Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 223(5)2024 May 06.
Article in English | MEDLINE | ID: mdl-38530280

ABSTRACT

Most mitochondrial proteins originate from the cytosol and require transport into the organelle. Such precursor proteins must be unfolded to pass through translocation channels in mitochondrial membranes. Misfolding of transported proteins can result in their arrest and translocation failure. Arrested proteins block further import, disturbing mitochondrial functions and cellular proteostasis. Cellular responses to translocation failure have been defined in yeast. We developed the cell line-based translocase clogging model to discover molecular mechanisms that resolve failed import events in humans. The mechanism we uncover differs significantly from these described in fungi, where ATPase-driven extraction of blocked protein is directly coupled with proteasomal processing. We found human cells to rely primarily on mitochondrial factors to clear translocation channel blockage. The mitochondrial membrane depolarization triggered proteolytic cleavage of the stalled protein, which involved mitochondrial protease OMA1. The cleavage allowed releasing the protein fragment that blocked the translocase. The released fragment was further cleared in the cytosol by VCP/p97 and the proteasome.


Subject(s)
Metalloendopeptidases , Mitochondria , Protein Transport , Humans , Endopeptidases , Mitochondria/metabolism , Proteasome Endopeptidase Complex , Proteolysis , Metalloendopeptidases/metabolism
2.
Toxicol In Vitro ; 88: 105556, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36681286

ABSTRACT

Metal-based agents in cancer therapy, like cisplatin and its derivates, have established clinical applications but also can induce serious side effects. Thus, metallotherapeutic alternatives for platinum derivatives are developed and intensively studied. Platinum is replaced by several transition metals including gold. Especially gold (III) complexes can have the same square-planar structure and are isoelectric with platinum (II). Hence, they are developed as potential anti-cancer drugs. Thus, our group projected and developed a group of novel cyanide-based gold (III) complexes. Within this work, we aimed to characterize the safety and effectivity of one of them, TGS 121. TGS 121 in our preliminary work was selective for Ras-hyperactivated cells. Here we studied the effects of the novel complex in cancerous Ras-3 T3 and non-cancerous NIH-3 T3 cells. The complex TGS 121 turned out to be non-toxic for NIH-3 T3 cells and to induce death and alternations in Ras-hyperactivated cells. We found induction of ER stress, mitochondria swelling, proteasome inhibition, and cell cycle block. Moreover, TGS 121 inhibited cell migration and induced the accumulation of perinuclear organelles that was secondary to proteasome inhibition. Results presented in this report suggest that stable gold-cyanide TGS 121 complex is non-toxic, with a targeted mechanism of action and it is promising in anticancer drug discovery.


Subject(s)
Antineoplastic Agents , Proteasome Endopeptidase Complex , Platinum/chemistry , Cyanides/toxicity , Antineoplastic Agents/toxicity , Antineoplastic Agents/chemistry , Gold/toxicity , Gold/chemistry , Cell Line, Tumor
3.
Mol Metab ; 67: 101659, 2023 01.
Article in English | MEDLINE | ID: mdl-36529318

ABSTRACT

Abnormalities that characterize the pathophysiology of type 2 diabetes (T2D) include deficiencies of ß-cells and the expansion of α-cells in pancreatic islets, manifested by lower insulin release and glucagon oversecretion. The molecular mechanisms that determine intra-islet interactions between pancreatic α- and ß-cells are still not fully understood. The present study showed that stearoyl-coenzyme A (CoA) desaturase 1 (SCD1), an enzyme that is implicated in fatty acid metabolism, serves as a checkpoint in the control of endocrine cell equilibrium in pancreatic islets. Our data showed that SCD1 activity is essential for proper α-cell and ß-cell lineage determination during morphogenesis of the pancreas and the maintenance of mature ß-cell identity. The inhibition of SCD1 expression/activity led to both a decrease in the expression of ß-cell signature genes (e.g., Pdx1, Nkx6.1, MafA, and Neurod1, among others) and induction of the expression of the dedifferentiation marker Sox9 in mature pancreatic islets. The transcriptional repression of Pdx1 and MafA in SCD1-deficient ß-cells was related to the excessive methylation of promoter regions of these transcription factors. In contrast, SCD1 ablation favored the formation of α-cells over ß-cells throughout pancreas organogenesis and did not compromise α-cell identity in adult pancreatic islets. Such molecular changes that were caused by SCD1 downregulation resulted in the mislocalization of α-cells within the core of islets and increased the ratio of pancreatic α- to ß-cell mass. This was followed by islet dysfunction, including impairments in glucose-stimulated insulin release, simultaneously with elevations of basal glucagon secretion. Altogether, these findings provide additional mechanistic insights into the role of SCD1 in the pathogenesis of T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Secreting Cells , Islets of Langerhans , Mice , Animals , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Glucagon-Secreting Cells/metabolism , Morphogenesis
4.
Biochim Biophys Acta Gen Subj ; 1866(6): 130132, 2022 06.
Article in English | MEDLINE | ID: mdl-35307509

ABSTRACT

BACKGROUND: The interaction of N-terminal extension of the myosin A1 essential light chain (A1 ELC) with actin is receiving increasing attention as a target in utilizing synthetic A1 ELC N-terminal-derived peptides in cardiac dysfunction therapy. METHODS: To elucidate the mechanism by which these peptides regulate actin-myosin interaction, here we have investigated their effects on the myosin subfragment 1 (S1)-induced polymerization of G-actin. RESULTS: The MLCFpep and MLCSpep peptides spanning the 3-12 of A1 ELC sequences from fast and slow skeletal muscle, respectively, increased the rate of actin polymerization not only by S1(A2) but also the rate of S1(A1)-induced actin polymerization, suggesting that they did not interfere with the direct binding of A1 ELC with actin. The efficiency of actin polymerization in the presence of the N-terminal ELC peptides depended on their sequence. Substitution of aspartic acid for neutral asparagine at position 5 of MLCFpep dramatically enhanced its ability to stimulate S1-induced polymerization and enabled it to initiate polymerization of G-actin in the absence of S1. CONCLUSIONS: These and other results presented in this work suggest that the modulation of myosin motor activity by N-terminal ELC peptides is exerted through a change in actin filament conformation rather than through blocking the A1 ELC-actin interaction. GENERAL SIGNIFICANCE: The results imply the possibility of enhancing therapeutic effects of these peptides by modifications of their sequence.


Subject(s)
Actins , Myosin Light Chains , Actins/metabolism , Muscle, Skeletal/metabolism , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Myosin Subfragments/chemistry , Myosin Subfragments/metabolism
5.
Cell Commun Signal ; 19(1): 116, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34801048

ABSTRACT

BACKGROUND: Wolfram syndrome (WFS) is a rare autosomal recessive syndrome in which diabetes mellitus and neurodegenerative disorders occur as a result of Wolframin deficiency and increased ER stress. In addition, WFS1 deficiency leads to calcium homeostasis disturbances and can change mitochondrial dynamics. The aim of this study was to evaluate protein levels and changes in gene transcription on human WFS cell model under experimental ER stress. METHODS: We performed transcriptomic and proteomic analysis on WFS human cell model-skin fibroblasts reprogrammed into induced pluripotent stem (iPS) cells and then into neural stem cells (NSC) with subsequent ER stress induction using tunicamycin (TM). Results were cross-referenced with publicly available RNA sequencing data in hippocampi and hypothalami of mice with WFS1 deficiency. RESULTS: Proteomic analysis identified specific signal pathways that differ in NSC WFS cells from healthy ones. Next, detailed analysis of the proteins involved in the mitochondrial function showed the down-regulation of subunits of the respiratory chain complexes in NSC WFS cells, as well as the up-regulation of proteins involved in Krebs cycle and glycolysis when compared to the control cells. Based on pathway enrichment analysis we concluded that in samples from mice hippocampi the mitochondrial protein import machinery and OXPHOS were significantly down-regulated. CONCLUSIONS: Our results show the functional and morphological secondary mitochondrial damage in patients with WFS. Video Abstract.


Subject(s)
Wolfram Syndrome
6.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166209, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34246750

ABSTRACT

Tauopathies, including Alzheimer's disease (AD), are manifested by the deposition of well-characterized amyloid aggregates of Tau protein in the brain. However, it is rather unlikely that these aggregates constitute the major form of Tau responsible for neurodegenerative changes. Currently, it is postulated that the intermediates termed as soluble oligomers, assembled on the amyloidogenic pathway, are the most neurotoxic form of Tau. However, Tau oligomers reported so far represent a population of poorly characterized, heterogeneous and unstable assemblies. In this study, to obtain the oligomers, we employed the aggregation-prone K18 fragment of Tau protein with deletion of Lys280 (K18Δ280) linked to a hereditary tauopathy. We have described a new procedure of inducing aggregation of mutated K18 which leads either to the formation of nontoxic amyloid fibrils or neurotoxic globular oligomers, depending on its phosphorylation status. We demonstrate that PKA-phosphorylated K18Δ280 oligomers are toxic to hippocampal neurons, which is manifested by loss of dendritic spines and neurites, and impairment of cell-membrane integrity leading to cell death. We also show that N1, the soluble N-terminal fragment of prion protein (PrP), protects neurons from the oligomers-induced cytotoxicity. Our findings support the hypothesis on the neurotoxicity of Tau oligomers and neuroprotective role of PrP-derived fragments in AD and other tauopathies. These observations could be useful in the development of therapeutic strategies for these diseases.


Subject(s)
Neurons/pathology , Prion Proteins/metabolism , Protein Aggregation, Pathological/pathology , Tauopathies/pathology , tau Proteins/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/pathology , Humans , Phosphorylation , Primary Cell Culture , Prion Proteins/genetics , Prion Proteins/isolation & purification , Protein Aggregation, Pathological/genetics , Protein Binding , Rats , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tauopathies/genetics , tau Proteins/genetics , tau Proteins/isolation & purification
7.
Toxicol In Vitro ; 73: 105144, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33722735

ABSTRACT

Podophyllotoxin (PPT) is an antimitotic drug used topically in the treatment of anogenital warts. Due to its toxicity it cannot be administered systemically as an anticancer agent. However, modified PPT derivatives such as etoposide and teniposide are used clinically as systemic agents. Thus, we invented novel PPT derivative KL3 that was synthesized by photocyclization. Earlier we have shown that KL3 has an anticancer effect in various cell lines. Here we compared the toxicity of KL3 vs PPT on non-cancerous normal human keratinocytes (HaCaT) and peripheral blood mononuclear cells (PBMC) showing that KL3 is less toxic than PPT to non-cancerous cells. At concentrations that neither induced cell death, nor affected cell cycle, KL3 in HaCaT cells evoked transient ultrastructural features of ER stress, swelling of mitochondria and elongation of cytoplasmic processes. Those changes partially reversed with prolonged incubation while features of autophagy were induced. PPT in equivalent concentrations induced HaCaT cell death by cell cycle arrest, intrinsic apoptosis and finally disintegration of cell membranes followed by secondary necrosis. In conclusion, we show that the KL3 derivative of PPT in contrast to PPT allows repair of normal keratinocytes and triggers mechanisms that restore non-tumor cell homeostasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , HaCaT Cells , Humans , Leukocytes, Mononuclear/drug effects , Microscopy, Electron, Transmission
8.
Int J Mol Sci ; 22(2)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477664

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is a heritable neurodegenerative disease that displays great genetic heterogeneity. The genes and mutations that underlie this heterogeneity have been extensively characterized by molecular genetics. However, the molecular pathogenesis of the vast majority of CMT subtypes remains terra incognita. Any attempts to perform experimental therapy for CMT disease are limited by a lack of understanding of the pathogenesis at a molecular level. In this study, we aim to identify the molecular pathways that are disturbed by mutations in the gene encoding GDAP1 using both yeast and human cell, based models of CMT-GDAP1 disease. We found that some mutations in GDAP1 led to a reduced expression of the GDAP1 protein and resulted in a selective disruption of the Golgi apparatus. These structural alterations are accompanied by functional disturbances within the Golgi. We screened over 1500 drugs that are available on the market using our yeast-based CMT-GDAP1 model. Drugs were identified that had both positive and negative effects on cell phenotypes. To the best of our knowledge, this study is the first report of the Golgi apparatus playing a role in the pathology of CMT disorders. The drugs we identified, using our yeast-based CMT-GDAP1 model, may be further used in translational research.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Golgi Apparatus/genetics , Nerve Tissue Proteins/genetics , trans-Golgi Network/genetics , Charcot-Marie-Tooth Disease/pathology , Genetic Heterogeneity , Golgi Apparatus/pathology , HeLa Cells , Humans , Models, Genetic , Mutation/genetics , Pedigree , Structure-Activity Relationship , Yeasts/genetics
9.
Biochim Biophys Acta Biomembr ; 1862(9): 183337, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32380169

ABSTRACT

A causative agent of Alzheimer's disease (AD) is a short amphipathic peptide called amyloid beta (Aß). Aß monomers undergo structural changes leading to their oligomerization or fibrillization. The monomers as well as all aggregated forms of Aß, i.e., oligomers, and fibrils, can bind to biological membranes, thereby modulating membrane mechanical properties. It is also known that some isoforms of the large-conductance calcium-activated potassium (BKCa) channel, including the mitochondrial BKCa (mitoBKCa) channel, respond to mechanical changes in the membrane. Here, using the patch-clamp technique, we investigated the impact of full-length Aß (Aß1-42) and its fragment, Aß25-35, on the activity of mitoBKCa channels. We found that all forms of Aß inhibited the activity of the mitoBKCa channel in a concentration-dependent manner. Since monomers, oligomers, and fibrils of Aß exhibit different molecular characteristics and structures, we hypothesized that the inhibition was not due to direct peptide-protein interactions but rather to membrane-binding of the Aß peptides. Our findings supported this hypothesis by showing that Aß peptides block mitoBKCa channels irrespective of the side of the membrane to which they are applied. In addition, we found that the enantiomeric peptide, D-Aß1-42, demonstrated similar inhibitory activity towards mitoBKCa channels. As a result, we proposed a general model in which all Aß forms i.e., monomers, oligomers, and amyloid fibrils, contribute to the progression of AD by exerting a modulatory effect on mechanosensitive membrane components.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Potassium Channels, Calcium-Activated/economics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/pharmacology , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/genetics , Humans , Mitochondria/drug effects , Mitochondria/genetics , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/genetics
10.
PLoS One ; 14(6): e0218975, 2019.
Article in English | MEDLINE | ID: mdl-31247048

ABSTRACT

Bovine serum albumin (BSA) is often employed as a proteinaceous component for synthesis of luminescent protein-stabilized gold nanoclusters (AuNC): intriguing systems with many potential applications. Typically, the formation of BSA-AuNC conjugate occurs under strongly alkaline conditions. Due to the sheer complexity of intertwined chemical and structural transitions taking place upon BSA-AuNC formation, the state of albumin enveloping AuNCs remains poorly characterized. Here, we study the conformational properties of BSA bound to AuNCs using an array of biophysical tools including vibrational spectroscopy, circular dichroism, fluorescence spectroscopy and trypsin digestion. The alkaline conditions of BSA-AuNC self-assembly appear to be primary responsible for the profound irreversible disruption of tertiary contacts, partial unfolding of native α-helices, hydrolysis of disulfide bonds and the protein becoming vulnerable to trypsin digestion. Further unfolding of BSA-AuNC by guanidinium hydrochloride (GdnHCl) is fully reversible equally in terms of albumin's secondary structure and conjugate's luminescent properties. This suggests that binding to AuNCs traps the albumin molecule in a state that is both partly disordered and refractory to irreversible misfolding. Indeed, when BSA-AuNC is subjected to conditions favoring self-association of BSA into amyloid-like fibrils, the buildup of non-native ß-sheet conformation is less pronounced than in a control experiment with unmodified BSA. Unexpectedly, BSA-AuNC reveals a tendency to self-assemble into giant twisted superstructures of micrometer lengths detectable with transmission electron microscopy (TEM), a property absent in unmodified BSA. The process is accompanied by ordering of bound AuNCs into elongated streaks and simultaneous decrease in fluorescence intensity. The newly discovered self-association pathway appears to be specifically accessible to protein molecules with a certain restriction on structural dynamics which in the case of BSA-AuNC arises from binding to metal nanoclusters. Our results have been discussed in the context of mechanisms of protein misfolding and applications of BSA-AuNC.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Serum Albumin, Bovine/chemistry , Amino Acid Sequence , Animals , Cattle , Circular Dichroism , Metal Nanoparticles/ultrastructure , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Models, Molecular , Protein Aggregates , Protein Conformation , Protein Denaturation , Protein Stability , Serum Albumin, Bovine/genetics , Serum Albumin, Bovine/ultrastructure , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman
11.
PLoS One ; 13(7): e0201182, 2018.
Article in English | MEDLINE | ID: mdl-30024984

ABSTRACT

Amyloid aggregates of Tau protein have been implicated in etiology of many neurodegenerative disorders including Alzheimer's disease (AD). When amyloid growth is induced by seeding with preformed fibrils assembled from the same protein, structural characteristics of the seed are usually imprinted in daughter generations of fibrils. This so-called conformational memory effect may be compromised when the seeding involves proteins with non-identical sequences leading to the emergence of distinct structural variants of fibrils (amyloid 'strains'). Here, we investigate cross-seeding of full-length human Tau (FL Tau) with fibrils assembled from K18 and K18ΔK280 fragments of Tau in the presence of poly-L-glutamate (poly-Glu) as an enhancer of Tau aggregation. To study cross-seeding between Tau polypeptides and the role of the conformational memory effect in induction of Tau amyloid polymorphism, kinetic assays, transmission electron microscopy, infrared spectroscopy and limited proteolysis have been employed. The fastest fibrillization was observed for FL Tau monomers seeded with preformed K18 amyloid yielding daughter fibrils with unique trypsin digestion patterns. Morphological features of daughter FL Tau fibrils induced by K18 and K18ΔK280 seeds were reminiscent of the mother fibrils (i.e. straight paired fibrils and paired helical filaments (PHFs), respectively) but disappeared in the following generations which became similar to unpaired FL Tau amyloid fibrils formed de novo. The structural evolution observed in our study was accompanied by disappearance of the unique proteolysis profile originated from K18. Our findings may have implications for understanding molecular mechanisms of the emergence and stability of Tau amyloid strains.


Subject(s)
Amyloid/metabolism , tau Proteins/metabolism , Amyloid/chemistry , Amyloid/genetics , Amyloid/ultrastructure , Escherichia coli , Humans , Kinetics , Polyglutamic Acid/genetics , Polyglutamic Acid/metabolism , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Multimerization , Proteolysis , Trypsin/chemistry , Trypsin/metabolism , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/ultrastructure
12.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2143-2153, 2018 06.
Article in English | MEDLINE | ID: mdl-29604335

ABSTRACT

Soluble form of the prion protein (PrP) has been previously shown to interact with amyloid-ß (Aß) peptides, suppressing their fibrillization as well as toxicity, which indicates that this protein may play a protective role in Alzheimer's disease (AD). The shortest known PrP fragment retaining all of these properties corresponds to physiologically generated proteolytic polypeptide PrP23-110/111, called N1. Here we have identified two N1-derived synthetic peptides, encompassing residues 23-50 (PrP23-50) and 90-112 (PrP90-112), which bind to Aß1-42 protofibrillar oligomers as well as amyloid fibrils. We found that, akin to N1, the abovementioned synthetic peptides not only reduce the initial rate of Aß fibrillization, but also alter the aggregation pathway of Aß, inhibiting formation of protofibrillar oligomers and facilitating amorphous aggregation. Furthermore, our data show that N1, PrP23-50 and PrP90-112 protect cultured hippocampal neurons from neurotoxic effects of Aß oligomers, preventing oligomers-induced retraction of neurites and loss of cell membrane integrity. The above PrP fragments can also attenuate neuronal intake of Aß. Our results strongly suggest that synthetic peptides such as PrP23-50 and PrP90-112 can be useful in designing a novel class of therapeutics in AD.


Subject(s)
Alzheimer Disease/drug therapy , Neurofibrillary Tangles/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Protein Aggregation, Pathological/drug therapy , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid/toxicity , Amyloid beta-Peptides/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/pathology , Cells, Cultured , Hippocampus/cytology , Neurites/drug effects , Neurites/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/therapeutic use , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , PrPC Proteins/chemistry , Primary Cell Culture , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar
13.
Biochim Biophys Acta ; 1853(10 Pt A): 2228-39, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26149502

ABSTRACT

Prion protein (PrP) mislocalized in the cytosol has been presumed to be the toxic entity responsible for the neurodegenerative process in transmissible spongiform encephalopathies (TSE), also called prion diseases. The mechanism underlying the neurotoxicity of cytosolic PrP (cytoPrP) remains, however, unresolved. In this study we analyze toxic effects of the cell-penetrating PrP fragment, PrP1-30--encompassing residues responsible for binding and aggregation of tubulin. We have found that intracellularly localized PrP1-30 disassembles microtubular cytoskeleton of primary neurons, which leads to the loss of neurites and, eventually, necrotic cell death. Accordingly, stabilization of microtubules by taxol reduced deleterious effects of cytosolic PrP1-30. Furthermore, we have demonstrated that decreased phosphorylation level of microtubule-associated proteins (MAPs), which also increases stability of microtubular cytoskeleton, protects neurons from the toxic effects of PrP1-30. CHIR98014 and LiCl--inhibitors of glycogen synthase kinase 3 (GSK-3), a major kinase responsible for phosphorylation of MAPs, inhibited PrP1-30-induced disruption of microtubular cytoskeleton and increased viability of peptide-treated neurons. We have also shown that the N-terminal fragment of cytoPrP may cause the loss of dendritic spines. PrP1-30-induced changes at the level of spines have also been prevented by stabilization of microtubules by taxol as well as LiCl. These observations indicate that the neurotoxicity of cytoPrP is tightly linked to the disruption of microtubular cytoskeleton. Importantly, this study implies that lithium, the commonly used mood stabilizer, may be a promising therapeutic agent in TSE, particularly in case of the disease forms associated with accumulation of cytoPrP.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neurites/metabolism , Peptide Fragments/toxicity , Prions/toxicity , Aminopyridines/pharmacology , Animals , Antimanic Agents/pharmacology , Cells, Cultured , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Lithium Chloride/pharmacology , Microtubules/pathology , Necrosis/chemically induced , Necrosis/metabolism , Necrosis/pathology , Neurites/pathology , Prion Diseases/chemically induced , Prion Diseases/drug therapy , Prion Diseases/metabolism , Prion Diseases/pathology , Pyrimidines/pharmacology , Rats , Rats, Wistar
14.
PLoS One ; 9(8): e105660, 2014.
Article in English | MEDLINE | ID: mdl-25144464

ABSTRACT

Poly-L-glutamic acid (PLGA) often serves as a model in studies on amyloid fibrils and conformational transitions in proteins, and as a precursor for synthetic biomaterials. Aggregation of PLGA chains and formation of amyloid-like fibrils was shown to continue on higher levels of superstructural self-assembly coinciding with the appearance of so-called ß2-sheet conformation manifesting in dramatic redshift of infrared amide I' band below 1600 cm(-1). This spectral hallmark has been attributed to network of bifurcated hydrogen bonds coupling C = O and N-D (N-H) groups of the main chains to glutamate side chains. However, other authors reported that, under essentially identical conditions, PLGA forms the conventional in terms of infrared characteristics ß1-sheet structure (exciton-split amide I' band with peaks at ca. 1616 and 1683 cm(-1)). Here we attempt to shed light on this discrepancy by studying the effect of increasing concentration of intentionally induced defects in PLGA on the tendency to form ß1/ß2-type aggregates using infrared spectroscopy. We have employed carbodiimide-mediated covalent modification of Glu side chains with n-butylamine (NBA), as well as electrostatics-driven inclusion of polylysine chains, as two different ways to trigger structural defects in PLGA. Our study depicts a clear correlation between concentration of defects in PLGA and increasing tendency to depart from the ß2-structure toward the one less demanding in terms of chemical uniformity of side chains: ß1-structure. The varying predisposition to form ß1- or ß2-type aggregates assessed by infrared absorption was compared with the degree of morphological order observed in electron microscopy images. Our results are discussed in the context of latent covalent defects in homopolypeptides (especially with side chains capable of hydrogen-bonding) that could obscure their actual propensities to adopt different conformations, and limit applications in the field of synthetic biomaterials.


Subject(s)
Amyloid/chemistry , Lactic Acid/chemistry , Polyglutamic Acid/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
15.
ACS Chem Neurosci ; 5(5): 340-5, 2014 05 21.
Article in English | MEDLINE | ID: mdl-24669873

ABSTRACT

Recent studies indicate that the pathogenesis of Alzheimer disease may be related to the interaction between prion protein (PrP) and certain oligomeric species of Aß peptide. However, the mechanism of this interaction remains unclear and controversial. Here we provide direct experimental evidence that, in addition to previously demonstrated binding to Aß oligomers, PrP also interacts with mature Aß fibrils. However, contrary to the recent claim that PrP causes fragmentation of Aß fibrils into oligomeric species, no evidence for such a disassembly could be detected in the present study. In contrast, our data indicate that the addition of PrP to preformed Aß fibrils results in a lateral association of individual fibrils into larger bundles. These findings have potentially important implications for understanding the mechanism by which PrP might impact Aß toxicity as well as for the emerging efforts to use PrP-derived compounds as inhibitors of Aß-induced neurodegeneration.


Subject(s)
Amyloid beta-Peptides/metabolism , Prions/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Animals , Humans , Prions/chemistry , Prions/pharmacology , Protein Binding/drug effects
16.
PLoS One ; 9(1): e86320, 2014.
Article in English | MEDLINE | ID: mdl-24466022

ABSTRACT

Formation of amyloid fibrils in vivo has been linked to disorders such as Alzheimer's disease and prion-associated transmissible spongiform encephalopathies. One of the characteristic features of amyloid fibrils is the high thermodynamic stability relative both to native and disordered states which is also thought to underlie the perplexingly remarkable heat resistance of prion infectivity. Here, we are comparing high-temperature degradation of native and fibrillar forms of human insulin. Decomposition of insulin amyloid has been studied under helium atmosphere and in the temperature range from ambient conditions to 750°C using thermogravimetry and differential scanning calorimetry coupled to mass spectrometry. While converting native insulin into amyloid does upshift onset of thermal decomposition by ca. 75°C, fibrils remain vulnerable to covalent degradation at temperatures below 300°C, as reflected by mass spectra of gases released upon heating of amyloid samples, as well as morphology and infrared spectra of fibrils subjected to incubation at 250°C. Mass spectra profiles of released gases indicate that degradation of fibrils is much more cooperative than degradation of native insulin. The data show no evidence of water of crystallization trapped within insulin fibrils. We have also compared untreated and heated amyloid samples in terms of capacity to seed daughter fibrils. Kinetic traces of seed-induced insulin fibrillation have shown that the seeding potency of amyloid samples decreases significantly already after exposure to 200°C, even though corresponding electron micrographs indicated persisting fibrillar morphology. Our results suggest that amyloid-based biological activity may not survive extremely high temperature treatments, at least in the absence of other stabilizing factors.


Subject(s)
Amyloid/chemistry , Insulin/chemistry , Protein Denaturation , Crystallization , Hot Temperature , Humans , Kinetics , Temperature , Water/chemistry
17.
Biochemistry ; 51(47): 9460-9, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23127165

ABSTRACT

The irreversibility and autocatalytic character of amyloidogenesis and the polymorphism of amyloid fibrils underlie the phenomenon of self-propagating strains, wherein the mother seed, rather than the seeding environment, determines the properties of daughter fibrils. Here we study the formation of amyloid fibrils from bovine insulin and the recombinant Lys(B31)-Arg(B32) human insulin analog. The two polypeptides are similar enough to cross-seed but, upon spontaneous aggregation, form amyloid fibrils with distinct spectral features in the infrared amide I' band region. When bovine insulin is cross-seeded with the analog amyloid (and vice versa), the shape, absorption maximum, and even fine fingerprint features of the amide I' band are passed from the mother to daughter fibrils with a high degree of fidelity. Although the differences in primary structure between bovine insulin and the Lys(B31)-Arg(B32) analog of human insulin lie outside of the polypeptide's critical amyloidogenic regions, they affect the secondary structure of fibrils, possibly the formation of intermolecular salt bridges, and the susceptibility to dissection and denaturation with dimethyl sulfoxide (DMSO). All these phenotypic features of mother fibrils are imprinted in daughter amyloid upon cross-seeding. Analysis of noncooperative DMSO-induced denaturation of daughter fibrils suggests that the self-propagating polymorphism underlying the emergence of new amyloid strains is encoded on the level of secondary structure. Our findings have been discussed in the context of polymorphism of fibrils, amyloid strains, and possible implications for mechanisms of amyloidogenesis.


Subject(s)
Amyloid/chemistry , Insulin/analogs & derivatives , Insulin/chemistry , Amides/chemistry , Amino Acid Substitution , Animals , Cattle , Deuterium , Dimethyl Sulfoxide/pharmacology , Humans , Microscopy, Electron, Transmission , Protein Denaturation , Recombinant Proteins/chemistry , Spectroscopy, Fourier Transform Infrared
18.
Biochem Biophys Res Commun ; 425(4): 788-93, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22885185

ABSTRACT

Our previous studies have demonstrated that prion protein (PrP) leads to disassembly of microtubular cytoskeleton through binding to tubulin and its oligomerization. Here we found that PrP-treated cells exhibited improper morphology of mitotic spindles. Formation of aberrant spindles may result not only from altered microtubule dynamics - as expected from PrP-induced tubulin oligomerization - but also from impairing the function of molecular motors. Therefore we checked whether binding of PrP to microtubules affected movement generated by Ncd - a kinesin responsible for the proper organization of division spindles. We found that PrP inhibited Ncd-driven transport of microtubules. Most probably, the inhibition of the microtubule movement resulted from PrP-induced changes in the microtubule structure since Ncd-microtubule binding was reduced already at low PrP to tubulin molar ratios. This study suggests another plausible mechanism of PrP cytotoxicity related to the interaction with tubulin, namely impeding microtubule-dependent transport.


Subject(s)
Cell Division , Kinesins/metabolism , Prions/metabolism , Spindle Apparatus/metabolism , Animals , Kinesins/chemistry , Microtubules/chemistry , Microtubules/metabolism , PC12 Cells , Prions/chemistry , Prions/pharmacology , Protein Transport , Rats , Spindle Apparatus/drug effects , Tubulin/chemistry , Tubulin/metabolism
19.
Mol Pharm ; 9(3): 458-69, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22206488

ABSTRACT

Alzheimer's disease (AD) is characterized by pathological aggregation of ß-amyloid peptides and MAP-Tau protein. ß-Amyloid (Aß) is a peptide responsible for extracellular Alzheimer's plaque formation. Intracellular MAP-Tau aggregates appear as a result of hyperphosphorylation of this cytoskeletal protein. Small, oligomeric forms of Aß are intermediate products that appear before the amyloid plaques are formed. These forms are believed to be most neurotoxic. Dendrimers are highly branched polymers, which may find an application in regulation of amyloid fibril formation. Several biophysical and biochemical methods, like circular dichroism (CD), fluorescence intensity of thioflavin T and thioflavin S, transmission electron microscopy, spectrofluorimetry (measuring quenching of intrinsic peptide fluorescence) and MTT-cytotoxicity assay, were applied to characterize interactions of cationic phosphorus-containing dendrimers of generation 3 and generation 4 (CPDG3, CPDG4) with the fragment of amyloid peptide (Aß(1-28)) and MAP-Tau protein. We have demonstrated that CPDs are able to affect ß-amyloid and MAP-Tau aggregation processes. A neuro-2a cell line (N2a) was used to test cytotoxicity of formed fibrils and intermediate products during the Aß(1-28) aggregation. It has been shown that CPDs might have a beneficial effect by reducing the system toxicity. Presented results suggest that phosphorus dendrimers may be used in the future as agents regulating the fibrilization processes in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Dendrimers/chemistry , Phosphorus/chemistry , tau Proteins/chemistry , Alzheimer Disease/metabolism , Amyloid beta-Peptides/ultrastructure , Animals , Cell Line, Tumor , Circular Dichroism , Humans , Mice , Microscopy, Electron, Transmission , Tyrosine/chemistry
20.
J Phys Chem B ; 115(37): 11010-6, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21842891

ABSTRACT

Amyloid fibrils, which are often associated with certain degenerative disorders, reveal a number of intriguing spectral properties. However, the relationship between the structure of fibrils and their optical traits remains poorly understood. Poly(L-glutamic) acid is a model polypeptide shown recently to form amyloid-like fibrils with an atypical infrared amide I' band at 1595 cm(-1), which has been attributed to the presence of bifurcated hydrogen bonds coupling C═O and N-D groups of the main chains to glutamate side chains. Here we show that this unusual amide I' band is observed only for fibrils grown from pure enantiomers of the polypeptide, whereas fibrils precipitating from equimolar mixtures of poly(L-glutamic) and poly(D-glutamic) acids have amide I' bands at 1684 and 1612 cm(-1), which are indicative of a typical intermolecular antiparallel ß-sheet. Pure enantiomers of polyglutamic acid form spirally twisted superstructures whose handedness is correlated to the amino acid chirality, while fibrils prepared from the racemate do not form scanning electron microscopy (SEM)-detectable mesoscopically ordered structures. Vibrational circular dichroism (VCD) spectra of ß-aggregates prepared from mixtures of all L- or D-polyglutamic acid in varying ratios indicate that the enhancement of VCD intensity correlates with the presence of the twisted superstructures. Our results demonstrate that both IR absorption and enhanced VCD are sensitive to subtle packing defects taking place within the compact structure of amyloid fibrils.


Subject(s)
Amyloid/chemistry , Polyglutamic Acid/chemistry , Circular Dichroism , Hydrogen Bonding , Microscopy, Electron, Scanning , Spectroscopy, Fourier Transform Infrared , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...