Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Biophys J ; 118(5): 1142-1151, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32105649

ABSTRACT

The polypeptide hormone islet amyloid polypeptide (IAPP) forms islet amyloid in type 2 diabetes, a process which contributes to pancreatic ß-cell dysfunction and death. Not all species form islet amyloid, and the ability to do so correlates with the primary sequence. Humans form islet amyloid, but baboon IAPP has not been studied. The baboon peptide differs from human IAPP at three positions containing K1I, H18R, and A25T substitutions. The K1I substitution is a rare example of a replacement in the N-terminal region of amylin. The effect of this mutation on amyloid formation has not been studied, but it reduces the net charge, and amyloid prediction programs suggest that it should increase amyloidogenicity. The A25T replacement involves a nonconservative substitution in a region of IAPP that is believed to be important for aggregation, but the effects of this replacement have not been examined. The H18R point mutant has been previously shown to reduce aggregation in vitro. Baboon amylin forms amyloid on the same timescale as human amylin in vitro and exhibits similar toxicity toward cultured ß-cells. The K1I replacement in human amylin slightly reduces toxicity, whereas the A25T substitution accelerates amyloid formation and enhances toxicity. Photochemical cross-linking reveals that the baboon amylin, like human amylin, forms low-order oligomers in the lag phase of amyloid formation. Ion-mobility mass spectrometry reveals broadly similar gas phase collisional cross sections for human and baboon amylin monomers and dimers, with some differences in the arrival time distributions. Preamyloid oligomers formed by baboon amylin, but not baboon amylin fibers, are toxic to cultured ß-cells. The toxicity of baboon oligomers and lack of significantly detectable toxicity with exogenously added amyloid fibers is consistent with the hypothesis that preamyloid oligomers are the most toxic species produced during IAPP amyloid formation.


Subject(s)
Diabetes Mellitus, Type 2 , Islet Amyloid Polypeptide , Amino Acid Sequence , Amyloid/toxicity , Animals , Humans , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/toxicity , Papio
2.
ACS Chem Biol ; 13(9): 2747-2757, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30086232

ABSTRACT

Islet amyloid formation contributes to ß-cell death and dysfunction in type-2 diabetes and to the failure of islet transplants. Amylin (islet amyloid polypeptide, IAPP), a normally soluble 37 residue polypeptide hormone produced in the pancreatic ß-cells, is responsible for amyloid formation in type-2 diabetes and is deficient in type-1 diabetes. Amylin normally plays an adaptive role in metabolism, and the development of nontoxic, non-amyloidogenic, bioactive variants of human amylin are of interest for use as adjuncts to insulin therapy. Naturally occurring non-amyloidogenic variants are of interest for xenobiotic transplantation and because they can provide clues toward understanding the amyloidogenicity of human amylin. The sequence of amylin is well-conserved among species, but sequence differences strongly correlate with in vitro amyloidogenicity and with islet amyloid formation in vivo. Bovine amylin differs from the human peptide at 10 positions and is one of the most divergent among known amylin sequences. We show that bovine amylin oligomerizes but is not toxic to cultured ß-cells and that it is considerably less amyloidogenic than the human polypeptide and is only a low-potency agonist at human amylin-responsive receptors. The bovine sequence contains several nonconservative substitutions relative to human amylin, including His to Pro, Ser to Pro, and Asn to Lys replacements. The effect of these substitutions is analyzed in the context of wild-type human amylin; the results provide insight into their role in receptor activation, the mode of assembly of human amylin, and the design of soluble amylin analogues.


Subject(s)
Amyloid/metabolism , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Amyloid/chemistry , Amyloid/ultrastructure , Animals , Cattle , Cells, Cultured , Conserved Sequence , Humans , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/chemistry
3.
J Alzheimers Dis ; 64(3): 995-1007, 2018.
Article in English | MEDLINE | ID: mdl-29966194

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) is linked to cellular stress and inflammation during Alzheimer's disease (AD). RAGE signals through Diaphanous-1 (DIAPH1); however, the expression of DIAPH1 in the healthy and AD human brain has yet to be methodically addressed. OBJECTIVE: To delineate the cell- and disease-state specific expression of DIAPH1 in the human medial temporal cortex during healthy aging and AD. METHODS: We used semi-quantitative immunohistochemistry in the human medial temporal cortex paired with widefield and confocal microscopy and automated analyses to determine colocalization and relative expression of DIAPH1 with key cell markers and molecules in the brains of subjects with AD versus age-matched controls. RESULTS: We report robust colocalization of DIAPH1 with myeloid cells and increased expression during AD, which strongly correlated to increased neutral lipids and morphology of inflamed myeloid cells. DIAPH1 moderately colocalized with markers of endothelial cells, astrocytes, neurons, and oligodendrocytes. DISCUSSION: Our findings localize DIAPH1 particularly to myeloid cells in the CNS, especially in AD in the locations of lipid droplet accumulation, thereby implicating RAGE-DIAPH1 signaling in dysregulated lipid metabolism and morphological changes of inflamed myeloid cells in this disorder.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/pathology , Alzheimer Disease/pathology , Myeloid Cells/metabolism , Temporal Lobe/metabolism , Up-Regulation/physiology , Adaptor Proteins, Signal Transducing/genetics , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Apolipoproteins E/genetics , Case-Control Studies , Claudin-1/metabolism , Female , Formins , Glial Fibrillary Acidic Protein/metabolism , Humans , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Receptor for Advanced Glycation End Products/metabolism
4.
Biochemistry ; 57(21): 3065-3074, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29697253

ABSTRACT

Human islet amyloid polypeptide (hIAPP) is a hormone secreted from ß-cells in the Islets of Langerhans in response to the same stimuli that lead to insulin secretion. hIAPP plays an adaptive role in glucose homeostasis but misfolds to form insoluble, fibrillar aggregates in type II diabetes that are associated with the disease. Along the misfolding pathway, hIAPP forms species that are toxic to ß-cells, resulting in reduced ß-cell mass. hIAPP contains a strictly conserved disulfide bond between residues 2 and 7, which forms a small loop at the N-terminus of the molecule. The loop is located outside of the cross ß-core in all models of the hIAPP amyloid fibrils. Mutations in this region are rare, and the disulfide loop plays a role in receptor binding; however, the contribution of this region to the aggregation of hIAPP is not well understood. We define the role of the disulfide by analyzing a collection of analogues that remove the disulfide, by mutation of Cys to Ser, by reduction and modification of the Cys residues, or by deletion of the first seven residues. The cytotoxic properties of hIAPP are retained in the Cys to Ser disulfide-free mutant. Removal of the disulfide bond accelerates amyloid formation in all constructs, both in solution and in the presence of model membranes. Removal of the disulfide weakens the ability of hIAPP to induce leakage of vesicles consisting of POPS and POPC. Smaller effects are observed with vesicles that contain 40 mol % cholesterol, although N-terminal truncation still reduces the extent of leakage.


Subject(s)
Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Amyloid/chemistry , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Disulfides/chemistry , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism
5.
Protein Sci ; 27(7): 1166-1180, 2018 07.
Article in English | MEDLINE | ID: mdl-29664151

ABSTRACT

Proteotoxicity plays a key role in many devastating human disorders, including Alzheimer's, Huntington's and Parkinson's diseases; type 2 diabetes; systemic amyloidosis; and cardiac dysfunction, to name a few. The cellular mechanisms of proteotoxicity in these disorders have been the focus of considerable research, but their role in prevalent and morbid disorders, such as diabetes, is less appreciated. There is a large body of literature on the impact of glucotoxicity and lipotoxicity on insulin-producing pancreatic ß-cells, and there is increasing recognition that proteotoxicty plays a key role. Pancreatic islet amyloidosis by the hormone IAPP, the production of advanced glycation endproducts (AGE), and insulin misprocessing into cytotoxic aggregates are all sources of ß-cell proteotoxicity in diabetes. AGE, produced by the reaction of reducing sugars with proteins and lipids are ligands for the receptor for AGE (RAGE), as are the toxic pre-fibrillar aggregates of IAPP produced during amyloid formation. The mechanisms of amyloid formation by IAPP in vivo or in vitro are not well understood, and the cellular mechanisms of IAPP-induced ß-cell death are not fully defined. Here, we review recent findings that illuminate the factors and mechanisms involved in ß-cell proteotoxicity in diabetes. Together, these new insights have far-reaching implications for the establishment of unifying mechanisms by which pathological amyloidoses imbue their injurious effects in vivo.


Subject(s)
Diabetes Mellitus, Type 2/etiology , Insulin-Secreting Cells/cytology , Islet Amyloid Polypeptide/toxicity , Receptor for Advanced Glycation End Products/metabolism , Animals , Cell Survival/drug effects , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism
6.
J Clin Invest ; 128(2): 682-698, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29337308

ABSTRACT

Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in ß cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces cellular stress or how IAPP-induced toxicity can be prevented or treated. We recently defined the properties of toxic IAPP species. Here, we have identified a receptor-mediated mechanism of islet amyloidosis-induced proteotoxicity. In human diabetic pancreas and in cellular and mouse models of islet amyloidosis, increased expression of the receptor for advanced glycation endproducts (RAGE) correlated with human IAPP-induced (h-IAPP-induced) ß cell and islet inflammation, toxicity, and apoptosis. RAGE selectively bound toxic intermediates, but not nontoxic forms of h-IAPP, including amyloid fibrils. The isolated extracellular ligand-binding domains of soluble RAGE (sRAGE) blocked both h-IAPP toxicity and amyloid formation. Inhibition of the interaction between h-IAPP and RAGE by sRAGE, RAGE-blocking antibodies, or genetic RAGE deletion protected pancreatic islets, ß cells, and smooth muscle cells from h-IAPP-induced inflammation and metabolic dysfunction. sRAGE-treated h-IAPP Tg mice were protected from amyloid deposition, loss of ß cell area, ß cell inflammation, stress, apoptosis, and glucose intolerance. These findings establish RAGE as a mediator of IAPP-induced toxicity and suggest that targeting the IAPP/RAGE axis is a potential strategy to mitigate this source of ß cell dysfunction in metabolic disease.


Subject(s)
Insulin-Secreting Cells/cytology , Receptor for Advanced Glycation End Products/metabolism , Amyloid/metabolism , Amyloidosis , Animals , Apoptosis , Cell Line , Diabetes Mellitus, Type 2/metabolism , Humans , Inflammation , Insulinoma/metabolism , Islet Amyloid Polypeptide/genetics , Islets of Langerhans/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism , Pancreas/metabolism , Protein Folding , Rats , Up-Regulation
7.
Isr J Chem ; 57(7-8): 750-761, 2017 Jul.
Article in English | MEDLINE | ID: mdl-29955200

ABSTRACT

Much of our knowledge of diabetes is derived from studies of rodent models. An alternative approach explores evolutionary solutions to physiological stress by studying organisms that face challenging metabolic environments. Polar bears eat an enormously lipid-rich diet without deleterious metabolic consequences. In contrast, transgenic rodents expressing the human neuropancreatic polypeptide hormone amylin develop hyperglycemia and extensive pancreatic islet amyloid when fed a high fat diet. The process of islet amyloid formation by human amylin contributes to ß-cell dysfunction and loss of ß-cell mass in type-2 diabetes. We show that ursine amylin is considerably less amyloidogenic and less toxic to ß-cells than human amylin, consistent with the hypothesis that part of the adaptation of bears to metabolic challenges might include protection from islet amyloidosis-induced ß-cell toxicity. Ursine and human amylin differ at four locations: H18R, S20G, F23L, and S29P. These are interesting from a biophysical perspective since the S20G mutation accelerates amyloid formation but the H18R slows it. An H18RS20G double mutant of human amylin behaves similarly to the H18R mutant, indicating that the substitution at position 18 dominates the S20G replacement. These data suggest one possible mechanism underpinning the protection of bears against metabolic challenges and provide insight into the design of soluble analogs of human amylin.

8.
Elife ; 52016 05 23.
Article in English | MEDLINE | ID: mdl-27213520

ABSTRACT

Islet amyloidosis by IAPP contributes to pancreatic ß-cell death in diabetes, but the nature of toxic IAPP species remains elusive. Using concurrent time-resolved biophysical and biological measurements, we define the toxic species produced during IAPP amyloid formation and link their properties to induction of rat INS-1 ß-cell and murine islet toxicity. These globally flexible, low order oligomers upregulate pro-inflammatory markers and induce reactive oxygen species. They do not bind 1-anilnonaphthalene-8-sulphonic acid and lack extensive ß-sheet structure. Aromatic interactions modulate, but are not required for toxicity. Not all IAPP oligomers are toxic; toxicity depends on their partially structured conformational states. Some anti-amyloid agents paradoxically prolong cytotoxicity by prolonging the lifetime of the toxic species. The data highlight the distinguishing properties of toxic IAPP oligomers and the common features that they share with toxic species reported for other amyloidogenic polypeptides, providing information for rational drug design to treat IAPP induced ß-cell death.


Subject(s)
Amyloidogenic Proteins/metabolism , Amyloidogenic Proteins/toxicity , Amyloidosis/physiopathology , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/toxicity , Amyloidosis/therapy , Animals , Cell Survival , Cells, Cultured , Inflammation/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Protein Conformation , Protein Denaturation , Protein Multimerization , Rats , Reactive Oxygen Species/analysis , Time Factors
9.
Methods Mol Biol ; 1345: 55-66, 2016.
Article in English | MEDLINE | ID: mdl-26453205

ABSTRACT

Amyloid formation and aberrant protein aggregation are hallmarks of more than 30 different human diseases. The proteins that form amyloid can be divided into two structural classes: those that form compact, well-ordered, globular structures in their unaggregated state and those that are intrinsically disordered in their unaggregated states. The latter include the Aß peptide of Alzheimer's disease, islet amyloid polypeptide (IAPP, amylin) implicated in type 2 diabetes and α-synuclein, which is linked to Parkinson's disease. Work in the last 10 years has highlighted the potential role of pre-amyloid intermediates in cytotoxicity and has focused attention on their properties. A number of intrinsically disordered proteins appear to form helical intermediates during amyloid formation. We discuss the spectroscopic methods employed to detect and characterize helical intermediates in homogenous solution and in membrane-catalyzed amyloid formation, with the emphasis on the application of circular dichroism (CD). IAPP is used as an example, but the methods are generally applicable.


Subject(s)
Amyloidogenic Proteins/isolation & purification , Intrinsically Disordered Proteins/isolation & purification , Molecular Biology/methods , Protein Aggregation, Pathological , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/isolation & purification
10.
J Diabetes Res ; 2016: 2798269, 2016.
Article in English | MEDLINE | ID: mdl-26649319

ABSTRACT

The hormone islet amyloid polypeptide (IAPP, or amylin) plays a role in glucose homeostasis but aggregates to form islet amyloid in type-2 diabetes. Islet amyloid formation contributes to ß-cell dysfunction and death in the disease and to the failure of islet transplants. Recent work suggests a role for IAPP aggregation in cardiovascular complications of type-2 diabetes and hints at a possible role in type-1 diabetes. The mechanisms of IAPP amyloid formation in vivo or in vitro are not understood and the mechanisms of IAPP induced ß-cell death are not fully defined. Activation of the inflammasome, defects in autophagy, ER stress, generation of reactive oxygen species, membrane disruption, and receptor mediated mechanisms have all been proposed to play a role. Open questions in the field include the relative importance of the various mechanisms of ß-cell death, the relevance of reductionist biophysical studies to the situation in vivo, the molecular mechanism of amyloid formation in vitro and in vivo, the factors which trigger amyloid formation in type-2 diabetes, the potential role of IAPP in type-1 diabetes, the development of clinically relevant inhibitors of islet amyloidosis toxicity, and the design of soluble, bioactive variants of IAPP for use as adjuncts to insulin therapy.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans/metabolism , Humans , Structure-Activity Relationship
11.
Diabetes ; 64(12): 4046-60, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26253613

ABSTRACT

Diabetes exacerbates cardiovascular disease, at least in part through suppression of macrophage cholesterol efflux and levels of the cholesterol transporters ATP binding cassette transporter A1 (ABCA1) and ABCG1. The receptor for advanced glycation end products (RAGE) is highly expressed in human and murine diabetic atherosclerotic plaques, particularly in macrophages. We tested the hypothesis that RAGE suppresses macrophage cholesterol efflux and probed the mechanisms by which RAGE downregulates ABCA1 and ABCG1. Macrophage cholesterol efflux to apolipoprotein A1 and HDL and reverse cholesterol transport to plasma, liver, and feces were reduced in diabetic macrophages through RAGE. In vitro, RAGE ligands suppressed ABCG1 and ABCA1 promoter luciferase activity and transcription of ABCG1 and ABCA1 through peroxisome proliferator-activated receptor-γ (PPARG)-responsive promoter elements but not through liver X receptor elements. Plasma levels of HDL were reduced in diabetic mice in a RAGE-dependent manner. Laser capture microdissected CD68(+) macrophages from atherosclerotic plaques of Ldlr(-/-) mice devoid of Ager (RAGE) displayed higher levels of Abca1, Abcg1, and Pparg mRNA transcripts versus Ager-expressing Ldlr(-/-) mice independently of glycemia or plasma levels of total cholesterol and triglycerides. Antagonism of RAGE may fill an important therapeutic gap in the treatment of diabetic macrovascular complications.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Cholesterol/metabolism , Diabetic Angiopathies/metabolism , Glycation End Products, Advanced/metabolism , Macrophages/metabolism , Receptor for Advanced Glycation End Products/agonists , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Biological Transport , Cell Line , Cells, Cultured , Diabetic Angiopathies/blood , Diabetic Angiopathies/immunology , Diabetic Angiopathies/pathology , Glycation End Products, Advanced/blood , Humans , Ligands , Lipoproteins/antagonists & inhibitors , Lipoproteins/genetics , Lipoproteins/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/pathology , Male , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Promoter Regions, Genetic , Receptor for Advanced Glycation End Products/blood , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
12.
Biochemistry ; 53(37): 5876-84, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25140605

ABSTRACT

Human islet amyloid polypeptide (hIAPP or amylin) is a polypeptide hormone produced in the pancreatic ß-cells that plays a role in glycemic control. hIAPP is deficient in type 1 and type 2 diabetes and is a promising adjunct to insulin therapy. However, hIAPP rapidly forms amyloid, and its strong tendency to aggregate limits its usefulness. The process of hIAPP amyloid formation is toxic to cultured ß-cells and islets, and islet amyloid formation in vivo has been linked to ß-cell death and islet graft failure. An analogue of hIAPP with a weakened tendency to aggregate, denoted pramlintide (PM), has been approved for clinical applications, but suffers from poor solubility, particularly at physiological pH, and its unfavorable solubility profile prevents coformulation with insulin. We describe a strategy for rationally designing analogues of hIAPP with improved properties; key proline mutations are combined with substitutions that increase the net charge of the molecule. An H18R/G24P/I26P triple mutant and an H18R/A25P/S28P/S29P quadruple mutant are significantly more soluble at neutral pH than hIAPP or PM. They are nonamyloidogenic and are not toxic to rat INS ß-cells. The approach is not limited to these examples; additional analogues can be designed using this strategy. To illustrate this point, we show that an S20R/G24P/I26P triple mutant and an H18R/I26P double mutant are nonamyloidogenic and significantly more soluble than human IAPP or PM. These analogues and second-generation derivatives are potential candidates for the coformulation of IAPP with insulin and other polypeptides.


Subject(s)
Islet Amyloid Polypeptide/chemistry , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Insulin/chemistry , Insulin-Secreting Cells , Molecular Sequence Data , Mutation , Peptides/chemical synthesis , Proline , Protein Engineering/methods , Rats
13.
Proc Natl Acad Sci U S A ; 110(48): 19279-84, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218607

ABSTRACT

Islet amyloid polypeptide (IAPP) is responsible for amyloid formation in type 2 diabetes and contributes to the failure of islet cell transplants, however the mechanisms of IAPP-induced cytotoxicity are not known. Interactions with model anionic membranes are known to catalyze IAPP amyloid formation in vitro. Human IAPP damages anionic membranes, promoting vesicle leakage, but the features that control IAPP-membrane interactions and the connection with cellular toxicity are not clear. Kinetic studies with wild-type IAPP and IAPP mutants demonstrate that membrane leakage is induced by prefibrillar IAPP species and continues over the course of amyloid formation, correlating additional membrane disruption with fibril growth. Analyses of a set of designed mutants reveal that membrane leakage does not require the formation of ß-sheet or α-helical structures. A His-18 to Arg substitution enhances leakage, whereas replacement of all of the aromatic residues via a triple leucine mutant has no effect. Biophysical measurements in conjunction with cytotoxicity studies show that nonamyloidogenic rat IAPP is as effective as human IAPP at disrupting standard anionic model membranes under conditions where rat IAPP does not induce cellular toxicity. Similar results are obtained with more complex model membranes, including ternary systems that contain cholesterol and are capable of forming lipid rafts. A designed point mutant, I26P-IAPP; a designed double mutant, G24P, I26P-IAPP; a double N-methylated variant; and pramlintide, a US Food and Drug Administration-approved IAPP variant all induce membrane leakage, but are not cytotoxic, showing that there is no one-to-one relationship between disruption of model membranes and induction of cellular toxicity.


Subject(s)
Amyloid/biosynthesis , Diabetes Mellitus, Type 2/physiopathology , Islet Amyloid Polypeptide/metabolism , Membranes, Artificial , Amino Acid Sequence , Animals , Benzothiazoles , Biophysics , Humans , Microscopy, Electron, Transmission , Molecular Sequence Data , Oxazines , Rats , Species Specificity , Thiazoles , Xanthenes
14.
FEBS Lett ; 587(8): 1106-18, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23380070

ABSTRACT

Pancreatic islet amyloid is a characteristic feature of type 2 diabetes. The major protein component of islet amyloid is the polypeptide hormone known as islet amyloid polypeptide (IAPP, or amylin). IAPP is stored with insulin in the ß-cell secretory granules and is released in response to the stimuli that lead to insulin secretion. IAPP is normally soluble and is natively unfolded in its monomeric state, but forms islet amyloid in type 2 diabetes. Islet amyloid is not the cause of type 2 diabetes, but it leads to ß-cell dysfunction and cell death, and contributes to the failure of islet cell transplantation. The mechanism of IAPP amyloid formation is not understood and the mechanisms of cytotoxicity are not fully defined.


Subject(s)
Amyloid/chemistry , Islet Amyloid Polypeptide/chemistry , Protein Structure, Secondary , Amino Acid Sequence , Amyloid/genetics , Amyloid/metabolism , Biophysical Phenomena , Cell Survival , Humans , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Sequence Homology, Amino Acid
15.
FEBS Lett ; 587(8): 1119-27, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23337872

ABSTRACT

Amyloid formation by the neuropancreatic hormone, islet amyloid polypeptide (IAPP or amylin), one of the most amyloidogenic sequences known, leads to islet amyloidosis in type 2 diabetes and to islet transplant failure. Under normal conditions, IAPP plays a role in the maintenance of energy homeostasis by regulating several metabolic parameters, such as satiety, blood glucose levels, adiposity and body weight. The mechanisms of IAPP amyloid formation, the nature of IAPP toxic species and the cellular pathways that lead to pancreatic ß-cell toxicity are not well characterized. Several mechanisms of toxicity, including receptor and non-receptor-mediated events, have been proposed. Analogs of IAPP have been approved for the treatment of diabetes and are under investigation for the treatment of obesity.


Subject(s)
Amyloidosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Amyloidosis/genetics , Humans , Islet Amyloid Polypeptide/genetics , Models, Biological , Molecular Sequence Data , Mutation , Receptors, Islet Amyloid Polypeptide/metabolism
16.
Curr Opin Struct Biol ; 23(1): 82-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23266002

ABSTRACT

Amyloid formation in the pancreas by islet amyloid polypeptide (IAPP) leads to ß-cell death and dysfunction, contributing to islet transplant failure and to type-2 diabetes. IAPP is stored in the ß-cell insulin secretory granules and cosecreted with insulin in response to ß-cell secretagogues. IAPP is believed to play a role in the control of food intake, in controlling gastric emptying and in glucose homeostasis. The polypeptide is natively unfolded in its monomeric state, but is one of the most amyloidogenic sequences known. The mechanisms of IAPP amyloid formation in vivo and in vitro are not understood; the mechanisms of IAPP induced cell death are unclear; and the nature of the toxic species is not completely defined. Recent work is shedding light on these important issues.


Subject(s)
Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Amyloid/chemistry , Amyloid/metabolism , Animals , Biophysics , Extracellular Matrix/chemistry , Humans , Insulin-Secreting Cells/drug effects , Islet Amyloid Polypeptide/toxicity , Lipopeptides/chemistry , Protein Structure, Secondary
17.
Proc Natl Acad Sci U S A ; 109(49): 19965-70, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23161913

ABSTRACT

Antibodies hold significant potential for inhibiting toxic protein aggregation associated with conformational disorders such as Alzheimer's and Huntington's diseases. However, near-stoichiometric antibody concentrations are typically required to completely inhibit protein aggregation. We posited that the molecular interactions mediating amyloid fibril formation could be harnessed to generate antibodies with potent antiaggregation. Here we report that grafting small amyloidogenic peptides (6-10 residues) into the complementarity-determining regions of a single-domain (V(H)) antibody yields potent domain antibody inhibitors of amyloid formation. Grafted AMyloid-Motif AntiBODIES (gammabodies) presenting hydrophobic peptides from Aß (Alzheimer's disease), α-Synuclein (Parkinson's disease), and islet amyloid polypeptide (type 2 diabetes) inhibit fibril assembly of each corresponding polypeptide at low substoichiometric concentrations (1:10 gammabody:monomer molar ratio). In contrast, sequence- and conformation-specific antibodies that were obtained via immunization are unable to prevent fibrillization at the same substoichiometric concentrations. Gammabodies prevent amyloid formation by converting monomers and/or fibrillar intermediates into small complexes that are unstructured and benign. We expect that our antibody design approach--which eliminates the need for immunization or screening to identify sequence-specific domain antibody inhibitors--can be readily extended to generate potent aggregation inhibitors of other amyloidogenic polypeptides linked to human disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/antagonists & inhibitors , Islet Amyloid Polypeptide/metabolism , Protein Engineering/methods , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/pharmacology , alpha-Synuclein/metabolism , Amino Acid Sequence , Benzothiazoles , Chromatography, Gel , Circular Dichroism , Cloning, Molecular , Drug Design , Electrophoresis, Polyacrylamide Gel , Fluorescence , Humans , Immunoblotting , Microscopy, Atomic Force , Molecular Sequence Data , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Thiazoles
18.
J Mol Biol ; 421(2-3): 282-95, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22206987

ABSTRACT

Islet amyloid polypeptide (IAPP, amylin) is responsible for amyloid formation in type 2 diabetes and in islet cell transplants. The only known natural mutation found in mature human IAPP is a Ser20-to-Gly missense mutation, found with small frequency in Chinese and Japanese populations. The mutation appears to be associated with increased risk of early-onset type 2 diabetes. Early measurements in the presence of organic co-solvents showed that S20G-IAPP formed amyloid more quickly than the wild type. We confirm that the mutant accelerates amyloid formation under a range of conditions including in the absence of co-solvents. Ser20 adopts a normal backbone geometry, and the side chain makes no steric clashes in models of IAPP amyloid fibers, suggesting that the increased rate of amyloid formation by the mutant does not result from the relief of steric incompatibility in the fiber state. Transmission electronic microscopy, circular dichroism, and seeding studies were used to probe the structure of the resulting fibers. The S20G-IAPP peptide is toxic to cultured rat INS-1 (transformed rat insulinoma-1) ß-cells. The sensitivity of amyloid formation to the identity of residue 20 was exploited to design a variant that is much slower to aggregate and that inhibits amyloid formation by wild-type IAPP. An S20K mutant forms amyloid with an 18-fold longer lag phase in homogeneous solution. Thioflavin T binding assays, together with experiments using a p-cyanophenylalanine (p-cyanoPhe) variant of human IAPP, show that the designed S20K mutant inhibits amyloid formation by human IAPP. The experiments illustrate how p-cyanoPhe can be exploited to monitor amyloid formation even in the presence of other amyloidogenic proteins.


Subject(s)
Islet Amyloid Polypeptide/metabolism , Mutation, Missense , Amino Acid Sequence , Cells, Cultured , Circular Dichroism , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/genetics , Microscopy, Electron, Transmission , Molecular Sequence Data , Protein Conformation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Am Chem Soc ; 132(41): 14340-2, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20873820

ABSTRACT

Amyloid formation plays a role in over 25 human disorders. A range of strategies have been applied to the problem of developing inhibitors of amyloid formation, but unfortunately, many inhibitors are effective only in molar excess and typically either lengthen the time to the onset of amyloid formation, (the lag time), while having modest effects on the total amount of amyloid fibrils produced, or decrease the amount of amyloid without significantly reducing the lag time. We demonstrate a general strategy whereby two moderate inhibitors of amyloid formation can be rationally selected via kinetic assays and combined in trans to yield a highly effective inhibitor which dramatically delays the time to the appearance of amyloid and drastically reduces the total amount of amyloid formed. A key feature is that the selection of the components of the mixture is based on their effect on the time course of amyloid formation rather than on just the amount of amyloid produced. The approach is validated using inhibitors of amyloid formation by islet amyloid polypeptide, the causative agent of amyloid formation in type 2 diabetes and the Alzheimer's disease Aß peptide.


Subject(s)
Amyloid/antagonists & inhibitors , Islet Amyloid Polypeptide/chemistry , Amino Acid Sequence , Amyloid/biosynthesis , Circular Dichroism , Kinetics , Microscopy, Electron, Transmission , Molecular Sequence Data
20.
Biochemistry ; 49(37): 8127-33, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20707388

ABSTRACT

Islet amyloid polypeptide (IAPP, amylin) is the major protein component of the islet amyloid deposits associated with type 2 diabetes. The polypeptide lacks a well-defined structure in its monomeric state but readily assembles to form amyloid. Amyloid fibrils formed from IAPP, intermediates generated in the assembly of IAPP amyloid, or both are toxic to ß-cells, suggesting that islet amyloid formation may contribute to the pathology of type 2 diabetes. There are relatively few reported inhibitors of amyloid formation by IAPP. Here we show that the tea-derived flavanol, (-)-epigallocatechin 3-gallate [(2R,3R)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-1-benzopyran-3-yl 3,4,5-trihydroxybenzoate] (EGCG), is an effective inhibitor of in vitro IAPP amyloid formation and disaggregates preformed amyloid fibrils derived from IAPP. The compound is thus one of a very small set of molecules which have been shown to disaggregate IAPP amyloid fibrils. Fluorescence-detected thioflavin-T binding assays and transmission electron microscopy confirm that the compound inhibits unseeded amyloid fibril formation as well as disaggregates IAPP amyloid. Seeding studies show that the complex formed by IAPP and EGCG does not seed amyloid formation by IAPP. In this regard, the behavior of IAPP is similar to the reported interactions of Aß and α-synuclein with EGCG. Alamar blue assays and light microscopy indicate that the compound protects cultured rat INS-1 cells against IAPP-induced toxicity. Thus, EGCG offers an interesting lead structure for further development of inhibitors of IAPP amyloid formation and compounds that disaggregate IAPP amyloid.


Subject(s)
Amyloid/antagonists & inhibitors , Amyloid/metabolism , Amyloid/chemistry , Amyloid beta-Protein Precursor , Animals , Benzothiazoles , Catechin/analogs & derivatives , Cell Culture Techniques , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Flavonoids , Islet Amyloid Polypeptide , Microscopy, Electron, Transmission , Phenols , Polyphenols , Protease Nexins , Rats , Receptors, Cell Surface , Thiazoles , alpha-Synuclein/analysis , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...