Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Molecules ; 27(3)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35164285

RESUMO

Amyloidosis is a common pathological event in which proteins self-assemble into misfolded soluble and insoluble molecular forms, oligomers and fibrils that are often toxic to cells. Notably, aggregation-prone human islet amyloid polypeptide (hIAPP), or amylin, is a pancreatic hormone linked to islet ß-cells demise in diabetics. The unifying mechanism by which amyloid proteins, including hIAPP, aggregate and kill cells is still matter of debate. The pathology of type-2 diabetes mellitus (T2DM) is characterized by extracellular and intracellular accumulation of toxic hIAPP species, soluble oligomers and insoluble fibrils in pancreatic human islets, eventually leading to loss of ß-cell mass. This review focuses on molecular, biochemical and cell-biology studies exploring molecular mechanisms of hIAPP synthesis, trafficking and degradation in the pancreas. In addition to hIAPP turnover, the dynamics and the mechanisms of IAPP-membrane interactions; hIAPP aggregation and toxicity in vitro and in situ; and the regulatory role of diabetic factors, such as lipids and cholesterol, in these processes are also discussed.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Pâncreas/patologia , Agregação Patológica de Proteínas/patologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/análise , Pâncreas/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas/metabolismo , Conformação Proteica , Dobramento de Proteína , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia
2.
Biochem J ; 478(6): 1261-1282, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33650632

RESUMO

Here, we investigated transcriptional and trafficking mechanisms of human islet amyloid polypeptide (hIAPP) in normal and stressed ß-cells. In high glucose-challenged human islets and rat insulinoma cells overexpressing hIAPP, cell fractionation studies revealed increased accumulation of hIAPP. Unexpectedly, a significant fraction (up to 22%) of hIAPP was found in the nuclear soluble and chromatin-enriched fractions of cultured human islet and rat insulinoma cells. The nucleolar accumulation of monomeric forms of hIAPP did not have any adverse effect on the proliferation of ß-cells nor did it affect nucleolar organization or function. However, intact nucleolar organization and function were essential for hIAPP expression under normal and ER-stress conditions as RNA polymerase II inhibitor, α-amanitin, reduced hIAPP protein expression evoked by high glucose and thapsigargin. Promoter activity studies revealed the essential role of transcription factor FoxA2 in hIAPP promoter activation in ER-stressed ß-cells. Transcriptome and secretory studies demonstrate that the biosynthetic and secretory capacity of islet ß-cells was preserved during ER stress. Thus, the main reason for increased intracellular hIAPP accumulation is its enhanced biosynthesis under these adverse conditions.


Assuntos
Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica , Glucose/farmacologia , Fator 3-beta Nuclear de Hepatócito/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , RNA Polimerase II/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , RNA Polimerase II/genética , Ratos , Edulcorantes
3.
Biochim Biophys Acta Biomembr ; 1860(9): 1721-1733, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29627323

RESUMO

Misfolded toxic human islet amyloid polypeptide or amylin (hA) and plasma membrane-associated redox complex, NADPH oxidase (NOX), have been implicated in the islet ß-cell demise associated with type-2 diabetes mellitus (T2DM). Studies show that hA accumulation is stressful to ß-cells and that misfolding of human amylin evokes redox stress and activates mitogen activated protein (MAP) kinases, p38 MAPK and c-Jun N-terminal (JNK) kinase. However, the molecular link and causality between hA-evoked redox stress, NOX activity and MAP kinases signaling in pancreatic ß-cells is incompletely understood. Here, we show that in the process of activating JNK, aggregation prone hA also activates an upstream apoptosis signal regulating kinase-1 (ASK1) with concomitant decrease in intracellular levels of reduced glutathione. Inhibition of ASK1 kinase activity, either by specific ASK1 inhibitor, NQDI1 or by thiol antioxidants reduces human amylin-evoked ASK1 and JNK activation and consequently human amylin toxicity in rat insulinoma Rin-m5F cells and human islets. ß-cell specific overexpression of human amylin in mouse islets elicited ASK1 phosphorylation and activation in ß-cells but not in other rodent's islet or exocrine cells. This ASK1 activation strongly correlated with islet amyloidosis and diabetes progression. Cytotoxic human amylin additionally stimulated pro-oxidative activity and expressions of plasma membrane bound NADPH oxidase (NOX) and its regulatory subunits. siRNA mediated NOX1 knockdown and selective NOX inhibitors, ML171 and apocynin, significantly reduced hA-induced mitochondrial stress in insulinoma beta-cells. However, NOX inhibitors were largely ineffective against hA-evoked redox stress and activation of cytotoxic ASK1/JNK signaling complex. Thus, our studies suggest that NOX1 and ASK1 autonomously mediate human amylin-evoked redox and mitochondrial stress in pancreatic ß-cells.

4.
Handb Exp Pharmacol ; 245: 271-312, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29043504

RESUMO

Human islet amyloid polypeptide or amylin (hA) is a 37-amino acid peptide hormone produced and co-secreted with insulin by pancreatic ß-cells. Under physiological conditions, hA regulates a broad range of biological processes including insulin release and slowing of gastric emptying, thereby maintaining glucose homeostasis. However, under the pathological conditions associated with type 2 diabetes mellitus (T2DM), hA undergoes a conformational transition from soluble random coil monomers to alpha-helical oligomers and insoluble ß-sheet amyloid fibrils or amyloid plaques. There is a positive correlation between hA oligomerization/aggregation, hA toxicity, and diabetes progression. Because the homeostatic balance between hA synthesis, release, and uptake is lost in diabetics and hA aggregation is a hallmark of T2DM, this chapter focuses on the biophysical and cell biology studies investigating molecular mechanisms of hA uptake, trafficking, and degradation in pancreatic cells and its relevance to h's toxicity. We will also discuss the regulatory role of endocytosis and proteolytic pathways in clearance of toxic hA species. Finally, we will discuss potential pharmacological approaches for specific targeting of hA trafficking pathways and toxicity in islet ß-cells as potential new avenues toward treatments of T2DM patients.


Assuntos
Amiloidose/etiologia , Diabetes Mellitus Tipo 2/etiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/metabolismo , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Endocitose , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Complexo de Endopeptidases do Proteassoma/fisiologia , Agregados Proteicos
5.
Biochem J ; 473(17): 2655-70, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27340132

RESUMO

Toxic human amylin (hA) oligomers and aggregates are implicated in the pathogenesis of type 2 diabetes mellitus (T2DM). Although recent studies demonstrated a causal connection between hA uptake and toxicity in pancreatic cells, the mechanism of amylin's clearance following its internalization and its relationship to toxicity is yet to be determined, and hence was investigated here. Using pancreatic rat insulinoma ß-cells and human islets as model systems, we show that hA, following its internalization, first accumulates in the cytosol followed by its translocation into nucleus, and to a lesser extent lysosomes, keeping the net cytosolic amylin content low. An increase in hA accumulation in the nucleus of pancreatic cells correlated with its cytotoxicity, suggesting that its excessive accumulation in the nucleus is detrimental. hA interacted with 20S core and 19S lid subunits of the ß-cell proteasomal complex, as suggested by immunoprecipitation and confocal microscopy studies, which subsequently resulted in a decrease in the proteasome's proteolytic activity in these cells. In vitro binding and activity assays confirmed an intrinsic and potent ability of amylin to interact with the 20S core complex thereby modulating its proteolytic activity. Interestingly, less toxic and aggregation incapable rat amylin (rA) showed a comparable inhibitory effect on proteasome activity and protein ubiquitination, decoupling amylin aggregation/ toxicity and amylin-induced protein stress. In agreement with these studies, inhibition of proteasomal proteolytic activity significantly increased intracellular amylin content and toxicity. Taken together, our results suggest a pivotal role of proteasomes in amylin's turnover and detoxification in pancreatic cells.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Pâncreas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/toxicidade , Microscopia Confocal , Pâncreas/citologia , Ratos
6.
Anal Chem ; 87(19): 9829-9837, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26352401

RESUMO

Native electrospray ionization (ESI) mass spectrometry (MS) is often used to monitor noncovalent complex formation between peptides and ligands. The relatively low throughput of this technique, however, is not compatible with extensive screening. Laser ablation electrospray ionization (LAESI) MS combined with ion mobility separation (IMS) can analyze complex formation and provide conformation information within a matter of seconds. Islet amyloid polypeptide (IAPP) or amylin, a 37-amino acid residue peptide, is produced in pancreatic beta-cells through proteolytic cleavage of its prohormone. Both amylin and its precursor can aggregate and produce toxic oligomers and fibrils leading to cell death in the pancreas that can eventually contribute to the development of type 2 diabetes mellitus. The inhibitory effect of the copper(II) ion on amylin aggregation has been recently discovered, but details of the interaction remain unknown. Finding other more physiologically tolerated approaches requires large scale screening of potential inhibitors. Here, we demonstrate that LAESI-IMS-MS can reveal the binding stoichiometry, copper oxidation state, and the dissociation constant of human amylin-copper(II) complex. The conformations of hIAPP in the presence of copper(II) ions were also analyzed by IMS, and preferential association between the ß-hairpin amylin monomer and the metal ion was found. The copper(II) ion exhibited strong association with the -HSSNN- residues of the amylin. In the absence of copper(II), amylin dimers were detected with collision cross sections consistent with monomers of ß-hairpin conformation. When copper(II) was present in the solution, no dimers were detected. Thus, the copper(II) ions disrupt the association pathway to the formation of ß-sheet rich amylin fibrils. Using LAESI-IMS-MS for the assessment of amylin-copper(II) interactions demonstrates the utility of this technique for the high-throughput screening of potential inhibitors of amylin oligomerization and fibril formation. More generally, this rapid technique opens the door for high-throughput screening of potential inhibitors of amyloid protein aggregation.


Assuntos
Cobre/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Agregados Proteicos , Multimerização Proteica , Sequência de Aminoácidos , Cátions Bivalentes/metabolismo , Humanos , Dados de Sequência Molecular , Oxirredução , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Espectrometria de Massas por Ionização por Electrospray
7.
Adv Exp Med Biol ; 855: 95-116, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26149927

RESUMO

Amyloidosis is a biological event in which proteins undergo structural transitions from soluble monomers and oligomers to insoluble fibrillar aggregates that are often toxic to cells. Exactly how amyloid proteins, such as the pancreatic hormone amylin, aggregate and kill cells is still unclear. Islet amyloid polypeptide, or amylin, is a recently discovered hormone that is stored and co-released with insulin from pancreatic islet ß-cells. The pathology of type 2 diabetes mellitus (T2DM) is characterized by an excessive extracellular and intracellular accumulation of toxic amylin species, soluble oligomers and insoluble fibrils, in islets, eventually leading to ß-cell loss. Obesity and elevated serum cholesterol levels are additional risk factors implicated in the development of T2DM. Because the homeostatic balance between cholesterol synthesis and uptake is lost in diabetics, and amylin aggregation is a hallmark of T2DM, this chapter focuses on the biophysical and cell biology studies exploring molecular mechanisms by which cholesterol and phospholipids modulate secondary structure, folding and aggregation of human amylin and other amyloid proteins on membranes and in cells. Amylin turnover and toxicity in pancreatic cells and the regulatory role of cholesterol in these processes are also discussed.


Assuntos
Amiloidose/fisiopatologia , Colesterol/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/fisiopatologia , Fosfolipídeos/fisiologia , Dobramento de Proteína , Sequência de Aminoácidos , Amiloidose/etiologia , Animais , Diabetes Mellitus Tipo 2/fisiopatologia , Humanos , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos
8.
PLoS One ; 8(9): e73080, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24019897

RESUMO

Toxic human amylin oligomers and aggregates are implicated in the pathogenesis of type 2 diabetes mellitus (TTDM). Although recent studies have shown that pancreatic cells can recycle amylin monomers and toxic oligomers, the exact uptake mechanism and trafficking routes of these molecular forms and their significance for amylin toxicity are yet to be determined. Using pancreatic rat insulinoma (RIN-m5F) beta (ß)-cells and human islets as model systems we show that monomers and oligomers cross the plasma membrane (PM) through both endocytotic and non-endocytotic (translocation) mechanisms, the predominance of which is dependent on amylin concentrations and incubation times. At low (≤ 100 nM) concentrations, internalization of amylin monomers in pancreatic cells is completely blocked by the selective amylin-receptor (AM-R) antagonist, AC-187, indicating an AM-R dependent mechanism. In contrast at cytotoxic (µM) concentrations monomers initially (1 hour) enter pancreatic cells by two distinct mechanisms: translocation and macropinocytosis. However, during the late stage (24 hours) monomers internalize by a clathrin-dependent but AM-R and macropinocytotic independent pathway. Like monomers a small fraction of the oligomers initially enter cells by a non-endocytotic mechanism. In contrast a majority of the oligomers at both early (1 hour) and late times (24 hours) traffic with a fluid-phase marker, dextran, to the same endocytotic compartments, the uptake of which is blocked by potent macropinocytotic inhibitors. This led to a significant increase in extra-cellular PM accumulation, in turn potentiating amylin toxicity in pancreatic cells. Our studies suggest that macropinocytosis is a major but not the only clearance mechanism for both amylin's molecular forms, thereby serving a cyto-protective role in these cells.


Assuntos
Endocitose , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Pâncreas/metabolismo , Animais , Biopolímeros/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Microscopia Confocal , Pâncreas/citologia , Ratos , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas/metabolismo
9.
Phys Chem Chem Phys ; 15(30): 12558-71, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23793354

RESUMO

Human amylin-derived oligomers and aggregates are believed to play an important role in the pathogenesis of type II diabetes mellitus (T2DM). In addition to amylin-evoked cell attrition, T2DM is often accompanied by elevated serum copper levels. Although previous studies have shown that human amylin, in the course of its aggregation, produces hydrogen peroxide (H2O2) in solution, and that this process is exacerbated in the presence of copper(ii) ions (Cu(2+)), very little is known about the mechanism of interaction between Cu(2+) and amylin in pancreatic ß-cells, including its pathological significance. Hence, in this study we investigated the mechanism by which Cu(2+) and human amylin catalyze formation of reactive oxygen species (ROS) in cells and in vitro, and examined the modulatory effect of Cu(2+) on amylin aggregation and toxicity in pancreatic rat insulinoma (RIN-m5F) ß-cells. Our results indicate that Cu(2+) interacts with human and rat amylin to form metalo-peptide complexes with low aggregative and oxidative properties. Human and non-amyloidogenic rat amylin produced minute (nM) amounts of H2O2, the accumulation of which was slightly enhanced in the presence of Cu(2+). In a marked contrast to human and rat amylin, and in the presence of the reducing agents glutathione and ascorbate, Cu(2+) produced µM concentrations of H2O2 surpassing the amylin effect by several fold. The current study shows that human and rat amylin not only produce but also quench H2O2, and that human but not rat amylin significantly decreases the amount of H2O2 in solution produced by Cu(2+) and glutathione. Similarly, human amylin was found to also decrease hydroxyl radical formation elicited by Cu(2+) and glutathione. Furthermore, Cu(2+) mitigated the toxic effect of human amylin by inhibiting activation of pro-apoptotic caspase-3 and stress-kinase signaling pathways in rat pancreatic insulinoma cells in part by stabilizing human amylin in its native conformational state. This sacrificial quenching of metal-catalyzed ROS by human amylin and copper's anti-aggregative and anti-apoptotic properties suggest a novel and protective role for the copper-amylin complex.


Assuntos
Cobre/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dicroísmo Circular , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Glutationa/química , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Radical Hidroxila/toxicidade , Íons/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos
10.
J Biol Chem ; 286(41): 36086-36097, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21865171

RESUMO

Self-assembly of the human pancreatic hormone amylin into toxic oligomers and aggregates is linked to dysfunction of islet ß-cells and pathogenesis of type 2 diabetes mellitus. Recent evidence suggests that cholesterol, an essential component of eukaryotic cells membranes, controls amylin aggregation on model membranes. However, the pathophysiological consequence of cholesterol-regulated amylin polymerization on membranes and biochemical mechanisms that protect ß-cells from amylin toxicity are poorly understood. Here, we report that plasma membrane (PM) cholesterol plays a key role in molecular recognition, sorting, and internalization of toxic amylin oligomers but not monomers in pancreatic rat insulinoma and human islet cells. Depletion of PM cholesterol or the disruption of the cytoskeleton network inhibits internalization of amylin oligomers, which in turn enhances extracellular oligomer accumulation and potentiates amylin toxicity. Confocal microscopy reveals an increased nucleation of amylin oligomers across the plasma membrane in cholesterol-depleted cells, with a 2-fold increase in cell surface coverage and a 3-fold increase in their number on the PM. Biochemical studies confirm accumulation of amylin oligomers in the medium after depletion of PM cholesterol. Replenishment of PM cholesterol from intracellular cholesterol stores or by the addition of water-soluble cholesterol restores amylin oligomer clustering at the PM and internalization, which consequently diminishes cell surface coverage and toxicity of amylin oligomers. In contrast to oligomers, amylin monomers followed clathrin-dependent endocytosis, which is not sensitive to cholesterol depletion. Our studies identify an actin-mediated and cholesterol-dependent mechanism for selective uptake and clearance of amylin oligomers, impairment of which greatly potentiates amylin toxicity.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Multimerização Proteica , Animais , Linhagem Celular Tumoral , Membrana Celular/patologia , Clatrina/metabolismo , Diabetes Mellitus Tipo 2/patologia , Endocitose , Humanos , Células Secretoras de Insulina/patologia , Ratos
11.
ScientificWorldJournal ; 10: 2054-69, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20953555

RESUMO

Essential physiological functions in eukaryotic cells, such as release of hormones and digestive enzymes, neurotransmission, and intercellular signaling, are all achieved by cell secretion. In regulated (calcium-dependent) secretion, membrane-bound secretory vesicles dock and transiently fuse with specialized, permanent, plasma membrane structures, called porosomes or fusion pores. Porosomes are supramolecular, cup-shaped lipoprotein structures at the cell plasma membrane that mediate and control the release of vesicle cargo to the outside of the cell. The sizes of porosomes range from 150 nm in diameter in acinar cells of the exocrine pancreas to 12 nm in neurons. In recent years, significant progress has been made in our understanding of the porosome and the cellular activities required for cell secretion, such as membrane fusion and swelling of secretory vesicles. The discovery of the porosome complex and the molecular mechanism of cell secretion are summarized in this article.


Assuntos
Membrana Celular/metabolismo , Exocitose , Lipoproteínas/metabolismo , Vesículas Secretórias/metabolismo , Animais , Humanos , Modelos Biológicos , Neurônios/metabolismo , Pâncreas Exócrino/metabolismo , Proteínas de Transporte Vesicular/metabolismo
12.
J Mol Biol ; 393(3): 765-75, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19720065

RESUMO

Amylin, a 37-aa pancreatic hormone, is the major constituent of islet amyloid, a hallmark of type II diabetes mellitus. Recent studies have revealed a pivotal role of anionic phospholipids in membrane-catalyzed amylin fibrillogenesis and aggregation. However, cholesterol, an integral component of eukaryotic cell membranes, also could have a role. In this study, we have examined the effect of cholesterol on amylin polymerization both on planar membranes and in solution. Using time-lapse atomic force microscopy, we have studied the dynamics and macromolecular organization of amylin on anionic and neutral planar membranes that lack or include cholesterol. On cholesterol-depleted planar membranes, amylin formed highly symmetrical tetrameric and pentameric pore-like supramolecular structures composed of 25- to 35-nm intermediate-sized globular structures or oligomers. Conversely, on membranes incorporating cholesterol, amylin formed highly compact approximately 200- to 500-nm protein clusters that constituted seeds or nuclei for continuing amylin binding and aggregation. However, cholesterol inhibited amylin nucleation with a 7-fold decrease in the number of amylin particles. Consequently, cholesterol-containing membranes accumulated significantly less amyloid with some membrane areas completely free of amyloid particles. The inhibitory effect of cholesterol on amylin aggregation in solution was also demonstrated as a 16-fold decrease in the aggregation rate. Consistent with this, circular dichroism spectroscopy revealed a stable, soluble random-coil conformation for amylin in the presence of cholesterol that could explain the inhibitory effect of cholesterol on amylin polymerization in solution and on membranes. The modulatory effect of cholesterol was largely independent of membrane charge or phospholipids, suggesting a novel cholesterol-regulated amylin polymerization process.


Assuntos
Amiloide/metabolismo , Colesterol/farmacologia , Membranas Artificiais , Peptídeos/metabolismo , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Microscopia de Força Atômica , Estrutura Quaternária de Proteína , Solubilidade/efeitos dos fármacos , Soluções
13.
Methods Cell Biol ; 90: 267-86, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19195555

RESUMO

Amylin, a 37-amino acid peptide hormone produced and secreted by pancreatic beta-cells, is the principal constituent of amyloid deposits in Type II Diabetes Mellitus (TTDM). Although much progress has been made in the understanding of amylin aggregation, molecular determinants that contribute to amylin aggregation in the pancreas and TTDM remain largely unknown. In order to better understand amylin aggregation and how membranes contribute to this process, visualization of amylin aggregation and deposition on membrane surface is of utmost importance. Here, we describe a new atomic force microscopy (AFM) approach to visualize amylin aggregation and to asses amylin-surface interactions. Using AFM in contact or tapping mode in fluid, amylin phase transitions on different supports were studied in real time and with high spatial nanometer-resolution. On mica, a two-stage sequential conversion of amylin from soluble monomer to small oligomers and further to mature amyloid fibrils was revealed by the AFM. This amylin conversion was accompanied by peptide conformational transition from random coil to beta-sheets assessed by CD spectroscopy. In contrast to mica, amylin formed amorphous amyloid deposits on planar lipid membranes consistent with pathological findings in diabetic subjects. Anionic lipid phosphatidylserine (PS) and membrane cholesterol had opposing effect on the kinetics and the extent of amylin aggregation. PS stimulated amylin aggregation, whereas cholesterol reversed the effect of PS. In addition, cholesterol sequestered amylin aggregates into membrane microdomains that in turn decreased amyloid deposition across the membranes. Hence, this reconstituted AFM approach offers new molecular insights to the etiology of diabetes that could be extended to investigate amylin aggregation in living islet cells at a subcellular resolution.


Assuntos
Amiloide/metabolismo , Membrana Celular/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Imageamento Tridimensional/métodos , Nanotecnologia/métodos , Silicatos de Alumínio , Amiloide/química , Amiloide/ultraestrutura , Animais , Benzotiazóis , Bioensaio , Membrana Celular/ultraestrutura , Colesterol/metabolismo , Dicroísmo Circular , Fluorescência , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Lipossomos/metabolismo , Microscopia de Força Atômica , Modelos Biológicos , Transição de Fase , Fosfolipídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Ratos , Soluções , Propriedades de Superfície , Tiazóis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...