Islet Amyloid: An Enigma of Type 2 Diabetes Anne Clark

Diabetes Research Laboratories, Radcliffe lnfirmary and Department of Human Anatomy, University of Oxford, U.K.

1. INTRODUCTION

Pancreatic islets have only recently reemerged as a site of a causative pathology in type 2 diabetes as a result of the identification in 1987-1988 of the peptide which is the principal component of islet amyloid.’,2 Early in the twentieth century, histopathological studies identified pancreatic lesions in d i a b e t e ~and ,~~ more than 30 years before the biochemical identification of two separate forms of diabetes, a far-sighted statement by Wright in 1927 described islet amyloid (hyalinosis) as a possible diagnostic feature.6 He suggested that ”insular changes. . . in younger individuals consist chiefly of inflammatory lesions,. . . hyaline change is characteristic of a group of cases in which the duration of the disease has been of some length,. . . and in which the patients are middle aged or older.” Since that time, islet amyloid has been shown unequivocally to be an important diagnostic feature for type 2 diabetes at postmortem, but the roles of the islet deposits and of the hormonal action of the amyloid peptide in the onset and progression of the disease are still unclear.

11. WHAT IS AMYLOID? Amyloid is the name given to amorphous masses of fibrils found in the body tissues Addressee for correspondence: Anne Clark, Diabetes Research Laboratories,Radcliffe Infirmary, Woodstock Road, Oxford OX3 6HE, U.K.

which have characteristic histological staining properties. The cotton dye Congo red binds to the fibrils, staining them pink. When viewed in polarized light, the stained fibrils exhibit a green/yellow birefringence due to the regular repeating pattern of Congo red binding sites.7 The term amyloid was coined in 1853 by Virchow, who found that amyloid deposits stained black with iodine, suggesting a “starchlike” material. This staining characteristic results both from polyiodide complexes which attach to the fibrils and from glycosylated compounds which are an integral part of amyloid deposits.8 However, the major fibrillar component is proteinaceous and each amyloid dyscrasia is characterized by a specific ~ e p t i d e Systemic .~ amyloidoses are commonly related to a generalized systemic disease. For example, in chronic inflammation, fibrils are formed from serum amyloid A protein,1° whereas in localized amyloidoses, deposits are restricted to the site of origin of the component peptide. Furthermore, amyloid which develops in medullary carcinoma of the thyroid is restricted to the thyroid and derived from calcitonin-related peptides;” Alzheimer’s amyloid in the brain is formed from A4 protein and found only in the brain;I2 islet amyloid is found only in the pancreas and is formed from islet amyloid polypeptide (IAPP), a peptide component of the islet B - ~ e l l s . ’ ~ , ’ ~ Amyloid fibrils are formed from two or more twisted filaments composed of stacked sheets of the peptide in a P-pleated conformation held together by hydrogen bonding. This structure confers a considerable degree of insolubility to the amyloid deposits.

Diabetes/Metabolism Reviews, Vol. 8, No. 2, 117-132 (1992)

0 1992 by John Wiley & Sons, Ltd.

07424221/92/020117- 16$8.00

118

CLARK

111. IS ISLET AMYLOID SPECIFIC FOR TYPE 2 DIABETES?

have untested glucose intolerance. Furthermore, islet amyloid deposits are not uniformly distributed throughout the organ and therefore multiple tissue sampling is The much quoted papers of Opie'5,'6 reported Islet amyloid is a characteristic of type 2 pathological "hyalinosis" in the pancreas of two diabetes in every ethnic group examined to date, diabetic patients. Although one subject was a including Pima Indians,25 Asian Indians,2h and middle-aged patient with a history of glycosuria, the Japanese.27 Furthermore, islet amyloid has the second subject was only 17 years old and been identified in older patients with diabetes post-morterm autolysis was so far advanced as to arising secondary to cystic f i b r o ~ i s . ' ~Extensive ,~~ obscure definition of the i ~ 1 e t s . lMore ~ rigorous amyloid deposits are found in many cases of large post-mortem studies showed the association polypeptide-producing tumours including insuof islet hyalinosis with d i a b e t e ~ . ~ ~BelP7 ,'~,~~ l i n ~ m a s . ~A ~ , peptide ~' component common to reported that the prevalence of islet amyloid was both islet and insulinoma amyloid was proposed greater than 40% in more than 1000 diabetic from early histochemical studies31 and subpatients over the age of 40 compared with 8 % in sequently identified as islet amyloid polypeptide. an age-matched non-diabetic group. However, this group of diabetic patients must have included some individuals who would today be classified as type 1 diabetic subjects, since diagnosis was IV. WHAT IS ISLET AMYLOID based only on the results of a urinary glucose POLYPEPTIDE? test. The reported prevalence of islet amyloid in post-mortem studies is very variable-between 25 and 92% in type 2 diabetic patients over the Histochemical and biochemical studies pionage of 40 years.1a22 However, in all reports, the eered by Westermark have shown that the amyloid occurrence of amyloid is less (between 0 and of islets and insulinomas differs biochemically 54%) in age-matched non-diabetic subjects'a22 from that formed in other systemic amyloidoses, (Figure 1). Accurate diagnosis of patients and being insoluble in distilled water and most organic solvents and lacking tryptophan.32 In sampling of pancreatic tissue are of critical 1986-1987 a novel peptide was extracted from importance, since many elderly patients dying in pancreatic tissue of type 2 diabetic patients and hospital of cardiovascular-related disease may

0Non-diabetic

Type 2 diabetic

80 U .0

-

2

60

5 '5

40

a

*

20

0

Caucasian

Japanese

Asian

Pima

Figure 1. Prevalence of islet amyloid in post-mortem pancreatic specimens from diabetic (B) and non-diabetic (Cl) subjects in different ethnic groups. The numbers of patients examined are indicated above the histograms. Data derived from published studies, refs 1&23 (Caucasian), 27 (Japanese), 26 (Asian), and 25 (Pima Indian).

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES

from an insulinoma and shown to be composed This , ~ ~discovery was made of 37 amino a ~ i d s . ' ~ by two separate groups in Sweden and the U.K. with the consequent result of multiple, and sometimes confusing, terminology for the same peptide: diabetes associated peptide (DAP), insulinoma amyloid polypeptide, islet amyloid polypeptide (IAPP), and "amylin". Subsequently, the terms IAPP and "amylin" have been retained and IAPP will be used throughout this review. Human IAPP (hIAPP) has a 46% structural homology with human calcitonin gene-related peptide (hCGRP-2), a neuropeptide produced largely by the C-cells of the t h y r ~ i d , ' ~the ,~~ homologous sequences being largely situated at the N- and C-terminal ends of the peptide. Although there are some similarities with insulin, particularly the N-terminal Cys bridge,35 IAPP has a closer association with the CGRP family of peptides than with insulin. The solubility of synthetic IAPP is low due to the primary and secondary structure of the peptide. The central region of hIAPP, particularly residues 20-24 (SNNFG) (Figure Z), is more hydrophobic than the comparable region ( G G W K ) of the more soluble CGRP.36 Secondary

119

structure predictions from circular dichroism spectroscopy data indicate that the hCGRP-1 molecule contains a greater OL helical component than IAPP but similar amounts of the p protein c o n f ~ r m a t i o n The . ~ ~ amount of p structure is an important factor in the amyloidogenic potential of a peptide: secondary structure predictions indicate that over 50% of the molecular structure of the amyloidogenic proteins tra n ~ th y r e tin ~' and P 2 - m i ~ r o g l o b u l i nis~ ~in the form of a P-pleated sheet. Aggregation of IAPP in vivo is critically dependent on the amino acid sequence of IAPP2(r2q, which varies between specie^.^^,^^ IAPP has been identified in every species examined to date: in the monkey, cat,44hamster,4scougar,"rat, mouse, g ~ i n e a - p i g , and ~ ~ -degu5I ~ ~ (Figure 2). The sequence of IAPP,-,, is highly conserved with a minimum of 78% homology present in the guinea pig and degu (Figure 2). IAPP also aggregates to form fibrils in vitro, a feature shared by other amyloidogenic proteins including residues 18-28 of the Alzheimer A4 p r ~ t e i n . ~The ' sequence Ala-Ile-Leu-Ser (hIAPP,,,,) appears to be critical , ~ ~ sequence is for fibrillogenesis in v i f r ~ . * ~This also important for amyloid formation in vivo since

NH2- terminal Signal peptide

propep t ide

CKIH-terminal I s l e t amyloid polypeptide

propeptide 37

p-pleated sheet forming region Figure 2. Amino acid sequence of the islet amyloid precursor protein, prepro-IAPP, in nine species of mammal. Whereas prepro-IAPP contains 89 amino acids in man, monkey, cat and dog, other species have 91-93 amino acids. The amino acid sequence which is critical for fibrillogenesis, hIAPP24_28(GAILS), is conserved in species in which amyloid develops in islets or insulinomas (human, cat, and dog). Data derived from refs 42-51. Amino acids are indicated by a single letter code: A, alanine; C, cysteine; D, aspartic acid; E, glutamic acid; F, phenylalanine; G, glycine; H, histidine; I, isoleucine; K, lysine; L, leucine; M, methionine; N, asparagine; P, proline; Q, glutamine; R, arginine; S, serine; T, threonine; V, valine; Y, tyrosine.

120

CLARK

in embryologial d e ~ e l o p m e n tIn . ~ addition, ~ islets it is common to man, domestic cat, and the cougar containing amyloid may be restricted to one or (Figure 2), which are all species in which diabetes two exocrine lobules. A lobular distribution of develops later in life. Diabetes-related amyloid pathological change is also a characteristic of the also occurs in older non-human primate^.^^,^^ early phases of autoimmune destruction of B-cells IAPP in macaque monkeys (Macaca nemestrina) in type 1 diabetes.59 Why this should occur is differs from hIAPP and feline IAPP in having unclear, but islets originating from different threonine at position 25,42 but since islet amyloid is present in other closely related macaque pancreatic duct stem cells could have different species (Macaca mufatta and Macaca n i g ~ a ) ~this ~ , ~ ~ ,susceptibilities to pathological processes. substitution is unlikely to affect the potential for amyloid formation. Residue 25 is proline in mice, B. Does Amyloid Originate Inside or rats, and hamsters, and Westermark et aL40 have Outside the B-Cells? suggested that this particular residue is a critical determinant for fibrillogenesis. However, analysis Electron microscopy has demonstrated that the margins of the B-cells adjacent to the amyloid of canine IAPP has shown that the amino acid show invaginations filled with fibrils.22,h"The sequence cannot be the only determining factor for islet amyloid formation: whereas the feline marginal B-cell plasma membrane is not clearly and canine IAPP2P29 sequence is identical, islet defined in the region of these invaginations, and amyloid is associated with diabetes in the cat cellular debris, including apparently isolated pockets of cytoplasm and insulin granules, are but not in the dog, even though amyloid formed from IAPP is present in some canine present within the amyloid masses (Figure 3b)." i n s ~ l i n o m a s . This ~ ~ , ~suggests ~ that in addition These observations suggest that amyloid could be formed at an intracellular site; accumulation to the IAPP sequence, physiological and/or biochemical factors, possibly determined by of insoluble IAPP within the cell could disrupt genetic or temporal features, are involved in the normal metabolism and lead to B-cell death amyloid formation. resulting in apparently extracellular masses of amyloid. Further support for this hypothesis comes from the finding of immunoreactive amyloid fibrils, apparently within the B-cell, in islets V. WHERE ARE AMYLOID AND IAPP of diabetic cats62and in human insulinoma tissue FOUND? (Figure 4).63In addition, many B-cells in amyloidcontaining islets show degenerative changes. A. Amyloid Deposits in the Pancreas IAPP is a normal constituent of B-cells in nondiabetic and diabetic individuals, where it is Islet amyloid deposits are situated within the boundaries of the islet between the endocrine located in insulin granules, and lysosomes.64~65 cells and islet capillaries and show immunoreactivThe highest density of labelling in human and ity for IAPP (Figure 3a). Islet amyloid in the South monkey B-cells was found in secondary lysosomes and in lipofuscin bodies, which are storage American rodent, the degu, is formed from insulin rather than from IAPP.s6 Islet amyloid, in common organelles for insoluble, partially degraded material. If polymerization of IAPP into fibrils with other forms of amyloid, contains other substances: notably, heparan sulphate and amyoccurred as a result of accumulations of the loid P protein. The role of these amyloidophilic peptide, lipofuscin bodies would represent a potential intracellular site for the initiation of compounds in the process of deposition of the amyloidosis. Abnormal cleavage of IAPP,_,, from fibrils is not ~ l e a r . Calcification, ~~,~~ in the form the N-terminal flanking peptide has been proof calcium phosphate crystals, is often associated posed in amyloidosis" but there is no evidence with extensive amyloid deposits, particularly those in monkey and cats. for involvement of the C-terminal flanking peptide The distribution of amyloid is far from in amy10id.~~ uniform throughout all of the islets of the pancreas, which makes accurate identification of C. IAPP in Non-8-Cells islet amyloidosis a tedious procedure. Amyloid has not been found in islets in the head of the Although IAPP is localized to B-cells in the human pancreas which contain few B-cells and pancreas of adult cats, monkeys, and man, which are derived from the ventral primordium expression during development (and possibly

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES

121 ^---

-_I

- -

_ I.L

~ " -

Figure 3. Amyloid deposits in islets of type 2 diabetic patients showing immunoreactivity for IAPP. (a) Immunoperoxidase-labelled amyloid deposits (arrows) within the boundaries of the islet. Amyloid is located as large deposits in the centre of the islet or adjacent to islet capillaries (arrow-heads). Scale bar = 100 pm. (b) Immunogold-labelled islet amyloid (AM) adjacent to an insulin-containing B-cell (B) and a glucagon-containing Acell (A). Cellular debris (C) including nuclei (N) and granules (I) appear to be engulfed by the amyloid. Scale bar = 1.0 pm.(c) Immunogold-labelled (anti-IAPP) islet amyloid fibrils in extracellular islet deposits. Scale bar = 0.1 pm.

during ontogeny) may not be so specific. During human foetal development, IAPP appears in Bcells at 10 weeks' gestation together with insulin.68 However, IAPP and insulin expression may be less tightly linked in foetal endocrine cells than in adult tissue: immunoreactivity has been identified in pluripotent foetal islet cells6' and IAPP can be co-expressed with glucagon (A-cells) or somatostatin (D-cells) in cell lines transformed from neonatal rat islet cells.70 Furthermore, IAPP has been identified in the stomach and duodenum of man and the rat; mRNA IAPP in extracts of rat stomach were approximately 5% of that found in the pancreas of and cells immunoreactive for IAPP but not for insulin have been localized to the mucosa of the pyloric antrum in rat and man.71

VI. IS ISLET AMYLOID A PRIMARY OR A SECONDARY FEATURE OF DIABETES? Diabetes-associated amyloid is found only in humans, non-human primates, and cats.7' Islet amyloid is not found in rodent models of obesity and diabetes, even though the physiological parameters may be similar to those of the syndrome of type 2 diabetes in man. Amyloid deposits can be detected before the onset of hyperglycaemia in the course of development of spontaneous diabetes in ~ a t sand ~ ~monkey^.^' , ~ ~ Whereas the amyloid deposits in the "prediabetic" period are relatively small and restricted to a few islets, the degree of amyloid formation and the number of islets affected increase with

122

CLARK

Figure 4. Fibrils immunoreactive for IAPP within B-cells of human insulinomas. (a) Radiating circular pattern of fibrils (arrows) within the cytoplasm adjacent to insulin granules (I), but within the cell membrane (CM). Scale bar = 1.0 pm. (b) Parallel arrays of immunogold-labelled fibrils not enclosed by detectable membranes (arrows). I = Insulin granule; M = Mitochondrion. Scale bar = 1.0 pm.

the progression of symptoms which lead to d i a b e t e ~ . The ~ ~ ,onset ~ ~ of hyperglycaemia could be closely related to the degree of B-cell loss which correlates with the progressive increase in amyloid deposition (Figure 5a). In man, it is not possible to study such a progression of islet pathology. The extent of islet amyloid formation at post-mortem in type 2 diabetic subjects who had received diet or oral hypoglycaemic agent therapy is very variable: these patients have less than 5% or more than 60% of islets affected with minimal reduction in islet cell population.zz Although the amyloid is located between the islet cells and capillaries in affected islets, it seems unlikely that this could seriously disrupt total pancreatic insulin production. However, if amyloid formation is initiated intracellularly, this could have a considerable effect on the normal function of the B-cell. In severe islet amyloidosis in type 2 diabetic

man, amyloid deposits can be found in up to 90% of the pancreatic islets, occupying up to 80% of the islet spacezz and in diabetic cats and monkeys, 60-100% of islets are affected.7677 In this severe pathological state, the few remaining islet cells are completely enclosed within the mass of amyloid (Figure 5b). The time course and extent of glucose stimuli and the resulting insulin release in such conditions are likely to be severely compromised by the physical barrier between the islet cells and capillaries. In spontaneously diabetic monkeys, fasting insulin levels are closely correlated with the proportion of the islet occupied by amy10id.~~ Furthermore, in type 2 diabetic man, the extent of islet amyloidosis has been shown to relate to the severity of the diabetic symptoms as defined by the need for insulin therapy.” Thus, is is likely that, once initiated, polymerization of IAPP to form fibrils continues over periods of years, causing progressive deposition of islet amyloid. This clinical undetectable process dis-

-

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES Islet

123 Islet Amyloid %

B-cell %

100

80

;

U

'

c

-ar

60

K!

Lc

0

0

c

40

5

?

c?

20

0

I1 normal

*

*

obese

* * * * * * diabetic *** **

hyperinsulinaemic

Figure 5. Islet amyloid and 8-cells in islets of spontaneously diabetic monkeys. (a) Morphometric data of the proportion of the islet occupied by amyloid ( 1 and B-cells (W in post-mortem tissue of 26 Macaca mulutta monkeys examined at different stages in progression to hyperglycaemia. Amyloid deposits (*) were found in some islets of 3/9 obese animals older than 12 years, 4/6 obese hyperinsulinaemic animals, and in all islets of 8/8 diabetic monkeys. B-cells occupied 4.540% of the islet area in non-diabetic animals but this area was reduced to less than 10% in most diabetic animals. (b) Pancreatic islet from a diabetic monkey. B-cells, immunoperoxidase labelled for insulin, (arrows) are severely reduced in number and enclosed in amyloid (arrow-heads). Scale bar = 100 pm. (c) Adjacent histological section immunoperoxidase-labelled for IAPP. Large amyloid deposits (arrow-heads) which occupy most of the islet area could severely compromise insulin secretion. (Studies in collaboration with Barbara Hansen, Baltimore USA.)

124

CLARK

rupts the islets and destroys the B-cells, resulting in diminishing islet function.

VII. IS THERE A GENETIC DEFECT IN ISLET AMYLOID POLYPEPTIDE IN DIABETES? Type 2 diabetes is a familial disease with a high concordance rate in identical twins.78 Although the glucokinase gene has been implicated in mild beta-cell d y ~ f u n c t i o nthe , ~ ~genetic basis of more severe type 2 diabetes has not been identified. The discovery of IAPP and the high prevalence of islet amyloid in type 2 diabetes offered an attractive candidate for a genetic defect. Following the identification of the amino acid sequence of the amyloid peptide, the cDNA encoding IAPP was determined from a h phage library of human genomic DNA and from an insulinoma cDNA library.80,8' The 37-amino acid peptide is derived from a larger precursor of 89 amino acids in man (molecular weight 9808 D).83 The hIAPP gene consists of three exons: exon 1 (104 bp) encodes most of the 5' untranslated part of the mRNA; exon 2 (95 bp) encodes 15 untranslated nucleotides, the signal sequence (22 residues), and the first nine amino acids of the N-terminal flanking peptide (N-IAPP); and exon 3 (1059 bp) encodes the dibasic processing site between N-IAPP and IAPPI-37, the C-terminal peptide, and the remainder of the 3' untranslated region (Figure 2).80-83 At least two sites for polyadenylation of the hIAPP and mRNA have been identified in extracts of human pancreas and i n s u l i n ~ m a . ~ ~ The IAPP gene has been located as a single copy gene on the short arm of chromosome 12.81,83 This contrasts with the related CALC-1,CALC-11, and CALC-I11 genes encoding CGRP and calcitonin which are assigned to chromosome 11. However, chromosomes 11 and 12 are thought to be related in evolution, and a possible common ancestral gene for IAPP, CGRP, and calcitonin has been proposed.84 Compared with IAPP, the CALC genes show a greater degree of complexity with five or six exons and alternative RNA processing in the CALC-I gene. Other forms of amyloid disease are associated with genetic abnormalities which result in single or multiple amino acid substitutions in the protein that forms the a m y l ~ i d . ~The ~ . ~amino ~ acid sequence for hIAPP deduced from cDNA was

identical to the peptide structure identified in extracts from a type 2 diabetic pancreas and from an i n ~ u l i n o m a , ' ~suggesting ,~~ that there is no causative structural change. Although abnormalities of the IAPP gene would be a convenient explanation for type 2 diabetes, extensive analyses have shown no variations in cDNA in diabetes: IAPP cDNA was examined in 25 well-defined type 2 diabetic individuals and found to be identical in each case, with the same encoding sequence on both alleles which matched that identified in the genomic library.87 Sequence polymorphisms do exist in the hIAPP cDNAX8 and these have been informative for linkage studies. It has been suggested that sequence polymorphisms are present in the 3' untranslated region of the gene.82 A population study of 88 type 2 diabetic and 67 non-diabetic age-matched subjects showed no linkage between diabetes and IAPP cDNA restriction fragment length p o l y r n o r p h i ~ m sAlthough .~~ subtle changes in the untranslated region cannot be excluded, it now seems unlikely that an abnormal IAPP gene plays a major role in the aetiology of type 2 diabetes. Whereas the nucleotide sequences in the IAPP,_,, domain show a high degree of conservation, there are considerable differences in the regions of the N- and C-flanking peptides."&" In man and cat, the N-terminal propeptide consists of 11 residues, but in the rat, mouse, and guineapig, three additional residues are encoded by nucleotides at the junction of exon 2 and the adjacent intron. Sizes of mRNA transcripts which have been reported indicate variation among species, suggesting that there may be multiple sites for initiation of transcription and/or termination or, possibly, alternative RNA splicing.8283 Studies on the transcriptional control of the human IAPP gene have identified a mechanism of negative control.93 Transcription of IAPP and insulin may be regulated by similar mechanisms. However, to date, quite extensive analyses of the IAPP gene have not provided any information which can explain islet amyloid formation.

VIII. IS ISLET AMYLOID FORMED AS A RESULT OF OVERPRODUCTION OF IAPP? Expression of mRNA and secretion of IAPP and insulin are closely linked in physiological responses to B-cell secretagogues and in animal

ISLET AMYLOID: A N ENIGMA OF TYPE 2 DIABETES

models of B-cell dysfunction. In the rat pancreas, the amount of pancreatic IAPP mRNA is approximately 5-10% that of mRNA for insulin and is reduced in parallel with insulin in streptozotocininduced diabetes, h y p ~ g l y c a e m i a , ~and ~ , ~ in ~,~~ spontaneously diabetic (BB) Wistar rats.95 IAPP is co-secreted with insulin in response to B-cell secretagogues. Studies on B-cells, islets and pancreas in vitro have indicated that, under most conditions, changes in IAPP secretion parallel changes in insulin secretion and represent 1-20% of the insulin production on a molar In non-diabetic man and animals, in U I Z I O plasma IAPP concentrations are low in comparison with insulin: basal levels in man vary from 2 to 13 pmol/l and are elevated in parallel with insulin secretion to 5-17 pmol/l after a glucose load (Figure 6).'00-106 Obese individuals in whom basal and stimulated insulin levels are elevated have proportionally higher levels of IAPP compared with normal-weight subject^.^^^-"^ However, there is no evidence for elevated or reduced basal or stimulated IAPP concentrations in type 2 diabetic subjects or in subjects with impaired glucose tolerance who may be at risk of developing diabetes. The highest reported plasma concentrations of IAPP (25 pmol/l) occurred in patients with chronic renal insufficiency. Following dialysis, the IAPP levels in these patients were reduced by SO%, suggesting that, like C-peptide, IAPP is normally excreted via the kidney.l" There are considerable differences in the reported absolute levels (Figure 6) and the molar ratio of secreted IAPP/insulin, and this area continues to be controversial since elevated IAPP secretion in the prediabetic period or in diabetes could contribute to amyloid formation. The measurement of IAPP in plasma samples by radioimmunoassay is difficult. The iodinated IAPP tracer is relatively unstable and the low plasma concentrations of IAPP require that plasma samples be concentrated and extracted before assay.'12 Furthermore, more than one molecular form of IAPP has been detected in the circulation: both intact IAPPl-,7 and a shorter form, IAPP17-37,have been identified, in variable ratios, in rat and human p1a~rna.l'~ If amyloid formation was caused by overexpression of IAPP, elevated IAPP concentrations would be expected not only in the islets but also in the general circulation, where the IAPP concentration would be dependent on both secretion and clearance. The lack of elevated circulating IAPP levels in type 2 diabetic subjects

125

with or without insulin resistance suggests that plasma levels of IAPP, as measured by the present methods, are not robust markers for amyloid formation. In non-diabetic patients with endstage renal failure, which is associated with elevated plasma concentrations of IAPP,"' there is a higher prevalence of islet amyloid compared with age-matched non-diabetic subjects without renal disease.l13 This could result from the higher than normal plasma IAPP concentration in the islet circulation, undiagnosed prediabetes, or the insulin resistance that often accompanies renal insufficiency. Secretion of incompletely processed B-cell products in the form of proinsulin have been implicated in the pathophysiology of type 2 diabete~,"~ and has been associated with islet amyloidosis in one patient.115

IX. WHAT IS THE FUNCTION OF IAPP? A. Peripheral Effects The C-terminal amidation of IAPP suggests that it has endocrine activity, and effects of IAPP have been studied in a variety of tissues, most notably, skeletal muscle."~"2" High concentrations of IAPP have been shown to reduce insulin-stimulated uptake of glucose and incorporation of glucose into glycogen, and to stimulate production of phosphorylase-a in isolated human and rat skeletal muscle strips.l2' It was therefore proposed that IAPP acts to modulate insulin resistance and, as such, would be a significant factor in the decreased insulin sensitivity of obesity and type 2 diabetes. However, confirmation of the in vitro findings in intact animals and in man has not been universal. Concentrations of IAPP required to inhibit insulin action on skeletal muscle in vitro range between 10 and 100 nM. Infusions of high concentrations of IAPP significantly reduced insulin-dependent disposal of a glucose load and suppressed hepatic glucose output in some experiments and showed no effects in Ho wever, lower plasma concentrations within the range of measured IAPP levels in man (5-50 pM) showed no effects on glucose handling in intact rats or dogs.'22,123,12x It has been suggested that effects of IAPP on glucose metabolism are critically dependent on the molecular conformation (intact cysteine bridge and C-terminal amidation) of the synthetic peptide and that these factors are not necessarily reflected in the measurements of plasma IAPP. However,

126

CLARK

st i mu1at ed

basal

la n 15 0

+

+

A

b

12

0 0

9

+

0

6

+ o 3

+

+

+

A

4

. +

a 0

0

0

N H B M L S J Studies

N H B M L

Figure 6. Measurements of plasma IAPP concentrations (pmol/l) in man under basal conditions and peak values following an oral glucose tolerance test (stimulated). Data are derived from seven published studies: N = ref. 103; H = ref. 109; B = ref. 105; M = ref. 104; L = ref. 108; S = ref.107; J = ref.106. + = Non-diabetic patients; 0 = type 1 diabetic subjects; 0 = type 2 diabetic subjects, diet or oral therapy; = type 2 diabetic subjects, insulin therapy; A = obese, normal glucose tolerance test; A = obese glucose-intolerant non-diabetic patients. There is an increase in circulating IAPP levels following glucose stimulation, but no clear indication of elevated basal concentrations in any of the patient groups examined.

even with intact IAPP.NH2, concentrations of 10 nM are required to induce effects in vitro,"* whereas the highest concentration recorded in vivo is 25 pmol/l.lll CGRP and IAPP have similar biological effects and both peptides act via CGRPbinding sites in skeletal muscle and hepatic cells'29 possibly involving activation of adenylate c y ~ 1 a s e . The l ~ ~ hypothesis of a specific peripheral action of IAPP on carbohydrate metabolism in diabetes therefore remains attractive, but unproven. An action of IAPP on calcium metabolism has been proposed because infusions of IAPP decrease plasma calcium concentration^.^^^

IAPP also affects bone resorption, although, compared with calcitonin, IAPP is 10 to 100 times less potent in this r e ~ p e c t . ' ~ l , l ~ ~ B. Effects within the islet CGRP affects insulin and glucagon secretion under various physiological conditions in rats and mice,133,134 pigs,13' and dogs.136It is therefore possible that the structurally homologous IAPP molecule could act as a paracrine regulator within the islets. Although high concentrations of IAPP (10-500 pM) inhibit insulin secretion, IAPP concen-

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES

127

trations less than 5 p M failed to reduce stimulated insulin production from either isolated rat islets137 or the more sensitive perfused pancreas preparation or in intact Furthermore, 10 pM amidated IAPP had no effect on proinsulin biosynthesi~.'~~ A paracrine/autocrine regulatory role for IAPP within the islet is equally attractive but remains unproven. IAPP partially shares the potent vasodilator properties of CGRP,143,144and CGRP immunoreactive nerves are present in pancreatic i ~ 1 e t s . A I ~possibility ~ therefore exists that IAPP, co-secreted with insulin, and CGRP from nerves could facilitate insulin delivery to the circulation by dilatation of vessels in the islets or pancreas.

remains a marker for an unidentified dysfunction of 8-cells associated with type 2 diabetes but may also provide valuable information about the clinical progression of the disease to insulin requirement. To understand the genetically determined abnormalities which result in a predisposition for type 2 diabetes we need to identify the physiological and biochemical factors involved in the accumulation of IAPP as islet amyloid.

X. CONCLUSION

The physiological function of IAPP remains unresolved partially since its effects and the receptors specific for IAPP have yet to be identified. The proposal to regulate hepatic glycogen stores in type l diabetes by combined therapeutic use of IAPP and insulin'46 may be premature whilst a physiological role for IAPP remains unclear. However, even though the cause of islet amyloid formation remains unknown, the effects of the deposits in the islets are clearly associated with decreased islet function and, in severe amyloidosis, destruction of 8-cells. In sumary, islet amyloid is a characteristic feature of type 2 diabetes and forms a unifying pathological marker for the heterogeneous clinical features of the disease. IAPP, the peptide component of islet amyloid, is a normal constituent of islet B-cells. However, unlike in many systemic forms of amyloidosis, there is no evidence for a molecular abnormality of IAPP in amyloid and little evidence for significant hypersecretion of the peptide in type 2 diabetes. Although amyloid formation precedes the onset of hyperglycaemia in animals, it seems unlikely that small amounts of extracellular amyloid would have a causative role in the onset of the disease. However, as in other amyloidoses, progressive accumulation of the deposits results in dysfunction of the affected organ, which in type 2 diabetes is manifested by a progressive decrease in insulin secretion. The physiological role of IAPP remains unclear although extensive experimental investigations have revealed pharmacological actions of IAPP which are similar to those of CGRP. Islet amyloid

Acknowledgements I am very grateful to John Morris, Eelco d e Koning, and my collegues at the Diabetes Research Laboratories for their helpful discussions and to Victoria Summers for her assistance in the preparation of the manuscript. I thank the British Diabetic Association and Novo Nordisk for financial support.

References 1. Westermark P, Wernstedt C, O'Brien TD, Hayden DW, and Johnson KH: Islet amyloid in type 2 human diabetes mellitus and adult diabetic cats contains a novel putative polypeptide hormone. Am Pathol 127:414-417, 1987. 2. Clark A, Cooper GJS, Lewis CE, Morris JF, Willis AC, Reid KBM, and Turner RC: Islet amyloid formed from diabetes associated peptide may be pathogenic in type 2 diabetes. lancet ii:231-234, 1987. 3. Cecil RL: A study of the pathological anatomy of the pancreas in ninety cases of diabetes mellitus. J E x p Med 11:266-290, 1909. 4. Ehrlich JC, and Ratner IM: Amyloidosis of the isletsof Langerhans. Am J Pathol 38:49-59, 1961. 5. Maclean N, and Ogilvie RF: Quantitative estimation of the pancreas islet tissue in diabetic subjects. Diabetes 4:367-376, 1955. 6. Wright AW: Hyaline degeneration of the islets of Langerhans in non-diabetics. Am J Pathol 31461-482, 1927. 7. Putchler H, Sweat F, and Levine M: On the binding of Congo red by amyloid. J Histochem Cytochem 10:355-364, 1962. 8. Linker A, and Carney HC: Presence and role of glycosaminoglycans in amyloidosis. Lab Invest 57:297-305,1987. 9. Glenner GG: AmyIoid deposits and amyloidosis: the B-fibrilloses. N Engl J Med 302:1333-1343, 1980. 10. Tape C, Tan R, Nesheim N, and Kisilevsky R: Direct evidence for circulating SAA as the precursor of tissue AA deposits. Scand J lmmunol 42:139, 1988. 11. Sletten K, Westermark P, and Natvig JB: Characterisation of amyloid fibril proteins from medullary carcinoma of the thyroid. 1 E x p Med 143:993-998, 1976. 12. Kang J, Lemaire HG, Unterbeck A, Salbaum

CLARK

128

JM, Masters CL, Grzeschik K-H, Multhaup G, Beyreuther K, and Muller-Hill B: The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor. Nature 325:733-736, 1987. 13. Westermark P, Wernstedt C, Wilander E, Hayden DW, O'Brien TD, and Johnson KH: Amyloid fibrils in human insulinoma and islets of Langerhans of the diabetic cat are derived from a neuropeptide-like protein also present in normal islet cells. Proc Nut1 Acad Sci U S A 84:3881-3885, 1987. 14. Cooper GJC, Willis AC, Clark A, Turner RC, Sim RB, and Reid KBM: Purification and characterisation of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proc Natl Acad Sci U S A 84:8628-8632, 1987. 15. Opie EL: On the relation of chronic interstitial pancreatitis to the islands of Langerhans and to diabetes mellitus. J Exp Med 5:397428, 1901. 16. Opie EL: The relation of diabetes mellitus to lesions of the pancreas: hyaline degeneration of the islets of Langerhans. j Exp Med 5:527-540, 1901. 17. Bell ET: Hyalinization of the islets of Langerhans in diabetes mellitus. Diabetes 1:341-344, 1952. 18. Westermark P, and Grimelius L: The pancreatic islet cell in insular amyloidosis in human diabetic and non-diabetic islets. Acta Pathol Microbiol Scund 81:291-300, 1973. 19. Schneider HM, Storkel S, and Will W: Das amyloid der Langerhansschen inseln und seine beziehung zum diabetes mellitus. Dtsch Med Wochenschr 105:1143-1147, 1980. 20. Rahier J,Goebbels RM, and Henquin JC: Cellular composition of the human diabetic pancreas. Diabetologia 24:366-371, 1983. 21. Westermark P, Wilander E, Westermark GT, and Johnson KH: Islet amyloid polypeptide-like immunoreactivity in the islet p-cells of type 2 non-insulin dependent diabetic and non-diabetic individuals. Diabetologia 30:887-892, 1987. 22. Clark A, Wells CA, Buley ID, Cruickshank JK, Vanhegan RI, Matthews DR, Cooper GJS, Holman RR, and Turner RC: Islet amyloid, increased Acells, reduced p-cells and exocrine fibrosis: quantitative changes in the pancreas in type 2 diabetes. Diabetes Res 9:151-160, 1988. 23. Maloy AL, Longnecker DS, and Greenberg ER: The relation of islet amyloid to the clinical type of diabetes. Hum Patkol 12:917-922, 1981. 24. Clark A, Holman RR, Matthews DR, Hockaday TDR, and Turner RC: Non-uniform distribution of islet amyloid in the pancreas of 'maturityonset' diabetic patients. Diabetologia 27:527-528, 1984. 25. Clark A, Saad MF, Nezzer T, Uren C, Knowler WC, Bennett PH, and Turner RC: Islet amyloid polypeptide in diabetic and non-diabetic Pima Indians. Diabetologia 33:285-289, 1990. 26. Vishwanathan KA, Bazaz-Malik G, Dandekar J, and Vaishnava 0: A qualitative and quantitative histological study of the islets of Langerhans in diabetes mellitus. Indian J Med Sci 26:807-812, 1972.

27. Saito K, Yaginuma N, and Takahashi T: Differential volumetry of A, B and D cells in the pancreatic islets of diabetic and non-diabetic subjects. Tokoku J Exp Med 129:27>283, 1979. 28. Iannucci A, Mukai K, Johnson D, and Burke B: Endocrine pancreas in cystic fibrosis: an Pathol immunohistochemical study. H u m 15:27%284, 1984. 29. Clark A, Stead RJ, Hodson ME, Batten JC, and Turner RC: Quantitative morphometry of pancreatic islet cells in cystic fibrosis. Diabetic Med 2:514A, 1985. 30. Pearse AGE, Ewen WB, and Polak JM: The genesis of APUD amyloid in endocrine polypeptide tumours: histochemical distinction from immunoamyloid. Virchows Arck B Zellyatlzol 10:9>107, 1972. 31. Westermark P,Grimellius L, Polak JM, Larsson L1,Van Noorden S, Wilander E, and Pearse AGE: Amyloid in polypeptide hormone producing tumours. Lab Invest 37:212-215, 1977. 32. Westermark P: Amyloid of human islets of Langerhans 1. Isolation and some characteristics. Acta Pathol Microbiol Scand Sect C 83:439-446, 1975. 33. Westermark P, Wernstedt C, Wilander E, and Sletten K: A novel peptide in the calcitonin gene related peptide family as an amyloid fibril protein in the endocrine pancreas. Biochem Biophys Res Commun 140327-831, 1986. 34. Sanke T, Bell GI, Sample C, Rubenstein AH, and Steiner DF: An isIet amyloid peptide is derived from an 89 amino acid precursor by proteolytic processing. 1 Biol Chem 263:17243-17246, 1988. 35. Cooper GJS, Leighton B, Willis AC, and Day AJ: The amylin superfamily: a novel grouping of biologically active polypeptides related to the insulin A-chain. Prog Growth Factor Res 1:99-105, 1989. 36. Glenner GG, Eanes E, and Clayton A: Amyloid fibrils formed from a segment of the pancreatic islet protein. Biockem Biophys Res Cornmun 155:608-614, 1988. 37. Hubbard JA, Martin SR, Chaplin LC, Bose C, Kelly SM, and Price NC: Solution structures of calcitonin gene related peptide analogues of calcitonin gene related peptide and amylin. Bioclzem J 275:785-788, 1991. 38. Blake CCF,Geisow MJ, Oatley SJ, Rerat E, and Rerat C: Structure of prealbumin, secondary, tertiary and quaternary interactions determined by Fourier refinement at 1.8 A. j Mol Biol 121:339-356, 1978. 39. Becker JW, and Reeke GN. Three dimensional structure of p-2 microglobulin. Pror Natl Acnd Sri U S A 82:4225-4229, 1985. 40. Westermark P, Engstrom U, Johnson KH, Westermark GT, and Betzholtz C: Islet amyloid polypeptide: pinpointing amino acid residues linked to amyloid fibril formation. Proc Natl Acad Sci U S A 87:5036-5040, 1990. 41. Westermark P, Johnson KH, Engstrom U,Westermark GT, and Betzholtz C: Islet amyloid polypeptide: synthetic peptides for study of the pathogenesis of islet amyloid. In Amyloid and Amyloidosis,

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53. 54.

Natvig JB, Ferre 0, Husby G, Husebekk A, Skogen B, Sletten K, Westermark P, Eds. Kluwer, Dordrecht, 1991, pp 449-452. Ohagi S, Nishi M, Bell GI, Ensinck JW, and Steiner DF: Sequences of islet amyloid polypeptide precursors of an Old World monkey, the pigtailed macaque Macaca nemestrina and the dog Canis familiaris. Diabetologia 34:555558, 1991. Jordon K, Murtaugh MP, O’Brien TD, Westermark P, Betsholtz C, and Johnson KH: Canine IAPP cDNA sequence provides important clues regarding diabetogenesis and amyloidogenesis in type 2 diabetes. Biochem Biophys Res Commun 169:502-508, 1990. Betzholtz C, Christmanson L, Engstrom U,Rorsman F, Jordan K, O’Brien TD, Murtaugh M, Johnson KH, and Westermark P: Structure of cat islet amyloid polypeptide and identification of amino acid residues of potential significance for islet amyloid formation. Diabetes 39:11&122,1990. Nishi M, Bell GI, and Steiner DF: Sequence of a cDNA encoding Syrian hamster islet amyloid polypeptide precursor. Nucleic Acids Res 18:6726, 1990. Johnson KH, Wernstedt C, O’Brien TD, and Westermark P: Amyloid in the pancreatic islets of the cougar Felis concolor is derived from islet amyloid polypeptide IAPP. Comp Biochem Physiol B 98:115-119, 1991. Leffert JD, Newgard CB, Okamoto H, Milbunn J L , and Luskey KL: Rat amylin: cloning and tissue-specific expression in pancreatic islets. Proc Nafl Acad Sci U S A 86:3127-3130, 1989. Asai J, Nakazato M, Miyazato M, Kangawa K, Matsuo H, and Matsukura S: Regional distribution and molecular forms of rat islet amyloid polypeptide. Biochem Biophys Res Commuri 169m-795, 1990. Ferrier GJM, Pierson AM, Jones PM, Bloom SR, Girgis SI, and Legon S: Expression of the rat amylin IAPP/DAP gene. Mol Endocrinol 3:Rl-R4, 1989. Nishi M, Chau SJ, Nagamatsu S, Bell GI, and Steiner DF: Conservation of the sequence of islet amyloid polypeptide in five mammals is consistent with its putative role as an islet hormone. Proc Natl Acad Sci U S A 86:5738-5742, 1989. Nishi M, and Steiner DF: Cloning of complementary DNAs encoding islet amyloid polypeptide insulin and glucagon precursors from a New World rodent, the degu, Octodon degus. Mol Endocrinol 4:1192-1198, 1990. Kirschner DA, Inouye H, Duffy LK, Sinclair A, Lind M, and Selkoe DJ: Synthetic peptide homologous to beta protein from Alzheimer disease forms amyloid-like fibrils in vitro. Proc Natl Acad Sci USA 84:6953-6957, 1987. Howard CF: Longitudinal studies on the development of diabetes in individual Macaca nigra. Diabetologia 29:301-306, 1986. Clark A, de Koning EJP, Hansen 3, Bodkin N, and Morris JF: Islet amyloid in glucose intolerant and spontaneously diabetic Macaca mulatta monkeys. In Frontiers in Diabetes Research. Lessons from

12Y

55.

56.

57.

58.

59.

60. 61. 62.

63.

64.

65.

66.

67.

68.

Animal Diabetes 111, Shafrir E, Ed. Smith Gordon, London, 1981, pp 502-506. O’Brien TD, Westermark P, and Johnson KH: Islet amyloid polypeptide and calcitonin generelated peptide immunoreactivity in amyloid and tumour cells of canine pancreatic .endocrine tumours. Vet Pathol 27:194-198, 1990. Hellman U, Wernstedt C, Westermark P, O’Brien TD, Rathbun WB, and Johnson KH: Amino acid sequence from degu islet-amyloid derived insulin shows unique sequence characteristics. Biochem Biophys Res Cornmuti 169:571-577, 1990. Kisilevsky R: Heparan sulphate proteoglycans in amyloidogenesis: an epiphenomenon, a unique factor or the tip of a more fundamental process. Lab Invest 63:589-591, 1990. Pepys MB, Baltz M, de Beer FC, Dyck RF, Holford S, Breathnach SM, Black MM. Tribe CR,Evans DJ, and Feinstein A: Biology of serum amyloid P component. Ann N Y Acad Sci 389:28&297, 1982. Foulis AK, and Stewart JA: The pancreas in recent onset type I (insulin-dependent) diabetes mellitus: insulin content of islets, insulitis and associated changes in the exocrine acinar tissue. Diabetologia 26: 456-467, 1984. Fine structure of the islets of nsular amyloidosis. Virchows Arch A IPathol Anat] 359:l-18, 7973. Clark A: Islet amyloid and type 2 diabetes. Diabetic Med 6:561-567, 1989. Yano BL, Hayden DW, and Johnson KH: Feline insular amyloid: ultrastructural evidence for intracellular formation by non-endocrine cells. Lab invest 45:149-156, 1981. Clark A, Morris JF, Scott LA, McLay A, Foulis AK, Bodkin NL, and Hansen BC: Intracellular formation of amyloid fibrils of human insulinoma and pre-diabetic monkey islets. In Amyloid and Amyloidosis, Natvig JB, Ferre 0, Husby G, Husebekk A, Skogen B, Sletten K, Westermark P., Eds. Kluwer, Dordrecht, p p 453-456. Lukinius A,Wilander E, Westermark GT, Engstrom U, and Westermark P: Co-localisation of islet amyloid polypeptide and insulin in the 6cell secretory granules of the human pancreatic islets. Diabetologia 32:240-244, 1989. Clark A, Edwards CA, Ostle LR, Sutton R, Rothbard JB, Morris JF, and Turner RC: Localisation of islet amyloid peptide in lipofuscin bodies and secretory granules of human p-cells and in islets of type 2 diabetic subjects. Crll Tissue Res 2571179-185, 1989. Westermark P, Engstrom U, Westermark GT, Johnson KH, Permerth J, and Betzholtz C: Islet amyloid polypeptide (IAPP) and proIAPP immunoreactivity in human islets of Langerhans. Diabetes Res Clin Prac 7:219-226, 1989. Clark A, Lloyd J, Novials A, Hutton JC, and Morris JF: Localisation of islet amyloid polypeptide and its carboxy-terminal flanking peptide in islets of diabetic man and monkey. Diabetologia 34:449451, 1991. In’t Veld PA, Zang F, Madsen OD, and Kloppel G: Islet amyloid polypeptide immunoreactivity in the human fetal pancreas. Diabetologia 35:

130 272-276, 1992. 69. De Krijger RR, Kranenburg G, Stevens M, Rahier JRD, and Bruining GJ: The presence of islet amyloid polypeptide during the development of the human fetal pancreas. Diabetologia 34:A43, 1991. 70. Madsen OD, Nielsen JH, Michelsen B, Westermark P, Betsholtz C, Nishi M, and Steiner DF: Islet amyloid polypeptide and insulin expression are controlled differently in primary and transformed islet cells. Mol Endocrinol 5:14>148, 1991. 71. Toshimori H, Navita R, Nakazato M, Asai J, Mitsukawa T, Kangawa K, Matsuo H, and Matsukura S: Islet amyloid polypeptide (IAPP) in the gastrointestinal tract and pancreas of man and rat. Cell Tissue Res 262:401406, 1990. Johnson KH, O’Brien TD, Betsholtz C, and Westermark P: Islet amyloid, islet amyloid polypeptide and diabetes mellitus. N Engl J Med 321:513-518, 1989. O’Brien TD, Hayden DW, Johnson KH, and Fletcher TF: Immunohistochemical morphometry of pancreatic endocrine cells in diabetic, normoglycaemic, glucose-intolerant and normal cats. J Comp Patkol 96:357-369, 1986. 74. Johnson KH, O’Brien TD, Jordan K, and Westermark P: Impaired glucose tolerance is associated with increased islet amyloid polypeptide (IAPP) immunoreactivity in pancreatic p-cells. A m ] Patkol 135:245250, 1989. 75. Clark A, Morris JF, and Hansen BC: Pancreatic islet amyloid in non-insulin-dependent diabetes. In Diabetes. Rifkin H, Colwell JA, and Taylor SI, Eds. Elsevier, Amsterdam, 1991, pp. 492-496. 76. De Koning EJP, Clark A, Bodkin N, and Hansen BC: Correlation of islet amyloid reduced p-cell mass with p-cell function in diabetic monkeys. Diabetologia 33:397A, 1990. 77. De Koning EJP: Unpublished results. 78. Newman B, Selby JV, King MC, Slemenda C, Fabsitz R, and Friedman GD: Concordance for type 2 (non-insulin-dependent) diabetes mellitus in male twins. Diabetologia 30:763-768, 1987. 79. Hattersley A, Turner RC, Permutt MA, Patel P, Tanizawa MD, Chiu KC, O’Rahilly S, Watkins P, and Wainscoat JS: Type 2 diabetes is linked to the glucokinase gene in a large pedigree. Lancet 339:1307-10, 1992. 80. Mosselman S, Hoppener JWM, Zandberg J, van Mansfield ADM, Geurts van Kessel AHM, Lips CJM, and Jansz HS: Islet amyloid polypeptide IAPP: identification and chromosomal localization of the human gene. FEBS Lett 239:227-232, 1988. 81. Christmanson L, Rorsman F, Stenman G, and Westermark P: The human islet amyloid polypeptide (IAPP) gene. Organization, chromosomal localisation and functional identification of a promoter region. FEBS Lett 267: 16C-166, 1990. 82. Nishi M, Sanke T, Seino S, Eddy RL, Fan Y, Byers MG, Shows TB, Bell GI, and Steiner DF: Human islet amyloid polypeptide gene: complete nucleotide sequence, chromosomal localisation and evolutionary history. Mol Endocrinol 3: 17751781, 1989.

CLARK 83. Mosselman S, Hoppener JWM, Lips CJM, and Jansz HS: The complete islet amyloid polypeptide precursor is encoded by two exons. FEBS Lett 247: 154-158, 1989. 84. Steiner DF, Ohagi S, Nagamatsu S, Bell GI, and Nishi M: Is islet amyloid polypeptide a significant factor in pathogenesis or pathophysiology of diabetes? Diabetes 40: 305-309, 1991. 85. Palsdottir A, Abrahamson M, Thorsteinsson L, Amason A, Olafsson I, Grubb A, and Jensson 0: Mutation in cystatin C gene causes hereditary brain haemorrhage. Lancet ii:603-604, 1988. 86. Pras M, PreIli F, Franklin EC, and Fragione B: Primary structure of an amyloid pre-albumin variant in familial polyneuropathy of Jewish origin. Proc Natl Acad Sci U S A 30:539-542, 1983. 87. Nishi M, Bell GI, and Steiner DF: Islet amyloid polypeptide (amylin): no evidence of an abnormal precursor sequence in 25 type 2 non-insulindependent diabetic patients. Diabetologia 33:628-630, 1990. 88. Patel P, Mosselman S, Hoppener JWM, Jansz HS, Clark A, O’Rahilly S, Turner RC, and Wainscoat JS: An RFLP associated with islet amyloid polypeptide locus (IAPP). N u c k i c Acid Res 17: 6758, 1989. 89. Cook JT, Patel P, Clark A, Hoppener JW, Lips CJ, Mosselman S, O’Rahilly S, Page RC, Wainscoat JS, and Turner RC: Non-linkage of the islet amyloid polypeptide gene with type 2 noninsulin-dependent diabetes mellitus. Diabrtologia 34: 103-108, 1991. 90. Betzholtz C, Svensson V, Rorsman F, Westermark GT, Wilander E, and Johnson KH: cDNA cloning and identification of an amyloidogenic region associated with the species-specific occurrence of age related diabetes mellitus. Exp Cell Res 1853484493, 1989. 91. Nishi M, Sanke T, Nagamatsu S, Bell GI, and Steiner DF: Islet amyloid polypeptide: a new pcell secretory product related to islet amyloid deposits. J Biol Chem 265:4173-4176, 1990. 92. Van Mansfeld ADM, Mosselman S, Hoppener JWM, Zandberg J, van Teeffelen HAAM, Baas PD, Lips CJM, and Jansz HS: Islet amyloid polypeptide: structure and upstream sequences of the IAPP genes in rat and man. Biockem Biopkys Acta 1087: 235-240, 1990. 93. Mosselman S, Hoppener JWM, De Wit L, Soeller W, Lips CJM, and Jansz HS: IAPP/amylin gene transcriptional control region: evidence for negative regulation. FEBS Lett 271:33-36, 1990. 94. Alam T, Chen L, Ogawa A, Leffert J, Unger RH, and Luskey KL: Co-ordinate regulation of amylin and insulin expression in response to hypoglycaemia and fasting. Diabetes 41:508-514, 1992. 95. Bretherton-Watt D, Ghatei MA, Legon 5, Jamal H, Suda K, and Bloom SR: Depletion of islet amyloid polypeptide in the spontaneously diabetic (BB) Wistar rat. J M o l Endocrinol 6: 3-7, 1991. 96. Kanatsuka A, Makino H, Ohsawa H, Tokuyama Y, Yamaguchi T, Yoshuda S, and Adachi M: Secretion of islet amyloid polypeptide in response to glucose. FEBS L e f t 259:199-201, 1989.

ISLET AMYLOID: AN ENIGMA OF TYPE 2 DIABETES 97

98.

99

I

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

Ogawa A, Harris V, McCorkle SK, Unger RH, and Luskey KL: Amylin secretion from the rat pancreas and its selective loss after streptozotocin treatment. Clin Znvest 85:973-978, 1989. Kahn SE, d’Alessio DA, Schwartz MW, Fujimoto WY, Ensinck JW, Taborsky GJ, and Porte D: Evidence of co-secretion of islet amyloid polypeptide and insulin by p-cells. Diabetes 393634638, 1990. lnoue K, Hisatomi A, Umeda F, and Nawata H: Release of amylin from perfused rat pancreas in response to glucose, arginine, beta hydroxybutyrate, and gliclazide. Diabetes 40: 100.5-1009, 1991. Moore CX, and Cooper GJS: Co-secretion of amylin and insulin from cultured islet p-cells: modulation by nutrient secretagogues, islet hormones and hypoglycaemic agents. Biochem Biophys Res Commun 179:l-9, 1991. O’Brien TD,Westermark P, and Johnson KH: Islet amyloid polypeptide and insulin secretion from isolated perfused pancreas of fed, fasted, glucose treated and dexamethasone-treated rats. Diabetes 40:1701-1706, 1991. Jamal H, Bretherton-Watt D, Suda K, Ghatei MA, and Bloom SR: Islet amyloid polypeptide-like immunoreactivity (amylin) in rats treated with dexamethasone and streptozotocin. J Endocrinol 126:425429, 1990. Nakazato M, Miyazato M, Asai J, Mitsukawa T, Kangawa K, Matsuo H, and Matsukura S: Islet amyloid polypeptide, a novel pancreatic peptide, is a circulating hormone secreted under glucose stimulation. Biochem Biophys Res Commun 169: 713-718, 1990. Mitsukawa T, Takemura J, Asai J, Nakazato M, Nagawa K, Matsuo H, and Mutsukura S: Islet amyloid polypeptide response to glucose insulin and somatostatin analogue administration. Diabetes 39:639-642, 1990. Butler PC, Chou J, Carter WB, Wang Y-N, Bu BH, Chang D, Chang J-K, and Rizza RA: Effects of meal ingestion on plasma amylin concentration in NIDDM and non-diabetic humans. Diabetes 39: 752-756, 1990. van Jaarsveld BC, Hackeng WHL, Nieuwenhuis MG, Erkelens DW, Geerdink RA, and Lips CJW: Islet amyloid polypeptide in human plasma. Lancet i:60, 1990. Sanke T, Hanabusa T, Nakamo Y, Oki C, Okai K, Nishimura S, Kondo M, and Nanjo K: Plasma islet amyloid polypeptide (amylin) levels and their responses to oral glucose in type 2 (noninsulin-dependent) diabetic patients. Diabetologia 34: 129-132, 1991. Ludvik B, Lell B, Hartter E, Schnack C, and Prager R: Decrease of stimulated amylin release precedes impairment of insulin secretion in type 2 diabetes. Diabetes 40:16151619, 1991. Hartter E, Svoboda T, Ludvik B, Schuller M, Lell B, Kuenburg E, Brunnbauer M, Woloszczuk W, and Prager R: Basal and stimulated plasma levels of pancreatic amylin indicate its co-secretion with insulin in humans. Diabetologia 34: 52-54, 1991. Gill AM, and Yen TT: Effects of ciglitazone on endogenous plasma islet amyloid polypeptide

131

111.

112.

113.

114. 115.

116.

117.

118. 119.

120.

121.

122.

123.

124.

and insulin sensitivity in obese-diabetic viable yellow mice. Life Sci 48:703-710, 1991. Ludvik B, Berzlanovich A, Hartter E, Lell B, Prager R, and Graf H: Increased amylin levels in patients on chronic haemodialysis. Neplirol Dial Transplant 8: 694-695A, 1990. Nakazato M, Asai J, Kangawa K, Matsukura S, and Matsuo H: Establishment of radioimmunoassay for human islet amyloid polypeptide and its tissue content and plasma concentration. Bioc/ieni Biophys Res Commun 164:394399, 1989. De Koning EJP, Fleming KA, Gray DWR, and Clark A: Pancreatic islet amyloid is present in non-diabetic subjects with end-stage chronic renal failure on dialysis treatment. Diabetic Med 9:13-14A. Porte D, and Kahn SE: Hyperproinsulinaemia and amyloid in NIDDM: clues to etiology of islet p-cell dysfunction. Diabetes 38: 1333-1336, 1989. Clark A, Matthews DR, Naylor BA, Wells CA, Hosker JP, and Turner RC: Pancreatic islet amyloid and elevated proinsulin secretion in familial maturity onset diabetes. Diabetes Res 4:51-55, 1987. Cooper GJS, Leighton B, Dimitriadis GD, ParryBillings M, Kowalchuk JM, Howland K, Rothbard JB, Willis AC, and Reid KBM: Amylin found in amyloid deposits in human type 2 diabetes mellitus may be a hormone that regulates glycogen metabolism in skeletal muscle. Proc Nafl Acad Sci U S A 85: 776S7766, 1988. Leighton B, and Cooper GJS: Pancreatic amylin and calcitonin gene-related peptide cause resistance to insulin in skeletal muscle in vifro. Nature 335~632-635, 1988. Leighton B, and Foot E: The effects of amylin on carbohydrate metabolism in skeletal muscle it7 vitro and in vivo. Biochem J 269:19-23, 1990. Young DA, Deems RO, Deacon RW, McIntosh RH, and Foley JE: Effects of amylin on glucose metabolism and glycogenolysis in vizio and it7 vitro. A m J Physiol 259: E457-461, 1990. Zierath JR, Galuska D, Engstrom A, Johnson KH, Betzholtz C, Westermark P, and WallbergHenriksson H: Human islet amyloid polypeptide at pharmacological levels inhibits insulin and phorbol-ester-stimulated glucose transport in it7 vitro incubated human muscle strips. Diabetologia 35: 2&31, 1992. Young AA, Mott DM, Stone K, and Cooper GJS: Amylin activates glycogen phosphorylase in the isolated soleus muscle of the rat. FEBS Lett 281: 149-151, 1991. Frontoni S , Choi SB, Banduch D, and Rossetti L: In vivo insulin resistance induced by amylin primarily through inhibition of insulin stimulated glycogen synthesis in skeletal muscle. Diabetes 40: 568-573, 1991. Koopmans SJ, van Mansfeld AD, Jansz HS, Krans HM, Radder JK, Frolich M, de Boer SF, Kreutter DK, Andrews GC, and Maassen JA: Amylin induced in vivo insulin resistance in conscious rats: the liver is more sensitive to amylin than peripheral tissues. Diabetologia 34:218-224, 1991. Molina JM, Cooper GJS, Leighton B, and Olefsky

CLARK

132

125.

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

JM: Induction of insulin resistance in vivo by amylin and calcitonin gene-related peptide. Diabetes 39: 260-265, 1990. Sowa R, Sanke T, Hrayama J, Tabata H, Furuta H, Nishimura S, and Nanjo K: Islet amyloid polypeptide amide causes peripheral insulin resistance in vivo in dogs. Diabetologia 33:11%120, 1990. Pettersson M, and Ahren B: Failure of islet amyloid polypeptide to inhibit basal and glucosestimulated insulin secretion in model experiments in mice and rats. Acta Physiol Scand 138:389-394, 1990. Tedstone AE, Nezzer T, Hughes SJ, Clark A, and Matthews DR: The effect of islet amyloid polypeptide (amylin) and calcitonin-gene-related peptide on glucose removal in the anaesthetised rat and on insulin secretion from rat pancreatic islets in vitro. Bioscience Rep 10:339-345, 1990. Kassir AA, Upadhyay AK, Lim TJ, Moossa AR, and Olefsky JM: Lack of effect of islet amyloid polypeptide in causing insulin resistance in conscious dogs during euglycemic clamp studies. Diabetes 40:99%1004, 1991. Chantry A, Leighton B, and Day AJ: Cross reactivity of amylin with calcitonin gene related peptide binding sites in rat liver and skeletal muscle membranes. Biochern J 277: 139-143, 1991. Morishita T, Yamaguchi A, Fujita T, and Chiba T: Activation of adenylate cyclase by islet amyloid polypeptide with COOH-terminal amide via calcitonin gene-related peptide receptors on rat liver plasma membranes. Diabetes 395375877, 1990. Datta HK, Zaidi M, Wimalawansa SI, Ghatei MA, Beacham JL, Bloom SR, and MacIntyre I: In vivo and in vitro effects of amylin and amylin-amide on calcium metabolism in the rabbit. Biochern Biophys Res Commun 162:876-881, 1989. Zaidi M, Datta HK, Bevis PJ, Wimalawansa SJ, and MacIntyre I: Amylin amide: a new bone conserving peptide from the pancreas. Exp Physiol 75:529-536, 1990. Pettersson M, Ahren B, Bottcher G, and Sundler F: Calcitonin-gene-related peptide: occurrence in pancreatic islets in the mouse and the rat, and inhibition of insulin secretion in the mouse. Endocrinology 1192365-869, 1986. Pettersson M, Lundquist I, and Ahren B: Neuropeptide Y and calcitonin gene-related peptide: effects on glucagon and insulin secretion in the mouse. Endocrinol Res 13:407-417, 1987. Ahren B, Martinsson H, and Nobin A: Effects of

136.

137.

138.

139.

140.

141.

142.

143. 144.

145.

146.

calcitonin-gene related peptide (CCRP) on islet hormone secretion in the pig. Diabetologia 30:354-359,1987. Hermanson K, and Ahren B: Dual effects of calcitonin gene-related peptide on insulin secretion in the perfused dog pancreas. Rrgul Pept 27:149-157, 1990. Ohsawa H, Kanatsuka A, Yamaguchi T, Makino H, and Yoshida S: lslet amyloid polypeptide inhibits glucose-stimulated insulin secretion from isolated rat pancreatic islets. Bioclieiti Biophys Res Commun 160: 961-967, 1989. Silvestre RA, Peiro E, Degano P, Miralles P, and Marco J: Inhibitory effect of rat amylin on the insulin responses to glucose and arginine in the perfused rat pancreas. Regul P e p t 31:23-31, 1990. Fehmann H-C, Weber V, Goke R, Goke B, Eissele R, and Arnold R: Islet amyloid polypeptide (IAPP; amylin) influences the endocrine but not the exocrine rat pancreas. Biochem Biophys Res Cornmun 167:1102-1108, 1990. O’Brien TD, Westermark P, and Johnson KH: Islet amyloid polypeptide (IAPP) does not inhibit glucose stimulted insulin secretion from isolated perfused rat pancreas. Bioclzern Biophys Res Cottimun 170: 122S1228, 1990. Ar’Rajab A, and Ahren B: Effects of amidated rat islet amyloid polypeptide on glucose stimulated insulin secretion in v i m and in vitro in rats. Eur J Pharrnacol 17:443-445, 1991. Nagamatsu S,Carroll J, Grodsky EM, and Steiner DF: Lack of islet amyloid polypeptide regulation of insulin biosynthesis or secretion in normal rat islets. Diabetes 39971-874, 1980. Brain SD, Williams TJ, Tippins JR, Morris HR, and MacIntyre I: Calcitonin gene-related peptide is a potent vasodilator. Nature 313:5456, 1985. Gardiner SM, Compton AM, Kemp PA, Bennett T, Bose C, Foulkes R, and Hughes B: Antagonistic effect of human alpha calcitonin gene related peptide (%37) on regional hemodynamic actions of rat islet amyloid polypeptide in conscious Long Evans rats. Diabetes 40:94%951, 1991. Sternini C, and Brecha N: Immunocytochemical identification of islet cells and nerve fibres containing calcitonin-gene-related peptide-like immunoreactivity in the rat pancreas. G a s t r o e n t w ology 90:1155-1163, 1986. Young AA, Crocker LB, Wolfe-Lopez D, and Cooper GJS: Daily amylin replacement reverses hepatic glycogen depletion in insulin-treated FEBS Lett streptozotocin diabetic rats. 287:203-205, 1991.

Islet amyloid: an enigma of type 2 diabetes.

Islet Amyloid: An Enigma of Type 2 Diabetes Anne Clark Diabetes Research Laboratories, Radcliffe lnfirmary and Department of Human Anatomy, Universit...
3MB Sizes 0 Downloads 0 Views