange broadening effects occur for residues in the hydrophobic pocket of Ca2+-S100A4. Most of these residues are not exposed in apo-S100A4 and explain the Ca2+ dependence of formation of theS100A4-MIIA complex. These studies provide the foundation for understanding S100A4 target recognition and may support the development of reagents that interfere with S100A4 function.


Subject(s)
Calcium/metabolism , Nonmuscle Myosin Type IIA/chemistry , Nonmuscle Myosin Type IIA/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , S100 Proteins/chemistry , S100 Proteins/metabolism , Crystallography, X-Ray , Dimerization , Humans , Models, Molecular , Muscles/chemistry , Muscles/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Quaternary , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Thermodynamics
20.
Biochim Biophys Acta ; 1763(11): 1284-97, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17010455

ABSTRACT

S100B is an EF-hand containing calcium-binding protein of the S100 protein family that exerts its biological effect by binding and affecting various target proteins. A consensus sequence for S100B target proteins was published as (K/R)(L/I)xWxxIL and matches a region in the actin capping protein CapZ (V.V. Ivanenkov, G.A. Jamieson, Jr., E. Gruenstein, R.V. Dimlich, Characterization of S-100b binding epitopes. Identification of a novel target, the actin capping protein, CapZ, J. Biol. Chem. 270 (1995) 14651-14658). Several additional S100B targets are known including p53, a nuclear Dbf2 related (NDR) kinase, the RAGE receptor, neuromodulin, protein kinase C, and others. Examining the binding sites of such targets and new protein sequence searches provided additional potential target proteins for S100B including Hdm2 and Hdm4, which were both found to bind S100B in a calcium-dependent manner. The interaction between S100B and the Hdm2 and/or the Hdm4 proteins may be important physiologically in light of evidence that like Hdm2, S100B also contributes to lowering protein levels of the tumor suppressor protein, p53. For the S100B-p53 interaction, it was found that phosphorylation of specific serine and/or threonine residues reduces the affinity of the S100B-p53 interaction by as much as an order of magnitude, and is important for protecting p53 from S100B-dependent down-regulation, a scenario that is similar to what is found for the Hdm2-p53 complex.


Subject(s)
Calgranulin B/chemistry , Nuclear Proteins/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins/chemistry , S100 Proteins/chemistry , Tumor Suppressor Protein p53/chemistry , Amino Acid Sequence , Calgranulin B/metabolism , Cell Cycle Proteins , Humans , Molecular Sequence Data , Peptides/chemistry , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , S100 Proteins/metabolism , Serine/chemistry , Serine/metabolism , Threonine/chemistry , Threonine/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...