Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Liposome Res ; 26(3): 188-98, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26340033

ABSTRACT

Amylin is a pancreatic hormone involved in the regulation of glucose metabolism and homeostasis. Restoration of the post-prandial and basal levels of human amylin in diabetic individuals is a key in controlling glycemia, controlling glucagon, reducing the insulin dose and increasing satiety, among other physiologic functions. Human amylin has a high propensity to aggregate. We have addressed this issue by designing a liposomal human amylin formulation. Nanoparticles of multilamellar liposomes comprising human amylin were obtained with 53% encapsulation efficiency. The in vitro kinetic release assay shows a biphasic profile. The stabilization of the lipidic nanoparticle against freeze-drying was achieved by using mannitol as a cryoprotectant, as evidenced by morphological characterization. The effectiveness of the human amylin entrapped in lipidic nanoparticles was tested by the measurement of its pharmacological effect in vivo after subcutaneous administration in mice. Collectively these results demonstrate the compatibility of human amylin with the lipidic interface as an effective pharmaceutical delivery system.


Subject(s)
Islet Amyloid Polypeptide/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Humans , Kinetics , Protein Conformation
2.
J Biol Inorg Chem ; 19(6): 839-51, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24557708

ABSTRACT

Conversion of prion protein (PrP) to an altered conformer, the scrapie PrP (PrP(Sc)), is a critical step in the development of transmissible spongiform encephalopathies. Both Cu(II) and nucleic acid molecules have been implicated in this conversion. Full-length PrP can bind up to six copper ions; four Cu(II) binding sites are located in the octarepeat domain (residues 60-91), and His-96 and His-111 coordinate two additional copper ions. Experimental evidence shows that PrP binds different molecules, resulting in diverse cellular signaling events. However, there is little information about the interaction of macromolecular ligands with Cu(II)-bound PrP. Both RNA and DNA sequences can bind PrP, and this interaction results in reciprocal conformational changes. Here, we investigated the interaction of Cu(II) and nucleic acids with amyloidogenic non-octarepeat PrP peptide models (comprising human PrP residues 106-126 and hamster PrP residues 109-149) that retain His-111 as the copper-anchoring residue. The effect of Cu(II) and DNA or RNA sequences in the aggregation, conformation, and toxicity of PrP domains was investigated at low and neutral pH. Circular dichroism and EPR spectroscopy data indicate that interaction of the PrP peptides with Cu(II) and DNA occurs at pH 7. This dual interaction induces conformational changes in the peptides, modulating their aggregation, and affecting the morphology of the aggregated species, resulting in different cytotoxic effects. These results provide new insights into the role of Cu(II) and nucleic acid sequences in the structural conversion and aggregation of PrP, which are both critical events related to prion pathogenesis.


Subject(s)
Copper/chemistry , Metalloproteins/pharmacology , Nucleic Acids/chemistry , Peptides/chemistry , Prions/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Metalloproteins/chemistry , Mice , Structure-Activity Relationship
3.
J Biol Chem ; 287(44): 37206-18, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22918834

ABSTRACT

The accumulation of amyloid fibrils is a feature of amyloid diseases, where cell toxicity is due to soluble oligomeric species that precede fibril formation or are formed by fibril fragmentation, but the mechanism(s) of fragmentation is still unclear. Neutrophil-derived elastase and histones were found in amyloid deposits from patients with different systemic amyloidoses. Neutrophil extracellular traps (NETs) are key players in a death mechanism in which neutrophils release DNA traps decorated with proteins such as elastase and histones to entangle pathogens. Here, we asked whether NETs are triggered by amyloid fibrils, reasoning that because proteases are present in NETs, protease digestion of amyloid may generate soluble, cytotoxic species. We show that amyloid fibrils from three different sources (α-synuclein, Sup35, and transthyretin) induced NADPH oxidase-dependent NETs in vitro from human neutrophils. Surprisingly, NET-associated elastase digested amyloid fibrils into short species that were cytotoxic for BHK-21 and HepG2 cells. In tissue sections from patients with primary amyloidosis, we also observed the co-localization of NETs with amyloid deposits as well as with oligomers, which are probably derived from elastase-induced fibril degradation (amyloidolysis). These data reveal that release of NETs, so far described to be elicited by pathogens, can also be triggered by amyloid fibrils. Moreover, the involvement of NETs in amyloidoses might be crucial for the production of toxic species derived from fibril fragmentation.


Subject(s)
Amyloid/physiology , Chromatin/metabolism , Neutrophils/pathology , Acetophenones/pharmacology , Amyloid/chemistry , Amyloid/genetics , Amyloid Neuropathies, Familial/enzymology , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Amyloidosis/enzymology , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Biomarkers/metabolism , Cell Nucleus/metabolism , Cell Survival/drug effects , Chromatin/enzymology , Cricetinae , Extracellular Space/enzymology , Extracellular Space/metabolism , Hep G2 Cells , Humans , Immunoglobulin Light-chain Amyloidosis , Lung/enzymology , Lung/metabolism , Lung/pathology , Mutation, Missense , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Onium Compounds/pharmacology , Pancreatic Elastase , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Prealbumin/chemistry , Prealbumin/genetics , Prealbumin/physiology , Protein Structure, Quaternary , Proteolysis , Reactive Oxygen Species/metabolism , Skin/enzymology , Skin/metabolism , Skin/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/physiology
4.
J Biol Chem ; 287(33): 28152-62, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22715097

ABSTRACT

Over 50% of all human cancers lose p53 function. To evaluate the role of aggregation in cancer, we asked whether wild-type (WT) p53 and the hot-spot mutant R248Q could aggregate as amyloids under physiological conditions and whether the mutant could seed aggregation of the wild-type form. The central domains (p53C) of both constructs aggregated into a mixture of oligomers and fibrils. R248Q had a greater tendency to aggregate than WT p53. Full-length p53 aggregated into amyloid-like species that bound thioflavin T. The amyloid nature of the aggregates was demonstrated using x-ray diffraction, electron microscopy, FTIR, dynamic light scattering, cell viabilility assay, and anti-amyloid immunoassay. The x-ray diffraction pattern of the fibrillar aggregates was consistent with the typical conformation of cross ß-sheet amyloid fibers with reflexions of 4.7 Å and 10 Å. A seed of R248Q p53C amyloid oligomers and fibrils accelerated the aggregation of WT p53C, a behavior typical of a prion. The R248Q mutant co-localized with amyloid-like species in a breast cancer sample, which further supported its prion-like effect. A tumor cell line containing mutant p53 also revealed massive aggregation of p53 in the nucleus. We conclude that aggregation of p53 into a mixture of oligomers and fibrils sequestrates the native protein into an inactive conformation that is typical of a prionoid. This prion-like behavior of oncogenic p53 mutants provides an explanation for the negative dominance effect and may serve as a potential target for cancer therapy.


Subject(s)
Amyloid/chemistry , Mutation, Missense , Neoplasms/chemistry , Prions , Protein Multimerization , Tumor Suppressor Protein p53/chemistry , Amino Acid Substitution , Amyloid/genetics , Amyloid/metabolism , Benzothiazoles , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Protein Structure, Quaternary , Protein Structure, Secondary , Thiazoles/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , X-Ray Diffraction
5.
Int J Mol Sci ; 14(3): 5284-311, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23466880

ABSTRACT

Transthyretin (TTR) is a homotetrameric protein that circulates in plasma and cerebral spinal fluid (CSF) whose aggregation into amyloid fibrils has been associated with at least two different amyloid diseases: senile systemic amyloidosis (SSA) and familial amyloid polyneuropathy (FAP). In SSA aggregates are composed of WT-TTR, while in FAP more than 100 already-described variants have been found in deposits. Until now, TTR-related diseases have been untreatable, although a new drug called Tafamidis has been approved only in Europe to specifically treat V30M patients. Thus, new strategies are still necessary to treat FAP caused by other variants of TTR. TTR has two channels in the dimer interface that bind to the hormone thyroxin and that have been used to accommodate anti-amyloidogenic compounds. These compounds stabilize the tetramers, rendering TTR less amyloidogenic. Here, we investigated the effects of three non-steroidal anti-inflammatory compounds-sulindac (SUL), indomethacin (IND) and lumiracoxib (LUM)-as tetramer stabilizers and aggregation inhibitors. WT-TTR and the very aggressive TTR variant L55P were used as models. These compounds were able to stabilize TTR against high hydrostatic pressure (HHP), increasing the ΔGf by several kcal. They were also effective in inhibiting WT-TTR and L55P acid- or HHP-induced aggregation; in particular, LUM and IND were very effective, inhibiting almost 100% of the aggregation of both proteins under certain conditions. The species formed when aggregation was performed in the presence of these compounds were much less toxic to cells in culture. The crystal structures of WT-TTR bound to the three compounds were solved at high resolution, allowing the identification of the relevant protein:drug interactions. We discuss here the ligand-binding features of LUM, IND and SUL to TTR, emphasizing the critical interactions that render the protein more stable and less amyloidogenic.

6.
Biochemistry ; 51(27): 5402-13, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22691027

ABSTRACT

A misfolded form of the prion protein (PrP) is the primary culprit in mammalian prion diseases. It has been shown that nucleic acids catalyze the misfolding of cellular PrP into a scrapie-like conformer. It has also been observed that the interaction of PrP with nucleic acids is nonspecific and that the complex can be toxic to cultured cells. No direct correlation has yet been drawn between changes in PrP structure and toxicity due to nucleic acid binding. Here we asked whether different aggregation, stability, and toxicity effects are detected when nonrelated DNA sequences interact with recombinant PrP. Using spectroscopic techniques to analyze PrP tertiary and secondary structure and cellular assays to assess toxicity, we found that rPrP-DNA interactions lead to different aggregated species, depending on the sequence and size of the oligonucleotide tested. A 21-mer DNA sequence (D67) induced higher levels of aggregation and also dissimilar structural changes in rPrP, compared to binding to oligonucleotides with the same length and different nucleotide sequences or different GC contents. The rPrP-D67 complex induced significant cell dysfunction, which appears to be correlated with the biophysical properties of the complex. Although sequence specificity is not apparent for PrP-nucleic acid interactions, we believe that particular nucleic acid patterns, possibly related to GC content, oligonucleotide length, and structure, govern PrP recognition. Understanding the structural and cellular effects observed for PrP-nucleic acid complexes may shed light on the still mysterious pathology of the prion protein.


Subject(s)
Cytotoxins/chemistry , Cytotoxins/toxicity , DNA/metabolism , Prions/chemistry , Prions/toxicity , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Cytotoxins/metabolism , DNA/genetics , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Prions/metabolism , Protein Binding , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Solubility
7.
Mater Sci Eng C Mater Biol Appl ; 79: 802-811, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28629083

ABSTRACT

Insulin-loaded calcium phosphate nanoparticles have been proposed as a potential drug delivery system for the oral treatment of diabetes and to stimulate bone cell proliferation and bone mineralization. The kinetics of insulin incorporation onto hydroxyapatite (HA) and Sr (SrHA)- and Zn (ZnHA)-substituted hydroxyapatite nanoparticles was investigated using X-ray photoelectron spectroscopy (XPS), Fourier transform infrared (FTIR) spectroscopy, zeta potential measurements and circular dichroism (CD) spectroscopy. The increase in insulin concentration on HA, SrHA and ZnHA was a typical physical adsorption process controlled by electrostatic forces and followed a Freundlich isotherm model. Zn substitution enhanced the capacity of the apatite surface to adsorb insulin, whereas Sr substitution inhibited insulin uptake. The surface stoichiometry and mesopore specific area induced by Zn and Sr substitution are proposed as the main causes of the difference in insulin adsorption. Despite the weak interaction between insulin and the apatite surface, the CD spectra revealed a decrease in the insulin ellipticity when the protein was adsorbed on the HA, SrHA and ZnHA nanoparticles. A reduction in alpha-helical structures and an increase in beta sheets were observed when insulin interacted with the HA surface. A less pronounced effect was found for ZnHA, for which a subtle decrease in alpha-helical structures was followed by an increase in turn structures. Interaction with the SrHA surface did not change the native insulin conformation. In vitro cell culture experiments lasting 24h using F-OST stromal cells showed that the insulin loaded on HA and ZnHA did not affect cell proliferation but the insulin loaded on SrHA improved cell proliferation. These results suggest that the stability of the native protein conformation is an important factor to consider when cells interact with insulin adsorbed on metal-substituted HA surfaces.


Subject(s)
Durapatite/chemistry , Adsorption , Insulin , Spectroscopy, Fourier Transform Infrared , Strontium , Zinc
8.
Int J Biochem Cell Biol ; 38(1): 110-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16183325

ABSTRACT

The C-domain of troponin C, the Ca(2+)-binding subunit of the troponin complex, has two high-affinity sites for Ca(2+) that also bind Mg(2+) (Ca(2+)/Mg(2+) sites), whereas the N-domain has two low-affinity sites for Ca(2+). Two more sites that bind Mg(2+) with very low affinity (K(a)<10(3)M(-1)) have been detected by several laboratories but have not been localized or studied in any detail. Here we investigated the effects of Ca(2+) and Mg(2+) binding to isolated C-domain, focusing primarily on low-affinity sites. Since TnC has no Trp residues, we utilized a mutant with Phe 154 replaced by Trp (F154W/C-domain). As expected from previous reports, the changes in Trp fluorescence revealed different conformations induced by the addition of Ca(2+) or Mg(2+) (Ca(2+)/Mg(2+) sites). Exposure of hydrophobic surfaces of F154W/C-domain was monitored using the fluorescence intensity of bis-anilino naphthalene sulfonic acid. Unlike the changes reported by Trp, the increments in bis-ANS fluorescence were much greater (4.2-fold) when Ca(2+)+Mg(2+) were both present or when Ca(2+) was present at high concentration. Bis-ANS fluorescence increased as a function of [Ca(2+)] in two well-defined steps: one at low [Ca(2+)], consistent with the Ca(2+)/Mg(2+) sites (K(a) approximately 1.5 x 10(6)M(-1)), and one of much lower affinity (K(a) approximately 52.3M(-1)). Controls were performed to rule out artifacts due to aggregation, high ionic strength and formation of the bis-ANS-TnC complex itself. With a low concentration of Ca(2+) (0.6mM) to occupy the Ca(2+)/Mg(2+) sites, a large increase in bis-ANS binding also occurred as Mg(2+) occupied a class of low-affinity sites (K(a) approximately 59 M(-1)). In skinned fibers, a high concentration of Mg(2+) (10-44 mM) caused TnC to dissociate from the thin filament. These data provide new evidence for a class of weak binding sites for divalent cations. They are located in the C-domain, lead to exposure of a large hydrophobic surface, and destabilize the binding of TnC to the regulatory complex even when sites III and IV are occupied.


Subject(s)
Amino Acid Substitution , Calcium/chemistry , Magnesium/chemistry , Multiprotein Complexes/chemistry , Point Mutation , Troponin C/chemistry , Animals , Binding Sites/genetics , Calcium/metabolism , Chickens , Hydrophobic and Hydrophilic Interactions , Magnesium/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Spectrometry, Fluorescence/methods , Troponin C/genetics , Troponin C/metabolism
9.
Biophys Chem ; 180-181: 135-44, 2013.
Article in English | MEDLINE | ID: mdl-23974296

ABSTRACT

Amylin is a pancreatic hormone co-secreted with insulin. Human amylin has been shown to form dimers and exhibit high propensity for amyloid fibril formation. We observed the ability of the water-soluble murine amylin to aggregate in water resulting in an insoluble material with Thioflavin T binding properties. Infrared spectroscopy analysis revealed beta-sheet components in the aggregated murine amylin. Morphological analysis by transmission electron microscopy and atomic force microscopy provided access to the fibril nature of the murine amylin aggregate which is similar to amyloid fibrils from human amylin. X-ray diffraction of the murine amylin fibrils showed peaks at 4.7Å and 10Å, a fingerprint for amyloid fibrils. Electron spray ionization-ion mobility spectroscopy-mass spectrometry (ESI-IMS-MS) analysis and crosslinking assays revealed self-association intermediates of murine amylin into high order oligomeric assemblies. These data demonstrate the stepwise association mechanism of murine amylin into stable oligomers, which ultimately converges to its organization into amyloid fibrils.


Subject(s)
Amyloid/metabolism , Islet Amyloid Polypeptide/metabolism , Amyloid/chemistry , Animals , Benzothiazoles , Humans , Islet Amyloid Polypeptide/chemistry , Mice , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Polymerization , Protein Binding , Protein Structure, Secondary , Solutions/chemistry , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Infrared , Thiazoles/chemistry , Thiazoles/metabolism
10.
J Mol Biol ; 405(1): 254-73, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21050861

ABSTRACT

Parkinson's disease (PD) is a movement disorder characterized by the loss of dopaminergic neurons in the substantia nigra and the formation of intraneuronal inclusions called Lewy bodies, which are composed mainly of α-synuclein (α-syn). Selegiline (Sel) is a noncompetitive monoamino oxidase B inhibitor that has neuroprotective effects and has been administered to PD patients as monotherapy or in combination with l-dopa. Besides its known effect of increasing the level of dopamine (DA) by monoamino oxidase B inhibition, Sel induces other effects that contribute to its action against PD. We evaluated the effects of Sel on the in vitro aggregation of A30P and wild-type α-syn. Sel delays fibril formation by extending the lag phase of aggregation. In the presence of Sel, electron microscopy reveals amorphous heterogeneous aggregates, including large annular species, which are innocuous to a primary culture enriched in dopaminergic neurons, while their age-matched counterparts are toxic. The inhibitory effect displayed by Sel is abolished when seeds (small fibril pieces) are added to the aggregation reaction, reinforcing the hypothesis that Sel interferes with early nuclei formation and, to a lesser extent, with fibril elongation. NMR experiments indicate that Sel does not interact with monomeric α-syn. Interestingly, when added in combination with DA (which favors the formation of toxic protofibrils), Sel overrides the inhibitory effect of DA and favors fibrillation. Additionally, Sel blocks the formation of smaller toxic aggregates by perturbing DA-dependent fibril disaggregation. These effects might be beneficial for PD patients, since the sequestration of protofibrils into fibrils or the inhibition of fibril dissociation could alleviate the toxic effects of protofibrils on dopaminergic neurons. In nondopaminergic neurons, Sel might slow the fibrillation, giving rise to the formation of large nontoxic aggregates.


Subject(s)
Antiparkinson Agents/metabolism , Selegiline/metabolism , alpha-Synuclein/metabolism , Humans , Magnetic Resonance Spectroscopy , Microscopy, Electron , Models, Biological , Protein Denaturation , alpha-Synuclein/ultrastructure
11.
Biochemistry ; 46(25): 7353-64, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17542560

ABSTRACT

The head of the P22 bacteriophage is interrupted by a unique dodecameric portal vertex that serves as a conduit for the entrance and exit of the DNA. Here, the in vitro unfolding/refolding processes of the portal protein of P22 were investigated at different temperatures (1, 25, and 37 degrees C) through the use of urea and high hydrostatic pressure (HHP) combined with spectroscopic techniques. We have characterized an intermediate species, IU, which forms at 25 degrees C during unfolding or refolding of the portal protein in 2-4 M urea. IU readily forms amorphous aggregates, rendering the folding process irreversible. On the other hand, at 1 degrees C, a two-state process is observed (DeltaGf = -2.2 kcal/mol). When subjected to HHP at 25 or 37 degrees C, the portal monomer undergoes partial denaturation, also forming an intermediate species, which we call IP. IP also tends to aggregate but, differently from IU, aggregates into a ring-like structure as seen by size-exclusion chromatography and electron microscopy. Again, at 1 degrees C the unfolding induced by HHP proved to be reversible, with DeltaGf = -2.4 kcal/mol and DeltaV = 72 mL/mol. Interestingly, at 25 degrees C, the binding of the hydrophobic probe bis-ANS to the native portal protein destabilizes it and completely blocks its aggregation under HHP. These data are relevant to the process by which the portal protein assembles into dodecamers in vivo, since species such as IP must prevail over IU in order to guarantee the proper ring formation.


Subject(s)
Bacteriophage P22/physiology , Protein Folding , Viral Proteins/physiology , Chromatography, Gel , Circular Dichroism , Dose-Response Relationship, Drug , Hydrostatic Pressure , Light , Models, Chemical , Protein Conformation , Protein Denaturation/drug effects , Protein Renaturation/drug effects , Protein Structure, Tertiary , Scattering, Radiation , Temperature , Urea/pharmacology , Viral Proteins/chemistry , Viral Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL