Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Critical Review

Islet Inflammation in Human Type 1 Diabetes Noel G. Morgan1*


Pia Leete1
Mellitus Alan K. Foulis2
Sarah J. Richardson1

1
Institute of Biomedical and Clinical Sciences, University of Exeter Medical
School, Exeter, UK
2
GG&C Pathology Department, Southern General Hospital, Glasgow, UK

Abstract
Type 1 diabetes mellitus (T1DM) is caused by the selective against specific islet antigenic peptides. The T cells are often
deletion of pancreatic b-cells in response to an assault accompanied by influent CD201 B cells, although new data
mounted within the pancreas by infiltrating immune cells. imply that the proportions of these individual cell types vary
However, this apparently clear and focussed annunciation and that patients fall into at least two distinct categories hav-
conceals a stark reality in which the cellular and molecular ing either a hyper-immune (CD20Hi) or a pauci-immune
events leading to b-cell loss remain poorly understood in (CD20Lo) phenotype. The overall rate of b-cell decline appears
humans. This reflects the difficulty of studying these proc- to correlate with these two phenotypes such that hyper-
esses in living individuals and the fact that, using pathological immune patients lose b-cells more quickly and tend to
specimens, islet inflammation has been analysed in fewer develop disease at an earlier age than those with the pauci-
than 200 recent-onset cases of T1DM worldwide, over the immune profile. In this article, we review the evidence which
past century. Nevertheless, insights have been gained and the underpins our current understanding of the aetiology of T1DM
composition of the islet infiltrate is being disclosed. This is and highlight both the established features as well as areas
shown to be primarily lymphocytic in nature, with populations of on-going ambiguity and debate. V C 2014 IUBMB Life,
of both CD81 and CD41 T cells displaying an autoreactivity 66(11):723–734, 2014

Keywords: cell death; pancreatic islet cells; b-cell; islets of Langerhans;


Insulin; Insulitis

Introduction article, we will attempt to highlight these ambiguities and indi-


cate where active research efforts are still required.
Type 1 diabetes mellitus (T1DM) in humans is considered to
There are various reasons why, almost 100 years after the
have an autoimmune aetiology in which the insulin-secreting
discovery of insulin, the molecular basis of human T1DM
b-cells of the pancreatic islets are destroyed selectively by
remains shrouded in mystery but, foremost among these, is
influent immune cells responding to the aberrant presentation
the fact that the pancreas is inaccessible to non-invasive inves-
of b-cell antigens (1–7). However, this seemingly straightfor-
tigation in humans. Currently there are no reliable ways to
ward summary conceals a considerable degree of ambiguity
image the organ such that the cellular and molecular events
that still surrounds almost every stage of the process. In this
associated with islet inflammation can be studied in living indi-
viduals, although efforts are continuing towards the achieve-
ment of this goal (8). Thus, the limited information which is
C 2014 International Union of Biochemistry and Molecular Biology
V
available has been derived from samples recovered post-
Volume 66, Number 11, November 2014, Pages 723–734
mortem from patients with T1DM (9–11), from glands recov-
Address correspondence to: Noel G. Morgan, Institute of Biomedical and
ered at the time of organ donation (6,12–14) or from surgical
Clinical Sciences, University of Exeter Medical School, Barrack Road,
Exeter EX2 5DW, UK. Tel: +01392 408300. biopsy samples recovered from living patients (15,16). Unfortu-
E-mail: n.g.morgan@exeter.ac.uk nately, even among these valuable resources, there are impor-
Received 27 October 2014; Accepted 17 November 2014 tant issues which militate against a full understanding of the
DOI 10.1002/iub.1330 disease pathology. First, access to the whole pancreas is only
Published online 7 November 2014 in Wiley Online Library possible at the end of a patient’s life and this is usually many
(wileyonlinelibrary.com) years after disease diagnosis. Second, the bulk of the pancreas

IUBMB Life 723


IUBMB LIFE

Insulitis patterns do not differ between autopsy or organ donor cases. Photomicrographs of pancreas sections from patients
FIG 1 with type 1 diabetes mellitus collected either at the time of autopsy (A) or upon organ donation (B). Sections were immuno-
stained for insulin (brown) and glucagon (red), and each image shows an individual islet. Evidence of immune cell infiltration
(seen as small nucleated cells; black arrows) is observed around each of the islets. [Color figure can be viewed in the online
issue, which is available at wileyonlinelibrary.com.]

plays a role in digestion, and during the post-mortem phase, sise that it is not our purpose to discuss in detail the data
the organ is often subject to autolysis such that the state of which have arisen from the study of the various animal models
preservation of the tissue may be less than optimal when it is of T1DM. This is not because we consider that these cannot
recovered. This may be offset by the rapid processing of an provide important insights (they clearly do!) but, rather,
organ at the time of donation in heart-beating donors; how- because the available evidence implies that the human disease
ever, such glands are rarely available from patients with is not recapitulated fully in any of the available animal models.
recent-onset T1DM. Figure 1 illustrates the process of insulitis
in individual islets from pancreases collected either at autopsy
or organ donation. The biopsy approach could, in principle,
provide a route by which many of these problems are over- b-Cell Loss in Type 1 Diabetes Mellitus
come; however, in practice, this also presents difficulties. Early studies of the pancreatic gland recovered from patients
Some of these are technical given that the surgical or post- with T1DM identified the presence of small nucleated cells
operative procedures can lead to unexpected complications, as around the periphery of (and sometimes within) the islets of
has happened recently in the DiViD study from Norway (16). Langerhans (19–21). These were correctly considered to be
In addition, this approach relies on the assumption that the immune cells, and it is now clear that such cells congregate in
disease process will inevitably be captured in the biopsied the vicinity of the islets during disease progression. However,
region; however, this does not necessarily hold true as there is islets do not become inflamed in synchrony; rather, the pro-
strong evidence that T1DM progresses in a focal manner. As cess follows a focal course. Thus, in patients studied soon after
such, both temporal and spatial differences exist in the distri- diagnosis, regions of the pancreas can be identified in which
bution of inflamed islets and, collectively, these limit the normal, apparently healthy, islets are found, whereas other
amount of useful information that may be available from regions exist (often located in close proximity) containing islets
within the biopsied area. in which b-cell destruction is complete (4,9,11,22). Such islets
Based on this finding, it will already be clear that a are sometimes referred to as ‘pseudoatrophic’ (18,23) to
detailed study of the cellular aetiology of T1DM in humans is denote the fact that although b-cells are absent, they retain a
not a trivial matter and that the quest to provide a more com- normal complement of the other islet endocrine cells. This
plete picture continues. In particular, consortia around the raises an important point in that immunological approaches
world are collaborating to develop new collections of samples have been (and remain) the mainstay of pathological investiga-
which build on the extant historical collections such that the tion and all conclusions about the loss of b-cells depend on an
process can be studied in molecular detail (6,12). Nevertheless, analysis of the presence (or absence) of immunoreactive insu-
we should emphasise that conclusions about the underlying lin as a means to identify these cells. In studies of T1DM pan-
pathology of human T1DM are drawn from the study of fewer creas, the loss of insulin immunoreactivity has almost univer-
than 200 cases worldwide. These cases and their historical sally been interpreted as synonymous with b-cell ablation;
context have been summarised very comprehensively in recent however, it should be understood that this has rarely been
reviews by In’t Veld (17,18) and will not be rehearsed here, proven conclusively. This is mainly because there are no other
although we will draw on the available information to present known immunological markers that define human b-cells
a contemporary view of the process of islet inflammation in uniquely. Thus, it is theoretically possible that degranulation
human T1DM. In stating this objective, we should also empha- of b-cells could be misinterpreted as cell ablation during

724 Process of Islet Inflammation in Human T1DM


Islet architecture is altered during b-cell destruction. Individual islets from a single nPOD case (6228) were immunostained for
FIG 2 insulin (brown) or glucagon (red). Islets in the initial phase of peri-insulitis tend to have a normal architecture with both a- and
b-cells present (A). Islets in which most of the b-cells have been destroyed (B) are usually smaller and adopt a more condensed
appearance. Occasionally, insulin-deficient islets (IDIs) adopt a more diffuse appearance (C), with loss of their typical cellular
organisation. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

histopathological analysis. One extension of this idea (which is feature is characteristic of the pseudoatrophic islets mentioned
increasingly being mooted as a possible cause of b-cell decline above, and these are taken to represent islets in which b-cell
in T2D; refs. 24–26) is that b-cells might undergo a process of loss is complete and in which the remaining cells have adopted
de-differentiation. In this view, the cells would not necessarily a reorganised orientation to occupy the newly available space.
be lost in significant numbers but, rather, they would lose Occasionally, insulin-deficient islets may also adopt a more dif-
insulin immunopositivity and acquire a more ‘mesenchymal’ fuse appearance suggesting that the demise of the b-cells can
phenotype associated with the expression of stem cell markers also lead to a more generalised loss of cellular organisation
such as Oct4, Ngn3 and vimentin. Although this finding has (Fig. 2C).
not yet been evaluated in a fully systematic way in the pan- These considerations raise a further important matter
creases of patients with T1DM (or, arguably, in human sub- which also remains largely unresolved. This relates to the
jects with T2D), our view from the examination of many pan- mechanism by which b-cell loss occurs in T1DM and what sub-
creatic glands recovered from a wide variety of patients with sequently happens to the associated debris. It has been widely
T1DM is that the vast majority of islet cells stain positively for supposed that the primary mechanism of b-cell death is by
one of the relevant hormones (glucagon, somatostatin, pancre- apoptosis (27–29) and that residual b-cell components are
atic polypeptide or ghrelin). Hence, we do not find islets con- cleared rapidly by macrophages which normally patrol and
taining large numbers of hormone-depleted cells. This implies monitor the islet milieu (30). Such hypotheses are based
that neither complete b-cell degranulation nor b-cell de-differ- mainly on evidence arising from in vitro studies which show
entiation is a frequent occurrence in human T1DM, and it that pro-inflammatory cytokines (e.g., interleukin-1b, inter-
seems reasonable to conclude that immune-mediated b-cell feron-c and tumour necrosis factor-a) can promote b-cell apo-
ablation remains the primary mechanism of loss. This does ptosis (31–35). Thus, it is concluded that if similar cytokines
not mean, of course, that processes such as de-differentiation bathe the endocrine cells in inflamed islets, then enhanced b-
or selective degranulation might not occur in some b-cells; cell apoptosis is likely to ensue. Increased apoptosis might also
however, we are not persuaded that they could account for the derive from Fas-mediated b-cell toxicity (35–38) or from
bulk of the apparent cell loss in T1DM. enhanced endoplasmic reticulum stress (39) as has been sug-
A further feature which suggests that the major mecha- gested in some studies. The concept of an apoptotic mode of b-
nism of insulin depletion in T1DM occurs by b-cell loss is that cell death has received limited support in that although apo-
the architecture is progressively altered in those islets display- ptotic b-cells have occasionally been identified in inflamed
ing reduced (or absent) b-cells. Islets frequently adopt a more islets (40), they are not usually present in large numbers, even
condensed appearance upon light microscopic examination as in those islets which appear to be entering the most active
they lose b-cells and they are often smaller and less ovoid in phase of destruction. This may simply reflect the efficiency of
appearance (as judged on sections of pancreas; Fig. 2). This the phagocytic clearance mechanisms and the fact that in any

Morgan et al. 725


IUBMB LIFE

Representative staining of immune cell subtypes in an islet of a patient with type 1 diabetes mellitus. Serial sections of the
FIG 3 same islet from nPOD case 6052 were stained to reveal the presence of the different immune cell subtypes and endocrine cells
using either immunofluorescence (A–C) or immunohistochemistry (D). Panel A: Insulin1 b-cells (blue), CD451 lymphocytes
(green); panel B: Glucagon1 a-cells (red), CD201 B cells (green); and panel C: CD81 cytotoxic T cells (red), CD681 macrophages
(green). Nuclei were visualised with either ToPro3 (A) or DAPI (B and C). Panel D: CD41 T-helper cells were labelled immuno-
histochemically (brown) and their nuclei counterstained with haematoxylin (blue).

pancreas sample, only a single point in time is being surveyed. a specific zinc transporter, ZnT8) and the cytosolic enzyme
Alternatively, it might mean that non-apoptotic mechanisms glutamate decarboxylase. Patients often develop antibodies to
also operate. This could be the case if, for example, the multiple autoantigens, and there is an on-going debate as to
release of granzymes and/or perforin from influent immune whether these antibodies are truly pathogenic or if they are
cells contributes to b-cell loss (41–44). However extensive islet generated secondarily, as markers of an underlying disease
cell necrosis might be expected to provoke a more intense process. Most evidence favours the latter interpretation,
immune response (as cellular contents will more readily enter although it should also be emphasised that individuals can
the extracellular space) and lead to widespread, less targeted develop autoantibodies but never progress to clinical T1DM.
damage. This is rarely seen upon histological examination of Thus, there is no absolute relationship between the two. What
T1DM pancreas and it remains unclear exactly how b-cells die is clear is that the presence of multiple autoantibodies is
in human T1DM. strongly predictive of diabetes development in susceptible indi-
viduals (50,51) and that non-diabetic subjects with three or
more circulating autoantibodies have a greatly increased risk
Islet Autoantibodies and Type 1 of progressing to T1DM.
Diabetes Mellitus
The definition of T1DM as an autoimmune disease has been
driven by the observation that many (although not all) patients
The Inflammatory Infiltrate in Type 1
develop circulating antibodies to particular b-cell proteins
(5,45–49). These include insulin itself (which is frequently the Diabetes Mellitus
earliest antigen to elicit an antibody response), certain secre- Dogma (or, at least, accepted wisdom) states that the islets of
tory granule proteins (such as a protein phosphatase, IA2, and patients with T1DM are inflamed; however, in practice, this

726 Process of Islet Inflammation in Human T1DM


Representative photomicrographs of islets from CD20Hi and CD20Lo cases among the nPOD collection. Serial sections of islets
FIG 4 from either an nPOD CD20Hi (A, C and E) or an nPOD CD20Lo (B, D and F) case were stained to identify the presence of CD201
B cells (green; A and B) and insulin1 b-cells (light blue; C and D). Merged images (E and F) also show the cell nuclei (stained
dark blue with DAPI). Increased numbers of CD201 B cells are observed around and within the islets of the CD20Hi cases when
compared with the CD20Lo cases. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

has been observed much less frequently than might be imag- often rather modest. Thus, when islets are observed in pan-
ined. Indeed, in one recent review, it was noted that human creas sections (i.e., in two dimensions), small numbers of
insulitis is so rare that most pathologists will not witness a sin- immune cells may be excluded from view because they lie
gle case during their entire careers as practitioners (18)! either above or below the plane of the section. As a result, an
Nevertheless, there is abundant evidence from the available islet might then be scored by an observer as ‘non-inflamed’,
pathological collections that insulitis does occur in human whereas in reality, it contains a few immune cells elsewhere
T1DM albeit in a relatively low (10%) proportion of islets. within its volume. It is also evident that insulitis is seen most
This is partly because the inflammation dissipates once b-cells frequently in patients with recent-onset T1DM and that this
are lost and that most pancreases of patients with T1DM con- frequency declines with disease duration.
tain a large number of insulin-deficient islets. It may also Despite these difficulties, both the number and the pheno-
reflect the focal nature of the disease and the fact that by con- type of the influent immune cells have been studied in the
trast with animals such as the non-obese diabetic mouse, the inflamed islets of humans with T1DM. Early experiments
total number of immune cells infiltrating any given islet is showed that the infiltrate is predominantly lymphocytic in

Morgan et al. 727


IUBMB LIFE

nature (Fig. 3A; ref. 52) and that it contains CD81 T cells (Fig.
3C), which are considered to be among the primary cytotoxic
mediators (53). However, additional immune cells are also
present, including CD41 T cells (Fig. 3D), B cells (Fig. 3B) and
macrophages (Fig. 3C; refs. 4 and 53). In addition, the sur-
rounding pancreatic exocrine tissue has recently been
reported to be enriched in both lymphocytes (54) and neutro-
phils (55) in T1DM and the suggestion made that these might
contribute to disease progression. Neutrophils do not appear
to be present within (or near to) inflamed islets in large num-
bers, and hence, their role remains ambiguous. In some stud-
The percentage of islets which retained insulin
ies, NK cells have also been found in the islet infiltrate (13), FIG 5 immunopositivity at the time of death in patients
although this is not universally observed (53). with recent-onset type 1 diabetes mellitus was higher
These considerations raise the issue of exactly how islet in the pauci-immune (CD20Lo) cases than in those
inflammation (insulitis) is defined. This is important because, with the hyper-immune (CD20 Hi) profile. The per-
centage of residual insulin-containing islets (ICI) was
with attempts to increase the number of available organs for
calculated in pancreas sections from among a cohort
study, it has been discovered that immune cell infiltration can of patients with recent-onset type 1 diabetes mellitus
occur in the pancreas in association with periods spent by sub- collected within the United Kingdom. These had
jects in intensive care units, prior to organ recovery (56). This been classified as either CD20Lo (green) or CD20Hi
occurs independently of the presence or absence of T1DM and (red) on the basis of their insulitis profiles. The
pauci-immune (CD20 Lo) cases retained more ICIs
could lead to misdiagnosis of insulitis in pathological specimens.
than those with a hyper-immune (CD20Hi) pheno-
Fortunately, a comprehensive consensus definition of insulitis type. [Color figure can be viewed in the online issue,
has recently been proposed (23). This deals with various aspects which is available at wileyonlinelibrary.com.]
of the pathological appearance of inflamed islets and concludes
that insulitis can be confirmed in a patient with T1DM if three or
more islets are shown to contain 15 or more lymphocytes in the T1DM, whereas the second is quite different. The main differ-
peri-islet region and/or within the islet structure. This definition ence lies in the number of influent CD201 cells which, as
does not, of course, define insulitis at the level of any given indi- noted above, can be extensive and mapped in parallel with the
vidual islet, and this remains a discretionary consideration on influx of CD81 cells or, as revealed in the more detailed recent
the part of individual investigators. In our hands, we have rarely study, can be minimal (or absent). As a consequence, the two
seen individual islets which contain more than five immune cells distinct profiles have been termed ‘CD20Hi’ or ‘CD20Lo’ to
(lymphocytes or macrophages) on a single cross-section of the denote the differential involvement of these cells (Fig. 4). In
islet in non-diabetic subjects, and therefore, we have used this fact, even this may be something of a misnomer as the abso-
as an operational threshold (53). lute numbers of all immune cell subtypes are reduced among
An analysis of the composition of the immune cell infiltrate patients displaying the CD20Lo profile, and in recognition of
in islets at apparently different stages of b-cell destruction (as this finding, an alternative designation of ‘hyper’- or ‘pauci’-
judged by the extent of residual insulin immunopositivity) has immune, respectively, has also been suggested.
led to the conclusion that CD81 T cells are the predominant In both forms of insulitis (CD20Hi and CD20Lo), it is clear
cell type throughout the period of inflammation (4,53,57). This that CD81 T cells predominate and it is likely, then, that these
is consistent with the notion that these cells ultimately mediate will mediate the demise of the b-cells in both situations.
b-cell death. Interestingly, however, the B-cell (CD201) profile Importantly, the two profiles also appear to reflect patient dif-
also changes during disease progression and, in an initial ferences rather than being islet-specific (or lobular) patterns.
study, was found to align closely with the migration of CD81 T This has not been evaluated completely as it would require the
cells (53). By contrast, the numbers of CD41 T cells and mac- analysis of all inflamed islets across an entire pancreas and
rophages (defined with anti-CD68) were much less variable this would represent a gargantuan task. Nevertheless, based
during the period of b-cell decline. This suggests the existence on the more limited analysis of islets from different regions of
of a dynamic interplay between some (at least) of the immune a given pancreas, it appears that patients with T1DM fall into
cell subsets during the progression of T1DM and this remains one of the two categories. The factor that then differentiates
to be understood. Moreover, very recent data imply that these the two is the apparent rate at which the b-cells are killed.
initial observations might represent an over-simplification and This cannot, of course, be deduced with complete certainty but
that the profile of insulitis is more variable (57). This variabili- the pathological evidence can be reconstructed to make a
ty is not simply stochastic but, rather, two closely regulated strongly supportive case. This is based on several notable fea-
patterns of insulitis can be defined. The first of these mirrors tures. First, the number of residual insulin-containing islets in
the pattern reported by Willcox et al. (53) based on the analy- patients displaying the CD20Lo profile is greater than in those
sis of insulitis in a cohort of 29 patients with recent-onset who are CD20Hi (Fig. 5). This implies that b-cell killing is less

728 Process of Islet Inflammation in Human T1DM


efficient in patients with the CD20Lo profile than in those who pendent of antibody synthesis and secretion. One possibility is
are defined as CD20Hi. In support of this, the residual insulin- that the B cells may be acting as local antigen-presenting cells
containing islets also tend to contain greater numbers of b- within the islet milieu and that they collaborate to promote the
cells in CD20Lo cases. Second (and, perhaps, most strikingly), b-cell-directed cytotoxic activity of CD81 T cells (61).
when a cohort of patients was studied who were diagnosed Recent studies have attempted to assess the specificity of
with T1DM within a fixed range of ages (between 0 and 20 the infiltrating CD81 cells using tetramer staining in which the
years), the group which displayed the CD20Hi profile was, on target antibodies are loaded with potentially autoreactive pep-
average, diagnosed 6 years younger than those designated tides derived from islet antigens (62). This has revealed that the
CD20Lo. Taken together, these results imply that two differing specificity of the total population of CD81 cells is relatively
profiles of insulitis exist in patients with T1DM and that these wide in that specific sub-population and appears to be reactive
are associated with either a more (CD20Hi) or a less (CD20Lo) against a range of different antigenic peptides. However, it was
aggressive autoimmune attack on the b-cells. also revealed that the majority of such cells within the vicinity
The factors that determine which profile occurs in any of any given islet display a common autoreactive phenotype,
given patient are entirely obscure (although genetic influences suggesting that they may be clonal in origin. Moreover, it
are an obvious, but untested, possibility); however, these new appears that the influent cells target defined b-cell antigens,
data offer an important perspective on the design of clinical implying that they are truly autoreactive (62). Whether there
trials intended to slow or halt the progression of autoimmun- are also additional CD81 cells present which target non-b-cell
ity. For example, it was previously proposed that the adminis- antigens (such as virus proteins) remains to be established.
tration of an anti-B-cell therapy might be effective in patients One subtype of immune cells which has not received as
with T1DM and trials with rituximab (a monoclonal anti-CD20 much attention is the population of CD41 T cells present
antibody; refs. 58 and 59) have been undertaken. These trials within insulitic infiltrates. This may be because their numbers
involved a short-term administration of rituximab followed by do not alter dramatically during the progress of b-cell destruc-
a lengthy period of follow-up, and they were, at best, only par- tion; however, it also remains unclear whether these cells dis-
tially successful. At first sight, this is disappointing, but the play a Th1 or Th2 phenotype. In future studies, it will be
newly available data on immune cell infiltration suggest that it important to determine the Th phenotype of these cells and to
might be premature to close the matter without further consid- examine whether this differs according to the CD20Hi or
eration. In particular, it is noteworthy that the mean age of CD20Lo status. In an important recent work, CD41 T cells
disease diagnosis among the patients recruited to the rituxi- were grown out from the islets of a patient with T1DM and
mab trials was 19 years (58,59), and, as such, it seems plausi- were shown to be reactive against epitopes present within
ble that many of them might have displayed the CD20Lo profile proinsulin in an HLA-restricted manner (63). Thus, these cells
of insulitis (although this prediction cannot be verified since it were shown to be truly autoreactive, and it was suggested that
is not possible to interrogate their pancreas). If so, then anti- they could be involved in b-cell destruction (63). Few, if any, of
CD20 antibody is likely to have been minimally effective the infiltrating CD41 cells can be classified as Tregs as they
(assuming that its efficacy reflects the targeting and deletion of do not stain positively for the presence of FoxP3 (53), a marker
CD201 cells having access to inflamed islets rather than sim- which defines this class.
ply those within the circulation). By contrast, administration of
rituximab showed more (albeit still limited) protection in the
younger patients. As such patients might be expected to pref-
Recruitment of Immune Cells to
erentially display the CD20Hi profile of insulitis, this may be Inflamed Islets
indicative of an altered insulitic profile in response to the anti- The fact that the number of infiltrating immune cells sur-
CD20 therapy, at least temporarily. Thus, is it conceivable that rounding (and/or within) inflamed islets is often modest pro-
renewed attempts to deplete CD201 cells in a selected sub- vokes questions relating to the nature of the attractant that
group of (younger-onset) patients might yield more striking draws these cells to the site and their origin. It is frequently
results? assumed that the immune cells migrate to the islets mainly
As antibodies are generated from activated B cells and from influent blood vessels and that they extravasate in the
there is evidence that B cells may comprise a significant pro- immediate vicinity of the islets (possibly from efferent capilla-
portion of the immune cell infiltrate in certain patients, it is ries carrying blood to collecting venules leaving the islet
important to understand whether islet-recruited B cells are milieu); however, this has not been formally proven. There is
actively secreting antibodies within the vicinity of islets. During little evidence that the cells reach the islet by egress from ves-
activation and differentiation into plasma cells, B cells usually sels which penetrate to the core of the structure as the mor-
down-regulate the expression of CD20 and up-regulate CD138 phology of the insulitis places the influent cells mainly in the
(60). However, islet-infiltrating B cells retain CD20 and they surrounding pancreatic parenchyma adjacent to islets (often
are not labelled by antisera directed against CD138, a marker close to one pole of an islet) rather than within the islet per se.
of terminally differentiated, antibody-secreting plasma cells It is commonly argued that immune cells are drawn to
(60). Thus, this population appears to play a role that is inde- islets by a chemokine gradient elaborated from the endocrine

Morgan et al. 729


IUBMB LIFE

Patients with type 1 diabetes mellitus with hyper-immune (CD20Hi) and pauci-immune (CD20Lo) phenotypes may follow differ-
FIG 6 ential paths of disease progression and have different rates of insulin secretion at disease onset. A hypothesis to describe the
progression of b-cell loss in patients with either a hyper-immune (CD20Hi) or pauci-immune (CD20Lo) phenotype of insulitis
(adapted from an original schema proposed by Eisenbarth; ref. 7). The functional b-cell mass of individuals with a genetic sus-
ceptibility to type 1 diabetes mellitus is shown on the y-axis and their relative age on the x-axis. Following an initial trigger, the
decline in b-cell mass occurs more rapidly in those individuals with the hyper-immune phenotype (red line), and overt diabetes
is diagnosed at an earlier age. At this point (A), few residual b-cells are retained within the pancreas, and diabetes results from
the reduced b-cell mass. By contrast, b-cell mass declines more slowly in individuals with the pauci-immune phenotype (yellow
line), and disease onset occurs at an older age. In these individuals, the proportion of islets still containing residual insulin at
the time of diagnosis is increased (compare B with A), suggesting that both b-cell loss and an insulin secretory defect (or a
greater requirement for endogenous insulin) could contribute to disease onset. [Color figure can be viewed in the online issue,
which is available at wileyonlinelibrary.com.]

cells themselves, and there is evidence that these cells can obvious route by which islet autoimmunity might be initiated
synthesise and release a range of relevant chemokines such as (71). Evidence of enteroviral infection has been reported in
CXCL10, MCP-1 and MIP-1a (64–68). If this is the case, then it human b-cells from patients with T1DM (13,71–73), and this
seems reasonable to deduce that this may represent one can occur in islets which display no signs of inflammation (74).
means by which the b-cells become visible to the immune cells Thus, it is possible that insulitis follows from the establishment
during an early phase in the development of T1DM. A second of an enteroviral infection, and this idea represents a working
mechanism occurs via the up-regulation of MHC class 1 mole- hypothesis which is still being pursued by many workers glob-
cules on the b-cell surface which, although not seen in all ally (71). Irrespective of the precise nature of the insult, it
islets, nevertheless represents a striking histological feature in seems probable that the extent to which b-cells become visible
patients with T1DM (9,69,70). Thus, it seems possible that the to the immune system is increased as an early event in the
primary ‘defect’ in T1DM may lie at the level of the b-cells development of T1DM and that insulitis develops as a conse-
themselves rather than in the immune system, as a dysfunc- quence of this.
tional immune system which aberrantly targets the b-cells As noted above, one of the consistent morphological obser-
should be largely independent of chemo-attractants and MHC- vations arising from the study of human insulitis is that the
1 hyper-expression. lesion tends to be diffuse and localised mainly at the periphery
One obvious possibility that continues to attract attention of each islet. There are certainly occasions when immune cells
as a means to explain why b-cells might become the target for can be detected among the endocrine cells within the islet
autoimmunity is that they are mounting an active response to structure; however, this is not seen in the majority of islets.
an environmental insult. This would then represent the pri- This arrangement implies that the influent immune cells may
mary ‘triggering’ event which initiates disease progression. be prevented from accessing the interior of the islet by the
Considerable effort is currently being invested to establish presence of a physical barrier which limits access. In support
whether this trigger could be a viral (particularly an enterovi- of this, it is known that islets are surrounded by a basement
ral) infection because such a mechanism would provide an membrane which serves to delineate the islet structure and

730 Process of Islet Inflammation in Human T1DM


which, following their isolation, allows islets to retain their it has been possible to examine insulin secretory responses in
three-dimensional organisation in vitro (75–78). The composi- vitro from islets isolated from at least three patients with
tion of this structure has not been defined systematically but it T1DM. One of these was a young woman who died 13 years
is almost certainly composed of a complex mixture of proteins after an initial diagnosis of diabetes (at the age of 19) (14).
and proteoglycans, including collagens, hyaluronic acids and Based on the age profiles of the patients with CD20Hi and Lo,
heparan sulphates. Thus, to access the islet interior, it is prob- it might be speculated that this patient would feature among
able that this physical barrier must be breached, and an the CD20Lo group at the time of diagnosis, but this conclusion
examination of the morphology of inflamed islets implies that is necessarily speculative. Nevertheless, histological analysis
this occurs at specific points around the perimeter of the islet suggested that the majority of islets were insulin deficient at
rather than as a more generalised dissolution of the basement the time of death with only a small proportion (15%)
membrane components. One possibility is that, on arrival retaining insulin immunopositivity. Such islets had reduced
within the peri-islet region, specific immune cells are induced insulin and elevated glucagon contents when compared
to secrete relevant proteases and/or other hydrolases that with controls, consistent with on-going, although still incom-
selectively degrade the basement membrane to facilitate the plete, b-cell destruction. The islets displayed a strong insulin
entry of the cytotoxic cells (79,80). If this is the case, then it secretory response to glucose in vitro, suggesting that, at least
might offer a therapeutic opportunity as the administration of upon isolation, there was no major secretory defect and that
inhibitors that attenuate the activity of such enzymes could be T1DM had resulted from insufficient b-cell mass in this
effective in slowing the progression of insulitis. The nature of patient.
the specific immune cell subset responsible for such hydrolase A second patient (18 years) who was studied immediately
secretion has not been established and immunolabelling of rel- after disease diagnosis (81) also retained insulin-positive islets
evant subsets in human pancreas sections has not, so far, which could be harvested following isolation; however, in this
revealed any unique cell type whose arrival appears to pre- case, no insulin secretory response to glucose (or forskolin)
sage the breaching of the basement membrane. Nevertheless, was evident, leading the authors to conclude that islet function
this possibility warrants further consideration, especially as was ‘disproportionately impaired’ (81). The third patient was a
there is increasing evidence that certain proteoglycans are lost girl of 14 years who died 8 months after diagnosis of T1DM
from islets during the progression of T1DM. and whose insulin-containing islets were harvested and then
studied after various periods of tissue culture (82). In perifu-
sion studies, these islets initially showed a modest but much
Insulin Secretion From Islets Isolated reduced secretory response to glucose when compared with
those isolated from a non-diabetic control subject. If the islets
From Patients with Type 1 Diabetes
were maintained in culture for a longer period (17 days), they
Mellitus recovered secretory function and became indistinguishable
It has been implicit in much of the above discussion that from controls in terms of the magnitude of the response to glu-
T1DM is caused by net b-cell loss, and many researchers have cose. Clearly, in total, these data are insufficient to allow firm
extrapolated from empirical observations to argue that the conclusions to be drawn, but it may be significant that an insu-
clinical symptoms arise when the extent of this loss reaches lin secretory defect was observed in the islets of two of three
80% (i.e., when 20% of the original b-cell number remains). patients with T1DM, suggesting that this could have contrib-
However, this has not been formally proven. Moreover, the uted to the disease phenotype in these individuals.
newly developed dual phenotype hypothesis (summarised in
Fig. 6) in which the inflammatory process is envisaged to pro-
gress by one of two distinct mechanisms (the CD20Hi and
Endocrine Cell Proliferation in Inflamed
CD20Lo phenotypes described above) leads to a situation in
which T1DM occurs when differing proportions of residual Islets
insulin-containing versus insulin-deficient islets remain. The A further area of interest and importance in T1DM is the
reasons for this are unknown; however, one possibility is that extent to which islet cells might attempt to regenerate in the
T1DM is not solely attributable to the absolute loss of b-cells face of an autoimmune insult. It is widely understood that b-
(which undoubtedly occurs in all patients) but also as a con- cell replication occurs at only a very low rate beyond the
tributory factor to a decline in the insulin secretory capacity of immediate post-natal period in humans (83). Indeed, in adults,
any residual b-cells (Fig. 6). This might be particularly true in the rate of b-cell proliferation is vanishingly small, and there
patients with a CD20Lo profile of islet inflammation where the seems little prospect, therefore, that islet cells could be
decline in b-cell numbers occurs more slowly and insulin- replaced at a sufficient rate to offset their decline during the
containing islets are present in greater proportion (relative to autoimmune attack leading to T1DM. Nevertheless, it appears
insulin-deficient islets) at diagnosis (Fig. 5). that islet cells are not inert in the face of this assault and that
There are rather few data available which allow firm con- they can respond by increasing their rate of mitosis. This was
clusions to be drawn in relation to these arguments; however, shown in both an older patient (84) and in two studies of

Morgan et al. 731


IUBMB LIFE

pancreas samples harvested post-mortem from patients with further windows of opportunity might be identifiable during
recent-onset T1DM, where it was revealed that islet cell prolif- the different phases when therapeutic intervention might be at
eration was increased by as much as 10-fold above that meas- its most effective.
ured in controls (74,85). More importantly, the increase was
most evident in those islets which were inflamed, implying Acknowledgements
that a component associated with the inflammatory process
This work received financial support from the European
was responsible for mediating the effect. Interestingly, the pro-
Union’s Seventh Framework Programme PEVNET (FP7/2007-
liferative response was not unique to b-cells but a similar
2013) under grant agreement number 261441. Additional sup-
increase was also noted in a-cells, suggesting that the endo-
port was received from a Diabetes Research Wellness Founda-
crine cells were responding to a general mitotic stimulus pres-
tion Non-Clinical Research Fellowship and, since 2014, a JDRF
ent within the islet milieu. It is reasonable to propose that this
Career Development Award (5-CDA-2014-221-A-N) to S.J.R.
stimulus might have been elaborated from one or more of the
The research was also performed with the support of the Net-
immune cell subtypes present in the inflammatory infiltrate;
work for Pancreatic Organ Donors with Diabetes (nPOD), a col-
however, this factor has not yet been formally identified or
laborative type 1 diabetes research project sponsored by the
characterised. In support of this conclusion, the rate of endo-
Juvenile Diabetes Research Foundation International (JDRF)
crine cell proliferation has also been examined in the inflamed
and with a JDRF research grant awarded to the nPOD-V con-
islets of two patients who died without a diagnosis of T1DM
sortium. Organ Procurement Organizations (OPO) partnering
but who were immunopositive for multiple islet autoantibodies
with nPOD to provide research resources are listed at
(86), suggesting that they might have been in a ‘pre-diabetic’
www.jdrfnpod.org/our-partners.php.
state. The pancreases of each of these individuals contained
islets which displayed enhanced rates of endocrine cell repli-
cation and, again, this correlated with the presence of inflam- References
mation (86). [1] Atkinson, M. A. (2012) The pathogenesis and natural history of type 1 diabe-
Clearly, it is of importance to identify the factors responsi- tes. Cold Spring Harbor Perspect. Med. 2, pii: a007641.
ble for mediating islet cell proliferation during inflammation. [2] Boitard, C. (2012) Pancreatic islet autoimmunity. Presse Med. 41, e636–e650.
Once identified, this might offer a novel therapeutic option to [3] La Torre, D. (2012) Immunobiology of b-cell destruction. Adv. Exp. Med. Biol.
771, 194–218.
increase endogenous b-cell mass in patients with T1DM if the
[4] Richardson, S. J., Morgan, N. G., and Foulis, A. K. (2014) Pancreatic pathol-
relevant factors could be administered in combination with ogy in type 1 diabetes mellitus. Endocr. Pathol. 25, 80–92.
approaches that limit the autoimmune attack. [5] Roep, B. O., and Tree, T. I. (2014) Immune modulation in humans: implica-
tions for type 1 diabetes mellitus. Nat. Rev. Endocrinol. 10, 229–242.
[6] Pugliese, A. (2014) Advances in the etiology and mechanisms of type 1 dia-
Conclusion betes. Discov. Med. 18, 141–150.

Despite the dearth of human samples in which the process of [7] Eisenbarth, G. S. (1986) Type I diabetes mellitus. A chronic autoimmune dis-
ease. N. Engl. J. Med. 314, 1360–1368.
islet inflammation can be studied, important advances have
[8] Di Gialleonardo, V., de Vries, E. F., Di Girolamo, M., Quintero, A. M., Dierckx,
been made which have improved our understanding of the R. A., et al. (2012) Imaging of b-cell mass and insulitis in insulin-dependent
underlying aetiology of human T1DM. These provide clear (type 1) diabetes mellitus. Endocr. Rev. 33, 892–919.
cause for continued optimism that the illness will yield its [9] Foulis, A. K., and Stewart, J. A. (1984) The pancreas in recent-onset type 1
secrets and that new and improved therapeutic options will (insulin-dependent) diabetes mellitus: insulin content of islets, insulitis and
emerge. Arguably, these are most likely to lead to a future in associated changes in the exocrine acinar tissue. Diabetologia 26, 456–461.
[10] Gepts, W. (1965) Pathologic anatomy of the pancreas in juvenile diabetes
which T1DM is preventable in susceptible individuals prior to
mellitus. Diabetes 14, 619–633.
disease onset rather than curable in those who already have [11] Kloppel, G., Lohr, M., Habich, K., Oberholzer, M., and Heitz, P. U. (1985) Islet
the disease. Either way, it is important that further studies are pathology and the pathogenesis of type 1 and type 2 diabetes mellitus
undertaken to address the remaining areas of uncertainty. revisited. Survey Synth. Pathol. Res. 4, 110–125.
As a final statement we also note that in most analyses, it [12] Campbell-Thompson, M., Wasserfall, C., Kaddis, J., Albanese-O’Neill, A.,
has been assumed that, once initiated, T1DM progresses in a Staeva, T., et al. (2012) Network for Pancreatic Organ Donors with Diabetes
(nPOD): developing a tissue biobank for type 1 diabetes. Diabetes Metab.
broadly uniform manner until b-cell destruction reaches a crit-
Res. Rev. 28, 608–617.
ical point at which the clinical diagnosis is made. However, we [13] Dotta, F., Censini, S., van Halteren, A. G., Marselli, L., Masini, M., et al.
have emphasised that the pathology reveals an asynchronous (2007) Coxsackie B4 virus infection of b cells and natural killer cell insulitis
progression at the islet level. This then raises the alternative in recent-onset type 1 diabetic patients. Proc. Natl. Acad. Sci. USA 104,
possibility that islet inflammation might wax and wane over 5115–5120.
time in a manner which is associated with alternating periods [14] Walker, J. N., Johnson, P. R., Shigeto, M., Hughes, S. J., Clark, A., et al.
(2011) Glucose-responsive b cells in islets isolated from a patient with long-
of relapse and remission as the illness proceeds. Such a mech-
standing type 1 diabetes mellitus. Diabetologia 54, 200–202.
anism would be consistent with the situation in various other [15] Itoh, N., Hanafusa, T., Miyazaki, A., Miyagawa, J., Yamagata, K., et al. (1993)
autoimmune conditions. At present, this idea remains only an Mononuclear cell infiltration and its relation to the expression of major his-
interesting hypothesis, but if it proves to have validity, then tocompatibility complex antigens and adhesion molecules in pancreas

732 Process of Islet Inflammation in Human T1DM


biopsy specimens from newly diagnosed insulin-dependent diabetes melli- [39] Marhfour, I., Lopez, X. M., Lefkaditis, D., Salmon, I., Allagnat, F., et al. (2012)
tus patients. J. Clin. Invest. 92, 2313–2322. Expression of endoplasmic reticulum stress markers in the islets of patients
[16] Krogvold, L., Edwin, B., Buanes, T., Ludvigsson, J., Korsgren, O., et al. with type 1 diabetes. Diabetologia 55, 2417–2420.
(2014) Pancreatic biopsy by minimal tail resection in live adult patients at [40] Meier, J. J., Bhushan, A., Butler, A. E., Rizza, R. A., and Butler, P. C. (2005)
the onset of type 1 diabetes: experiences from the DiViD study. Diabetologia Sustained b cell apoptosis in patients with long-standing type 1 diabetes:
57, 841–843. indirect evidence for islet regeneration? Diabetologia 48, 2221–2228.
[17] In’t Veld, P. (2011) Insulitis in human type 1 diabetes: the quest for an elu- [41] Graham, K. L., Sutherland, R. M., Mannering, S. I., Zhao, Y., Chee, J., et al.
sive lesion. Islets 3, 131–138. (2012) Pathogenic mechanisms in type 1 diabetes: the islet is both target
[18] In’t Veld, P. (2014) Insulitis in human type 1 diabetes: a comparison and driver of disease. Rev. Diabet. Stud. 9, 148–168.
between patients and animal models. Semin. Immunopathol. 36, 569–579. [42] Hahn, S., Gehri, R., and Erb, P. (1995) Mechanism and biological signifi-
[19] Lecompte, P. M. (1958) Insulitis in early juvenile diabetes. Arch. Pathol. 66, cance of CD4-mediated cytotoxicity. Immunol. Rev. 146, 57–79.
450–457. [43] Kreuwel, H. T., Morgan, D. J., Krahl, T., Ko, A., Sarvetnick, N., et al. (1999)
[20] Schmidt, M. B. (1902) Ueber die beziechung der langerhans’schen inseln Comparing the relative role of perforin/granzyme versus Fas/Fas ligand
des pankreas zum diabetes mellitus. Munch. Med. Wochenschr. 49, 51–54. cytotoxic pathways in CD81 T cell-mediated insulin-dependent diabetes
[21] Von Meyenburg, M. (1940) Ueber “Insulitis” bei diabetes. Schweiz. Med. mellitus. J. Immunol. 163, 4335–4341.
Wochenschr. 21, 554–557. [44] Thomas, H. E., Trapani, J. A., and Kay, T. W. (2010) The role of perforin and
[22] Kloppel, G., Drenck, C. R., Carstensen, A., and Heitz, P. U. (1984) The B cell granzymes in diabetes. Cell Death Differ. 17, 577–585.
mass at the clinical onset of type I diabetes. Behring Inst. Mitt. 75, 42–49. [45] Jaberi-Douraki, M., Liu, S. W., Pietropaolo, M., and Khadra, A. (2014)
[23] Campbell-Thompson, M. L., Atkinson, M. A., Butler, A. E., Chapman, N. M., Autoimmune responses in T1DM: quantitative methods to understand
Frisk, G., et al. (2013) The diagnosis of insulitis in human type 1 diabetes. onset, progression, and prevention of disease. Pediatr. Diabetes 15,
Diabetologia 56, 2541–2543. 162–174.
[24] Dor, Y., and Glaser, B. (2013) b-Cell dedifferentiation and type 2 diabetes. N. [46] Lernmark, A., and Larsson, H. E. (2013) Immune therapy in type 1 diabetes
Engl. J. Med. 368, 572–573. mellitus. Nat. Rev. Endocrinol. 9, 92–103.
[25] Talchai, C., Xuan, S., Lin, H. V., Sussel, L., and Accili, D. (2012) Pancreatic b [47] Pietropaolo, M., Towns, R., and Eisenbarth, G. S. (2012) Humoral autoim-
cell dedifferentiation as a mechanism of diabetic b cell failure. Cell 150, munity in type 1 diabetes: prediction, significance, and detection of distinct
1223–1234. disease subtypes. Cold Spring Harbor Perspect. Med. 2, pii:a012831.
[26] White, M. G., Marshall, H. L., Rigby, R., Huang, G. C., Amer, A., et al. (2013) [48] Christie, M. R., Hollands, J. A., Brown, T. J., Michelsen, B. K., and Delovitch,
Expression of mesenchymal and a-cell phenotypic markers in islet b-cells in T. L. (1993) Detection of pancreatic islet 64,000 M(r) autoantigens in insulin-
recently diagnosed diabetes. Diabetes Care 36, 3818–3820. dependent diabetes distinct from glutamate decarboxylase. J. Clin. Invest.
[27] Anuradha, R., Saraswati, M., Kumar, K. G., and Rani, S. H. (2014) Apoptosis 92, 240–248.
of b cells in diabetes mellitus. DNA Cell Biol. 33, 743–748. [49] Christie, M. R., Brown, T. J., and Cassidy, D. (1992) Binding of antibodies in
[28] Quan, W., Jo, E. K., and Lee, M. S. (2013) Role of pancreatic b-cell death sera from type 1 (insulin-dependent) diabetic patients to glutamate decar-
and inflammation in diabetes. Diabetes Obes. Metab. 15 (Suppl3), 141–151. boxylase from rat tissues. Evidence for antigenic and non-antigenic forms
[29] Santin, I., and Eizirik, D. L. (2013) Candidate genes for type 1 diabetes mod- of the enzyme. Diabetologia 35, 380–384.
ulate pancreatic islet inflammation and b-cell apoptosis. Diabetes Obes. [50] Bonifacio, E., Krumsiek, J., Winkler, C., Theis, F. J., and Ziegler, A. G. (2014)
Metab. 15 (Suppl 3), 71–81. A strategy to find gene combinations that identify children who progress
[30] Richardson, S. J., Willcox, A., Bone, A. J., Foulis, A. K., and Morgan, N. G. rapidly to type 1 diabetes after islet autoantibody seroconversion. Acta Dia-
(2009) Islet-associated macrophages in type 2 diabetes. Diabetologia 52, betol. 51, 403–411.
1686–1688. [51] Nokoff, N., and Rewers, M. (2013) Pathogenesis of type 1 diabetes: lessons
[31] Delaney, C. A., Pavlovic, D., Hoorens, A., Pipeleers, D. G., and Eizirik, D. L. from natural history studies of high-risk individuals. Ann. NY Acad. Sci.
(1997) Cytokines induce deoxyribonucleic acid strand breaks and apoptosis 1281, 1–15.
in human pancreatic islet cells. Endocrinology 138, 2610–2614. [52] Bottazzo, G. F., Dean, B. M., McNally, J. M., MacKay, E. H., Swift, P. G.,
[32] Dunger, A., Augstein, P., Schmidt, S., and Fischer, U. (1996) Identification of et al. (1985) In situ characterization of autoimmune phenomena and expres-
interleukin 1-induced apoptosis in rat islets using in situ specific labelling of sion of HLA molecules in the pancreas in diabetic insulitis. N. Engl. J. Med.
fragmented DNA. J. Autoimmun. 9, 309–313. 313, 353–360.
[33] Eizirik, D. L., and Mandrup-Poulsen, T. (2001) A choice of death—the signal- [53] Willcox, A., Richardson, S. J., Bone, A. J., Foulis, A. K., and Morgan, N. G.
transduction of immune-mediated b-cell apoptosis. Diabetologia 44, 2115– (2009) Analysis of islet inflammation in human type 1 diabetes. Clin. Exp.
2133. Immunol. 155, 173–181.
[34] Hadjivassiliou, V., Green, M. H., James, R. F., Swift, S. M., Clayton, H. A., [54] Rodriguez-Calvo, T., Ekwall, O., Amirian, N., Zapardiel-Gonzalo, J., and von
et al. (1998) Insulin secretion, DNA damage, and apoptosis in human and Herrath, M. G. (2014) Increased immune cell infiltration of the exocrine pan-
rat islets of Langerhans following exposure to nitric oxide, peroxynitrite, creas: a possible contribution to the pathogenesis of type 1 diabetes. Diabe-
and cytokines. Nitric Oxide 2, 429–441. tes 63, 3880–3890.
[35] Loweth, A. C., Williams, G. T., James, R. F., Scarpello, J. H., and Morgan, N. [55] Valle, A., Giamporcaro, G. M., Scavini, M., Stabilini, A., Grogan, P., et al.
G. (1998) Human islets of Langerhans express Fas ligand and undergo (2013) Reduction of circulating neutrophils precedes and accompanies type
apoptosis in response to interleukin-1b and Fas ligation. Diabetes 47, 1 diabetes. Diabetes 62, 2072–2077.
727–732. [56] In’t Veld, P., De Munck, N., Van Belle, K., Buelens, N., Ling, Z., et al. (2010)
[36] Signore, A., Annovazzi, A., Gradini, R., Liddi, R., and Ruberti, G. (1998) Fas b-Cell replication is increased in donor organs from young patients after
and Fas ligand-mediated apoptosis and its role in autoimmune diabetes. prolonged life support. Diabetes 59, 1702–1708.
Diabetes Metab. Rev. 14, 197–206. [57] Arif, S., Leete, P., Nguyen, V., Marks, K., Nor, N. M., et al. (2014) Blood and
[37] Stassi, G., Todaro, M., Richiusa, P., Giordano, M., Mattina, A., et al. (1995) islet phenotypes indicate immunological heterogeneity in type 1 diabetes.
Expression of apoptosis-inducing CD95 (Fas/Apo-1) on human b-cells sorted Diabetes 63, 3835–3845.
by flow-cytometry and cultured in vitro. Transplant. Proc. 27, 3271–3275. [58] Pescovitz, M. D., Greenbaum, C. J., Bundy, B., Becker, D. J., Gitelman, S. E.,
[38] Yamada, K., Takane-Gyotoku, N., Yuan, X., Ichikawa, F., Inada, C., et al. et al.; and Type 1 Diabetes TrialNet Anti-CD20 Study Group. (2014) B-lym-
(1996) Mouse islet cell lysis mediated by interleukin-1-induced Fas. Diabeto- phocyte depletion with rituximab and b-cell function: two-year results. Dia-
logia 39, 1306–1312. betes Care 37, 453–459.

Morgan et al. 733


IUBMB LIFE

[59] Pescovitz, M. D., Greenbaum, C. J., Krause-Steinrauf, H., Becker, D. J., [73] Richardson, S. J., Willcox, A., Bone, A. J., Foulis, A. K., and Morgan, N. G.
Gitelman, S. E., et al., and Type 1 Diabetes TrialNet Anti-CD20 Study Group. (2009) The prevalence of enteroviral capsid protein vp1 immunostaining
(2009) Rituximab, B-lymphocyte depletion, and preservation of b-cell func- in pancreatic islets in human type 1 diabetes. Diabetologia 52, 1143–1151.
tion. N. Engl. J. Med. 361, 2143–2152. [74] Willcox, A., Richardson, S. J., Bone, A. J., Foulis, A. K., and Morgan, N. G.
[60] Bataille, R., Jego, G., Robillard, N., Barille-Nion, S., Harousseau, J. L., et al. (2011) Immunohistochemical analysis of the relationship between islet cell
(2006) The phenotype of normal, reactive and malignant plasma cells. Iden- proliferation and the production of the enteroviral capsid protein, VP1, in
tification of “many and multiple myelomas” and of new targets for the islets of patients with recent-onset type 1 diabetes. Diabetologia 54,
myeloma therapy. Haematologica 91, 1234–1240. 2417–2420.
[61] Hinman, R. M., and Cambier, J. C. (2014) Role of B lymphocytes in the [75] Bollyky, P. L., Bogdani, M., Bollyky, J. B., Hull, R. L., and Wight, T. N. (2012)
pathogenesis of type 1 diabetes. Curr. Diabetes Rep. 14, 543. The role of hyaluronan and the extracellular matrix in islet inflammation
[62] Coppieters, K. T., Dotta, F., Amirian, N., Campbell, P. D., Kay, T. W., et al. and immune regulation. Curr. Diabetes Rep. 12, 471–480.
(2012) Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from [76] Otonkoski, T., Banerjee, M., Korsgren, O., Thornell, L. E., and Virtanen, I.
recent onset and long-term type 1 diabetes patients. J. Exp. Med. 209, 51–60. (2008) Unique basement membrane structure of human pancreatic islets:
[63] Pathiraja, V., Kuehlich, J. P., Campbell, P. D., Krishnamurthy, B., Loudovaris, implications for b-cell growth and differentiation. Diabetes Obes. Metab. 10
T., et al. Proinsulin specific, HLA-DQ8 and HLA-DQ8 transdimer restricted, (Suppl 4), 119–127.
CD41 T cells infiltrate the islets in type 1 diabetes. Diabetes, in press. [77] van Deijnen, J. H., Hulstaert, C. E., Wolters, G. H., and van Schilfgaarde, R.
[64] Berg, A. K., Korsgren, O., and Frisk, G. (2006) Induction of the chemokine (1992) Significance of the peri-insular extracellular matrix for islet
interferon-c-inducible protein-10 in human pancreatic islets during enterovi- isolation from the pancreas of rat, dog, pig, and man. Cell Tissue Res. 267,
rus infection. Diabetologia 49, 2697–2703. 139–146.
[65] Cardozo, A. K., Proost, P., Gysemans, C., Chen, M. C., Mathieu, C., et al. [78] Wang, R. N., and Rosenberg, L. (1999) Maintenance of b-cell function and
(2003) IL-1b and IFN-c induce the expression of diverse chemokines and IL- survival following islet isolation requires re-establishment of the islet–matrix
15 in human and rat pancreatic islet cells, and in islets from pre-diabetic relationship. J. Endocrinol. 163, 181–190.
NOD mice. Diabetologia 46, 255–266. [79] Korpos, E., Kadri, N., Kappelhoff, R., Wegner, J., Overall, C. M., et al. (2013)
[66] Chen, M. C., Proost, P., Gysemans, C., Mathieu, C., and Eizirik, D. L. (2001) The peri-islet basement membrane, a barrier to infiltrating leukocytes in
Monocyte chemoattractant protein-1 is expressed in pancreatic islets from type 1 diabetes in mouse and human. Diabetes 62, 531–542.
prediabetic NOD mice and in interleukin-1b-exposed human and rat islet [80] Irving-Rodgers, H. F., Choong, F. J., Hummitzsch, K., Parish, C. R., Rodgers,
cells. Diabetologia 44, 325–332. R. J., et al. (2014) Pancreatic islet basement membrane loss and remodeling
[67] Grewal, I. S., Rutledge, B. J., Fiorillo, J. A., Gu, L., Gladue, R. P., et al. (1997) after mouse islet isolation and transplantation: impact for allograft rejection.
Transgenic monocyte chemoattractant protein-1 (MCP-1) in pancreatic islets Cell Transplant. 23, 59–72.
produces monocyte-rich insulitis without diabetes: abrogation by a second [81] Conget, I., Fernandez-Alvarez, J., Ferrer, J., Sarri, Y., Novials, A., et al.
transgene expressing systemic MCP-1. J. Immunol. 159, 401–408. (1993) Human pancreatic islet function at the onset of type 1 (insulin-
[68] Roep, B. O., Kleijwegt, F. S., van Halteren, A. G., Bonato, V., Boggi, U., et al. dependent) diabetes mellitus. Diabetologia 36, 358–360.
(2010) Islet inflammation and CXCL10 in recent-onset type 1 diabetes. Clin. [82] Marchetti, P., Dotta, F., Ling, Z., Lupi, R., Del Guerra, S., et al. (2000) Func-
Exp. Immunol. 159, 338–343. tion of pancreatic islets isolated from a type 1 diabetic patient. Diabetes
[69] Foulis, A. K., Farquharson, M. A., and Meager, A. (1987) Immunoreactive a- Care 23, 701–703.
interferon in insulin-secreting b cells in type 1 diabetes mellitus. Lancet 2, [83] Butler, P. C., Meier, J. J., Butler, A. E., and Bhushan, A. (2007) The replica-
1423–1427. tion of b cells in normal physiology, in disease and for therapy. Nat. Clin.
[70] Harrison, L. C., Campbell, I. L., Allison, J., and Miller, J. F. (1989) MHC mole- Pract. Endocrinol. Metab. 3, 758–768.
cules and b-cell destruction. Immune and nonimmune mechanisms. Diabe- [84] Meier, J. J., Lin, J. C., Butler, A. E., Galasso, R., Martinez, D. S., et al. (2006)
tes 38, 815–818. Direct evidence of attempted b cell regeneration in an 89-year-old patient
[71] Morgan, N. G., and Richardson, S. J. Enteroviruses as causative agents in with recent-onset type 1 diabetes. Diabetologia 49, 1838–1844.
type 1 diabetes: loose ends or lost cause? Trends Endocrinol. Metab. 25, 611– [85] Willcox, A., Richardson, S. J., Bone, A. J., Foulis, A. K., and Morgan, N. G.
619. (2010) Evidence of increased islet cell proliferation in patients with recent-
[72] Richardson, S. J., Leete, P., Bone, A. J., Foulis, A. K., and Morgan, N. G. onset type 1 diabetes. Diabetologia 53, 2020–2028.
(2013) Expression of the enteroviral capsid protein VP1 in the islet cells of [86] In’t Veld, P., Lievens, D., De Grijse, J., Ling, Z., Van der Auwera, B., et al.
patients with type 1 diabetes is associated with induction of protein kinase (2007) Screening for insulitis in adult autoantibody-positive organ donors.
R and downregulation of Mcl-1. Diabetologia 56, 185–193. Diabetes 56, 2400–2404.

734 Process of Islet Inflammation in Human T1DM

You might also like