Enteroviruses as causative agents in type 1 diabetes loose ends or

advertisement
Enteroviruses as causative agents in type 1 diabetes: loose ends or lost cause?
Noel G Morgan & Sarah J Richardson
Institute of Biomedical & Clinical Science
University of Exeter Medical School
RILD Building, Barrack Road
Exeter EX2 5DW, UK
Correspondence:
n.g.morgan@exeter.ac.uk (+44-(0)1392-408300)
s.richardson@exter.ac.uk (+44-(0)1392-408225)
Keywords
Coxsackievirus; type 1 diabetes; persistent infection; pathogen recognition receptors; islets of
Langerhans
1
Abstract
Considerable evidence implies that an enteroviral infection may accelerate or precipitate type 1
diabetes (T1D) in some individuals. However, causality is not proven. We present and critically
assess evidence suggesting that islet β-cells can become infected with enterovirus, and argue that
this may result in one of several consequences. Occasionally, a fully lytic infection may arise and
this culminates in fulminant diabetes. Alternatively, an atypical persistent infection develops
which can be either benign or promote islet autoimmunity. We propose a model in which the
“strength” of the β-cell response to the establishment of a persistent enteroviral infection
determines the final disease outcome.
2
Linking enteroviruses to T1D
Enteroviruses are a genus of positive-sense, single stranded, non-enveloped, RNA viruses that
circulate widely and can cause a spectrum of mild respiratory and gastrointestinal (GI) symptoms
as well as more severe conditions such as myocarditis or poliomyelitis. In addition, some
enteroviruses, notably those belonging to the Coxsackievirus B species, have been implicated in
the development of T1D in humans, implying that an initial (mild) infection might have significant,
long-term, consequences in some individuals.
The cumulative evidence implying a firm association between enteroviral infection and the
development of T1D in humans is substantial but, unlike the situation in mouse models [1], this
evidence base remains largely correlative. Unequivocal confirmation of enteroviral infection
within the pancreatic islets has been hard to obtain and the majority of data have been derived
from epidemiological studies which show that markers of enteroviral infection occur commonly in
the circulation of patients at diagnosis of T1D [2-11]. One pertinent recent example comes from a
study of Italian families where enteroviral genome was detected at high frequency in the blood of
newly diagnosed T1D patients (79%) as well as in their parents (63%) and siblings (60%) but in only
1 of 29 (3%) relevant controls [8]. These data imply very firmly that enteroviral infections may
contribute to the onset of disease in some patients but the data also show that such infections are
not the sole determinants. This is not surprising, given that enteroviruses circulate widely in the
population whereas the incidence of T1D is much lower, even in populations where it is most
prevalent.
Clearly, any model which attempts to implicate enteroviruses in the aetiology of diabetes must
account for the fact that the vast majority of people who become infected do not go on to develop
diabetes. This is illustrated forcibly by studies among Cubans who were exposed to an epidemic of
echovirus early in the 21st century [12]. Within this group, large numbers of patients
seroconverted to islet auto-antibody positivity following enteroviral infection, but the prevalence
of overt T1D remained low. Thus, not all infected individuals went on to develop disease. This
situation is reminiscent of the paralysis associated with poliomyelitis which occurs in only a
minority of infected individuals [13]. One contributory factor in the case of T1D may be the
enteroviral serotype involved since data from the Finnish population implies that serotypes which
are closely related phylogenetically, have a differing potential to promote T1D [3, 6].
3
Autoimmunity and enteroviruses; what are the connections?
Is there a role for molecular mimicry?
The currently accepted wisdom considers that human T1D is mediated by a process of
autoimmunity in which pancreatic β-cells are destroyed in a highly selective manner [14].
Therefore, any attempt to relate autoimmunity and β-cell loss to enteroviral infection must
provide a plausible mechanism by which these processes are linked. One of the most obvious
possibilities for the development of autoimmunity is that some form of molecular mimicry occurs
in which the immune response to a viral antigen triggers the generation of antibodies which
serendipitously cross-react with sequences present in islet proteins. Support for this concept has
been presented since a conserved sequence within the enterovirus 2C protein is also present in
glutamate decarboxylase (GAD); a principal autoantigen in T1D [15, 16]. However, while this
epitope relationship may be true, it is not clear why this should serve as the trigger for islet
autoimmunity. Moreover, it is likely that the presence of circulating autoantibodies directed
against GAD is a marker of disease progression rather than of pathogenic significance. Thus, the
association between enteroviral peptides and islet autoantibody generation has been questioned
[17-21]. We conclude that, although the molecular mimicry mechanism has an attractive
symmetry, this is superficial and does not provide a firm basis to explain the mechanisms of β-cell
loss.
Migration of enteroviruses to islets
A second possibility is that enteroviruses may spread from their initial sites of infection to β-cells
and thereby provoke an (auto)immune response more directly. There is firm evidence that
enteroviruses can establish an infection in cells of the GI tract in patients with T1D [22], and this is
likely to be the source of islet infection. Supporting evidence comes from reports of the isolation
of enterovirus from the pancreas of two patients with recent onset T1D [23, 24] (although, in one
of these, the provenance of the virus has been challenged [1]).
It is also clear that other pathways exist by which an infection may spread to more remote organs,
since it is well understood that acute enteroviral infection can lead to myocarditis under certain
circumstances [25-27]. It is likely that, in such cases, a blood-borne infection ensues and, in
4
support of this, evidence of early viraemia during acute enteroviral infection has been provided
[28]. This might, then, lead to the spread of infectious particles to any internal organ that is
permissive for the virus. To address this possibility, pancreases recovered from several children
who had died of acute myocarditis following proven enteroviral infection were examined and the
islets found to contain enteroviral antigens [25, 29] and RNA [30, 31]. This provides very strong
evidence that enteroviruses can spread from the circulation to the pancreatic islets, at least under
conditions when the viral load is extremely high.
It is a moot point whether such transit also occurs during episodes of mild enteroviral infection but
the establishment of productive infection in the pancreatic islets appears to be uncommon.
Among a cohort of British children who died of causes unrelated to enteroviral infection (or T1D)
only 6% (3 of 50) had evidence of enteroviral infection in their islet cells (as determined by
immunostaining for the enteroviral capsid protein VP1). Moreover, in those who did, the number
of positively stained cells was vanishingly low. This contrasts with data obtained from the
pancreases of children who died soon after developing T1D, where VP1 was detected in the islets
of more than 60% (44 of 72) [29] and, in a subset of these same cases, the presence of enterovirus
was confirmed by in situ hybridisation [30]. Additional smaller studies have reached similar
conclusions [30, 32, 33]. Such evidence implies that enteroviral infection is intimately related to
diabetes; and might be causative. Alternatively, it is also possible that the islets of patients who
have contracted T1D are simply more susceptible to enteroviral infection; perhaps because of
ongoing damage to the β-cells. We do not favour this latter hypothesis since histological analysis
of human pancreas suggests that the majority (>60%) of insulin-containing, VP1-immunopositive,
islets are not insulitic [34], implying that viral infection precedes the process of immune cell
recruitment and β-cell damage. Moreover, the fact that seroconversion to islet autoantibody
positivity occurs in many patients (including monozygotic siblings [35]) who have contracted an
enteroviral infection but not developed T1D (at least, at the time of analysis) [12, 35] implies that
infection of the islet cells must be an early event in the process. Nevertheless, gaining firm
information about the pathological changes occurring within the pancreas during the initiation of
T1D remains difficult and the continuing dearth of appropriate samples for study represents a
significant impediment to progress.
5
Islet cell susceptibility to infection differs between cell types
That islet cells can become infected with enteroviruses under some circumstances is not in dispute
but begs a further important question; namely, which of the various endocrine cell types support
enteroviral replication in vivo? The majority of data indicate a clear selectivity for β-cells (as
judged by the cellular distribution of immunoreactive VP1) [29, 32, 33] and this is consistent with
the selectivity of cell loss seen in the disease. In this context, it is also important to note that the
exocrine pancreas is largely immunonegative for VP1 in most human T1D patients, although
occasional VP1 immunopositivity occurs in cells of the duct epithelium [29, 30]. This situation is
quite different from that seen in mouse models where enteroviral infection of the acinar tissue
within the exocrine pancreas is often widespread, suggesting that there are very different
mechanisms regulating pancreatic infectivity and replication in rodents and humans [36-38].
To clarify the infectivity of islet cells, recent studies have addressed the expression of two
potential enteroviral "receptor" proteins, Decay Accelerating Factor (DAF) and CoxsackieAdenovirus Receptor (CAR), in human pancreas. CAR is considered to be a tight-junctional
component, whereas the physiological role of DAF is less clear and it is reportedly absent from
pancreatic islets [30]. By contrast, CAR is expressed in islet cells [39, 40], and antibodies against
CAR can block enteroviral infection of islets incubated in vitro [30]. However, CAR exists in
multiple isoforms and the precise isoform distribution has not been mapped in islet cells; nor is it
clear exactly which isoforms preferentially mediate enteroviral entry [41]. Some workers have
found that CAR is present in the majority of human islet cells [42] whereas others, using different
antibodies, report that it is present in β- but not alpha-cells within islets [39]. Interestingly, in the
latter study, CAR was detectable on a unique sub-population of human alpha cells located outside
the islets and within the pancreatic parenchyma [39]. The origin and ultimate destination of these
"extra-islet" alpha cells is unclear and it is not known why they should express CAR. However, one
might speculate that the protein is present to assist in their coalescence into primordial islets, by
promoting tight junction formation with other, single, endocrine cells that are encountered within
the pancreas. Whatever the explanation, it has not yet been demonstrated that such CAR-positive
alpha cells can become infected with enterovirus in vivo, and the weight of evidence still points to
a primarily β-cell focussed infection mechanism.
6
Non-canonical viral replication
Importantly, there are aspects of the biology of enteroviruses which appear unusual when they
infect β-cells in vivo. Most notable is the fact that morphological examination of the islets of
patients with T1D does not reveal evidence of large-scale cell lysis. This contrasts with expectation
since enteroviruses normally subvert the cell's transcriptional and translational machinery to
generate newly synthesised capsids which are then released by a lytic mechanism [43]. These new
virions are able to infect neighbouring cells, leading to extensive tissue damage; but such damage
is not typically seen in human T1D. Despite this, it cannot be excluded that lysis of a small number
of islet cells may occur early in the disease process since any debris will have been cleared by the
time tissue is removed for analysis (which may be weeks, months or even years after disease
initiation). An equivalent situation has been described in the heart where an early episode of
cardiomyocyte lysis is followed by the establishment of a more sustained, non-lytic, infection [44].
The fact that extensive cell lysis is not seen in the pancreas of patients with T1D is unlikely to be
simply a property of the cells themselves since infection of isolated human islets maintained in
tissue culture, can readily lead to extensive damage [45-47]. Moreover, in a rare form of T1D that
has been described most frequently in Japanese populations (known as "fulminant" diabetes),
large-scale lysis of islet cells does occur in vivo, and this disease may well be mediated by an
acutely lytic enteroviral infection of the islet cells [48-50]. If so, it differs markedly from the
situation seen more typically in the islets of patients with T1D in Western populations.
Interestingly, in fulminant diabetes the destructive process is so intense that both β- and alpha cell
lysis occurs [51, 52]. This might imply that each cell type can support enteroviral infection in those
ethnic groups who are most susceptible to fulminant diabetes but this remains to be clarified. A
further possibility is that the net damage to β-cells is so intense in fulminant disease that adjacent
"bystander" cells undergo a rapid demise as a result of the large-scale release of β-cell hydrolases
within the islet milieu (Figure 1).
Given that dramatic cell lysis does not normally occur during β-cell enteroviral infection in more
typical (i.e. non-fulminant) T1D, this implies that the viral replication cycle cannot proceed
according to a canonical mechanism. Any such conclusion may seem radical but there is already
evidence that the biology of enteroviruses is altered when they infect the heart. In particular,
Chapman and colleagues have shown that infection of human cardiomyocytes with enteroviruses,
7
in vivo, results in the loss of genomic sequences from the 5' end of the RNA [44, 53, 54]. Such
"terminally deleted" viruses have much reduced propensity to replicate and they rarely form
functionally competent virions that can be released to infect neighbouring cells. As such, they
persist in the host cells in a form which replicates only very slowly over a prolonged period. This
scenario has clear parallels with the situation found in the islets of patients with T1D, where only
occasional VP1 immunopositive β-cells can be detected and wholesale cell lysis is not apparent.
The presence of slowly replicating (genomically modified?) virus in a tiny proportion of human islet
cells may also contribute to the difficulties encountered in detecting viral RNA by RT-PCR in
pancreatic extracts, by comparison with isolated islets acutely infected in vitro, where abundant
quantities of fully replicative virus are present in each infected cell [55].
Defective trans-differentiation of infected pancreatic duct cells
Additional data emerging from studies of persistent enteroviral infection in cultured cell models
has also pointed to another important issue, which might shed further light on the processes
leading to net β-cell loss in human T1D. Hober and colleagues have established a persistent
enteroviral infection in a line of pancreatic ductal cells in tissue culture, and reported that this
leads to a reduced propensity for these cells to undergo differentiation [56]. This is important since
one mechanism by which β-cells might be replenished in humans occurs by a process of transdifferentiation of pancreatic duct cells [56, 57]. Thus, if the establishment of a persistent
enteroviral infection in the duct cells impedes this process, then β-cell replacement might be
compromised, further contributing to their overall demise under conditions when they are being
depleted by autoimmune-mediated mechanisms. This is important since, as emphasised earlier,
VP1 immunopositivity is sometimes detected in pancreatic ducts in patients with T1D, which
would be consistent with this model.
Alternative mechanisms of persistent enteroviral infection
The mechanism by which enteroviral persistence might be maintained within islet cells remains an
important issue and there are reports that enteroviruses can exist in an atypical double-stranded
RNA genomic form as a means to promote persistence [58-60] (Figure 1). This may seem odd,
given that the genome is normally a molecule of positive single stranded RNA and the fact that
dsRNA is not usually tolerated in cells. Normally, the presence of dsRNA is carefully monitored by
8
various cellular surveillance systems which are designed to respond to its appearance by mounting
a vigorous anti-viral response [27]. Human islet cells are equipped with several intracellular
pattern recognition receptors [39, 47, 49, 61-63] (Figure 1), some of which are activated
exclusively by dsRNA and respond by initiating signalling cascades that promote type-I and type-III
interferon generation [64]. Frequently, the presence of longer length (i.e. enteroviral) dsRNA is
sensed most effectively by an RNA helicase enzyme, Mda5 [65] and this protein is present in
human islets. However, in islets, Mda5 is not expressed uniformly among the different endocrine
cells. Rather, it appears to be most abundant in alpha cells while β-cells are relatively deficient [39,
63, 66]. Thus, conceivably, the establishment of a persistent β-cell infection might be favoured by
this relative deficiency. Interestingly, Mda5 expression is increased in the residual β-cells of
patients with T1D [39, 66] which could be indicative of an enhanced response to viral infection
(Figure 1). This suggests, in turn, that the normally low activity of Mda5 in β-cells may be
important as a means to mitigate the development of autoimmunity.
Level of response to infection might determine final outcome
Following on from the evidence outlined above, it seems plausible that the magnitude of the antiviral response occurring at the level of β-cells during an enteroviral infection, could be critical to
determination of the final outcome. In this model, the better outcome arises when β-cells fail to
respond vigorously to enteroviral infection since, under these circumstances, they can continue to
survive and function. In support of this, it was shown that islet cells in 15-20% of adults without
diabetes, express VP1 (albeit at very low frequency) [29, 32]. This suggests that, if the cells allow
the virus to persist (perhaps in a slowly replicating form) without developing a robust response,
then a new equilibrium is established in which both the virus and the infected β-cell can co-exist.
If, however, this equilibrium is disturbed, such that the cell strenuously attempts to eliminate the
virus (e.g. by increasing the expression and activity of Mda5), this might then, counter-intuitively,
serve as the trigger that provokes autoimmunity and the progression towards T1D. In support of
this, certain polymorphic variants of Mda5 with reduced helicase activity are known to confer a
measure of protection against the development of T1D [67-70] and we individuals carrying these
protective variants are likely to respond only weakly to β-cell enteroviral infection.
Further evidence that a heightened β-cell response to enteroviral infection might mediate
autoimmunity comes from an analysis of the expression of MHC class I antigens on the islet cells of
9
patients and controls. Islet cell hyper-expression of MHC class I is a hallmark of T1D [71-73] and is
likely to occur as a result of increased interferon secretion during enteroviral infection [74, 75].
Importantly, islet cell hyper-expression of MHC class I is not seen in control individuals, even
among those whose islets stain positively for VP1. This supports the concept that weakly (or non-)
responding subjects are less likely to develop T1D.
This hypothesis inevitably raises additional questions, including whether it is the presence of
virally infected β-cells per se which triggers autoimmunity and then frank diabetes, or whether
more subtle mechanisms are operative. So far, the answers to these questions remain elusive, but
a recent study of the specificity of islet infiltrating T-cells in patients with T1D has shown that at
least a proportion of these are directed against islet antigens [76]. It is unclear whether a different
population may also be reactive against viral antigens. Nevertheless, the fact that some infiltrating
T-cells are reactive against islet proteins implies that, if viral infection underlies these processes,
then the virus may promote altered presentation of normally "silent" islet protein epitopes as the
primary means to elicit an immune response. Alternatively, (or additionally) the characteristic islet
cell hyper-expression of MHC class I noted above, may contribute to the enhanced presentation of
typical β-cell antigens thereby provoking an autoimmune response. Whichever mechanism is
operative, the virus can be considered to act as a Trojan horse, slipping quietly inside the normal
cellular defences but then, once recognised, eliciting an anti-viral response which alters the
intracellular environment to promote autoimmunity.
The role of interferons and protein kinase R (PKR)
It follows from this discussion, that the presence of enterovirus within the β-cells of patients with
T1D should be accompanied by a signature anti-viral response. In particular, it would be
anticipated that interferons type-I and -III, should be generated locally within the islet milieu
(Figure 1). This would then lead to the production of a characteristic interferon response in the
cells surrounding those carrying the persistent infection. One feature of this would be the hyperexpression of MHC class-I [71-73] (Figure 1) and direct evidence that this correlates with
interferon-alpha secretion in the islets of patients with recent-onset T1D has been provided [74].
Furthermore, a clear interferon signature has also been observed in the islets of the non-obese
diabetic (NOD) mouse early in life, although this is apparently not required for development of
10
T1D [77]. In addition, an interferon response has also been demonstrated in the blood of human
subjects who subsequently developed islet autoantibodies [78, 79] although this may reflect
interferon production outside the islets.
Although such evidence is persuasive, it must also be accepted that there are still many gaps to be
filled before the hypothesis that an islet viral response signature can be detected commonly in
human T1D, is considered watertight. In particular, as noted above in relation to MHC class-I
hyper-expression, the release of interferons should also promote the expression of additional
"interferon-response genes" (ISGs). Searching for this in human islet cells has not proved
straightforward. Thus, while interferon responses (and generation of ISGs) can be readily
demonstrated when human islets are isolated and exposed to enteroviruses in vitro [47, 61, 80,
81], detection of such gene products in the islets of patients with T1D has proved more elusive.
For example, one of the most prominent ISGs is "ISG15", a protein which becomes covalently
attached to other newly-synthesised viral proteins in virally infected cells, thereby targeting them
for degradation [82]. Surprisingly, ISG15 is readily detected in all human islets irrespective of the
presence or absence of enteroviruses, and it is present in delta- rather than β-cells [39]. The
reasons for this are unclear and the factors that drive expression of elevated ISG15 levels in delta
cells are unknown. However, the data imply that ISG15 is not expressed in response to viral
infection in these cells and, more significantly, there is little evidence that it is induced in β-cells, in
T1D. This seems surprising if the islets are bathed in interferons as a response to local viral
infection. Conceivably, this discrepancy might be explained by variations in the production of,
and/or the precise downstream response to, individual interferon isoforms which mediate
differences in receptor signalling [83].
A further unexpected observation relates to the up-regulation of a second anti-viral response
gene, PKR. It was shown that the small number of islet cells which are immunopositive for
enteroviral VP1 in human pancreas (and which, therefore, appear to be - or to have been infected with replicating virus), also express very high levels of PKR [32]. This provides very strong
support for the concept that β-cell VP1-immunoreactivity reflects an underlying enteroviral
infection, since PKR is a member of the family of PRRs known to be induced by these viruses [81,
84, 85], and whose primary role is to signal a state of cellular stress in which protein translation is
arrested (Figure 1). This then leads to the selective degradation of certain labile anti-apoptotic
11
proteins (such as Mcl-1 [32]) within the virally-infected cells, thereby increasing their vulnerability
to pro-apoptotic stimuli. The unexpected feature of the islet PKR response is that it occurs in such
a restricted manner. Rather than finding a marked up-regulation of this enzyme in all islet cells in
the vicinity of those which express VP1 (as might be expected if the release of a soluble factor such
as an interferon was involved), the most dramatic increase occurs uniquely in that subset of β-cells
which also express VP1. Indeed, this relationship is so strong that, under many circumstances,
immunodetection of PKR can be used, with confidence, as surrogate for VP1 in human pancreas.
Thus, it must be concluded that this component of the "anti-viral" response is mediated mainly by
an intracellular mechanism operating within each single infected cell, rather than by an
intercellular (soluble) mediator which acts on all neighbouring cells. How this happens is unclear.
Concluding remarks and future perspectives
In conclusion, there is growing evidence that enteroviral infection plays a direct role in promoting
islet autoimmunity in some patients with T1D and a recent meta-analysis of the cumulative
epidemiological evidence confirms a very clear association [11]. There is also strong evidence that
β-cells can become infected in vivo and can sustain an enteroviral infection. However, whether
(and, if so, how) such infections persist is unclear. The current state of play is summarised in
Figures 1 & 2 and it is evident that many loose ends must still be tied before causality is confirmed.
Nevertheless, all is not yet lost and the prospect that an enteroviral vaccination strategy might
ultimately be effective in, at least, some individuals who are predisposed to T1D, justifies a
continued effort.
Acknowledgements
We are pleased to acknowledge financial support from the European Union's Seventh Framework
Programme PEVNET [FP7/2007-2013] under grant agreement number 261441. Additional support
was from a Diabetes Research Wellness Foundation Non-Clinical Research Fellowship and, since
2014, a JDRF Career Development Award (5-CDA-2014-221-A-N) to S.J.R. The research was also
performed with the support of the Network for Pancreatic Organ Donors with Diabetes (nPOD), a
collaborative type 1 diabetes research project sponsored by the Juvenile Diabetes Research
12
Foundation International (JDRF) and with a JDRF research grant awarded to the nPOD-V
consortium. Organ Procurement Organizations (OPO) partnering with nPOD to provide research
resources are listed at www.jdrfnpod.org/our-partners.php.
We are grateful to Mark Russell and Pia Leete for stimulating discussions and assistance with the
preparation of images.
13
References
1 Tracy, S., et al. (2010) Enteroviruses, type 1 diabetes and hygiene: a complex relationship.
Reviews in medical virology 20, 106-116
2 Craig, M.E., et al. (2003) Reduced frequency of HLA DRB1*03-DQB1*02 in children with type 1
diabetes associated with enterovirus RNA. J Infect Dis 187, 1562-1570
3 Laitinen, O.H., et al. (2014) Coxsackievirus B1 is associated with induction of beta-cell
autoimmunity that portends type 1 diabetes. Diabetes 63, 446-455
4 Moya-Suri, V., et al. (2005) Enterovirus RNA sequences in sera of schoolchildren in the general
population and their association with type 1-diabetes-associated autoantibodies. Journal of medical
microbiology 54, 879-883
5 Nairn, C., et al. (1999) Enterovirus variants in the serum of children at the onset of Type 1
diabetes mellitus. Diabet Med 16, 509-513
6 Oikarinen, S., et al. (2014) Virus antibody survey in different European populations indicates risk
association between coxsackievirus B1 and type 1 diabetes. Diabetes 63, 655-662
7 Salminen, K., et al. (2003) Enterovirus infections are associated with the induction of beta-cell
autoimmunity in a prospective birth cohort study. J Med Virol 69, 91-98
8 Salvatoni, A., et al. (2013) Intrafamilial spread of enterovirus infections at the clinical onset of
type 1 diabetes. Pediatr Diabetes 14, 407-416
9 Sarmiento, L., et al. (2007) Occurrence of enterovirus RNA in serum of children with newly
diagnosed type 1 diabetes and islet cell autoantibody-positive subjects in a population with a low
incidence of type 1 diabetes. Autoimmunity 40, 540-545
10 Schulte, B.M., et al. (2010) Detection of enterovirus RNA in peripheral blood mononuclear cells
of type 1 diabetic patients beyond the stage of acute infection. Viral Immunol 23, 99-104
11 Yeung, W.C., et al. (2011) Enterovirus infection and type 1 diabetes mellitus: systematic review
and meta-analysis of observational molecular studies. BMJ (Clinical research ed 342, d35
12 Sarmiento, L., et al. (2013) Evidence of association between type 1 diabetes and exposure to
enterovirus in Cuban children and adolescents. MEDICC review 15, 29-32
13 Whitehead, J.E. (1972) Viral epidemiology: the hidden universality of infection. British medical
journal 2, 451-456
14 Roep, B.O. and Tree, T.I. (2014) Immune modulation in humans: implications for type 1
diabetes mellitus. Nat Rev Endocrinol 10, 229-242
15 Kaufman, D.L., et al. (1992) Autoimmunity to two forms of glutamate decarboxylase in insulindependent diabetes mellitus. The Journal of clinical investigation 89, 283-292
16 Hou, J., et al. (1994) Antibodies to glutamic acid decarboxylase and P2-C peptides in sera from
coxsackie virus B4-infected mice and IDDM patients. Diabetes 43, 1260-1266
17 Lonnrot, M., et al. (1996) Antibody cross-reactivity induced by the homologous regions in
glutamic acid decarboxylase (GAD65) and 2C protein of coxsackievirus B4. Childhood Diabetes in
Finland Study Group. Clin Exp Immunol 104, 398-405
18 Marttila, J., et al. (2001) Responses of coxsackievirus B4-specific T-cell lines to 2C proteincharacterization of epitopes with special reference to the GAD65 homology region. Virology 284,
131-141
19 Roep, B.O., et al. (2002) Molecular mimicry in type 1 diabetes: immune cross-reactivity between
islet autoantigen and human cytomegalovirus but not Coxsackie virus. Ann N Y Acad Sci 958, 163165
20 Schloot, N.C., et al. (1997) T-cell reactivity to GAD65 peptide sequences shared with coxsackie
virus protein in recent-onset IDDM, post-onset IDDM patients and control subjects. Diabetologia
40, 332-338
21 Afonso, G. and Mallone, R. (2013) Infectious triggers in type 1 diabetes: is there a case for
epitope mimicry? Diabetes, obesity & metabolism 15 Suppl 3, 82-88
22 Oikarinen, M., et al. (2008) Detection of enteroviruses in the intestine of type 1 diabetic patients.
Clin Exp Immunol 151, 71-75
23 Yoon, J.W., et al. (1979) Isolation of a virus from the pancreas of a child with diabetic
ketoacidosis. The New England journal of medicine 300, 1173-1179
14
24 Dotta, F., et al. (2007) Coxsackie B4 virus infection of beta cells and natural killer cell insulitis in
recent-onset type 1 diabetic patients. Proceedings of the National Academy of Sciences of the
United States of America 104, 5115-5120
25 Foulis, A.K., et al. (1990) A search for the presence of the enteroviral capsid protein VP1 in
pancreases of patients with type 1 (insulin-dependent) diabetes and pancreases and hearts of
infants who died of coxsackieviral myocarditis. Diabetologia 33, 290-298
26 Andreoletti, L., et al. (2009) Viral causes of human myocarditis. Archives of cardiovascular
diseases 102, 559-568
27 Harris, K.G. and Coyne, C.B. (2013) Enter at your own risk: how enteroviruses navigate the
dangerous world of pattern recognition receptor signaling. Cytokine 63, 230-236
28 Welch, J., et al. (2003) Frequency, viral loads, and serotype identification of enterovirus
infections in Scottish blood donors. Transfusion 43, 1060-1066
29 Richardson, S.J., et al. (2009) The prevalence of enteroviral capsid protein vp1 immunostaining
in pancreatic islets in human type 1 diabetes. Diabetologia 52, 1143-1151
30 Ylipaasto, P., et al. (2004) Enterovirus infection in human pancreatic islet cells, islet tropism in
vivo and receptor involvement in cultured islet beta cells. Diabetologia 47, 225-239
31 Hilton, D.A., et al. (1993) Demonstration of Coxsackie virus RNA in formalin-fixed tissue
sections from childhood myocarditis cases by in situ hybridization and the polymerase chain
reaction. J Pathol 170, 45-51
32 Richardson, S.J., et al. (2013) Expression of the enteroviral capsid protein VP1 in the islet cells
of patients with type 1 diabetes is associated with induction of protein kinase R and downregulation
of Mcl-1. Diabetologia 56, 185-193
33 Dotta, F., et al. (2007) Coxsackie B4 virus infection of beta cells and natural killer cell insulitis in
recent-onset type 1 diabetic patients. Proceedings of the National Academy of Sciences of the
United States of America 104, 5115-5120
34 Willcox, A., et al. (2011) Immunohistochemical analysis of the relationship between islet cell
proliferation and the production of the enteroviral capsid protein, VP1, in the islets of patients with
recent-onset type 1 diabetes. Diabetologia 54, 2417-2420
35 Stechova, K., et al. (2012) Case report: type 1 diabetes in monozygotic quadruplets. European
journal of human genetics : EJHG 20, 457-462
36 Hilton, D.A., et al. (1992) Demonstration of the distribution of coxsackie virus RNA in neonatal
mice by non-isotopic in situ hybridization. Journal of virological methods 40, 155-162
37 Alirezaei, M., et al. (2012) Pancreatic acinar cell-specific autophagy disruption reduces
coxsackievirus replication and pathogenesis in vivo. Cell Host Microbe 11, 298-305
38 Moon, M.S., et al. (2005) Distribution of viral RNA in mouse tissues during acute phase of
coxsackievirus B5 infection. Intervirology 48, 153-160
39 Spagnuolo, I., et al. (2013) Pancreatic islet expression of enteroviral receptors and dsRNA
sensors in human type 1 diabetes. Diabetologia 56, S139-140
40 Oikarinen, M., et al. (2008) Analysis of pancreas tissue in a child positive for islet cell
antibodies. Diabetologia 51, 1796-1802
41 Dorner, A., et al. (2004) Alternatively spliced soluble coxsackie-adenovirus receptors inhibit
coxsackievirus infection. J Biol Chem 279, 18497-18503
42 Drescher, K.M., et al. (2004) Coxsackievirus B3 infection and type 1 diabetes development in
NOD mice: insulitis determines susceptibility of pancreatic islets to virus infection. Virology 329,
381-394
43 Inal, J.M. and Jorfi, S. (2013) Coxsackievirus B transmission and possible new roles for
extracellular vesicles. Biochemical Society transactions 41, 299-302
44 Kim, K.S., et al. (2005) 5'-Terminal deletions occur in coxsackievirus B3 during replication in
murine hearts and cardiac myocyte cultures and correlate with encapsidation of negative-strand
viral RNA. J Virol 79, 7024-7041
45 Elshebani, A., et al. (2007) Effects on isolated human pancreatic islet cells after infection with
strains of enterovirus isolated at clinical presentation of type 1 diabetes. Virus Res 124, 193-203
46 Hodik, M., et al. (2013) Tropism Analysis of Two Coxsackie B5 Strains Reveals Virus Growth in
Human Primary Pancreatic Islets but not in Exocrine Cell Clusters In Vitro. The open virology
journal 7, 49-56
15
47 Anagandula, M., et al. (2014) Infection of human islets of langerhans with two strains of
coxsackie B virus serotype 1: Assessment of virus replication, degree of cell death and induction of
genes involved in the innate immunity pathway. J Med Virol 86, 1402-1411
48 Tanaka, S., et al. (2009) Enterovirus infection, CXC chemokine ligand 10 (CXCL10), and
CXCR3 circuit: a mechanism of accelerated beta-cell failure in fulminant type 1 diabetes. Diabetes
58, 2285-2291
49 Kobayashi, T., et al. (2011) Pathological changes in the pancreas of fulminant type 1 diabetes
and slowly progressive insulin-dependent diabetes mellitus (SPIDDM): innate immunity in fulminant
type 1 diabetes and SPIDDM. Diabetes/metabolism research and reviews 27, 965-970
50 Tanaka, S., et al. (2013) Pathophysiological mechanisms involving aggressive islet cell
destruction in fulminant type 1 diabetes. Endocr J 60, 837-845
51 Sayama, K., et al. (2005) Pancreatic beta and alpha cells are both decreased in patients with
fulminant type 1 diabetes: a morphometrical assessment. Diabetologia 48, 1560-1564
52 Foulis, A.K., et al. (1988) Massive synchronous B-cell necrosis causing type 1 (insulindependent) diabetes--a unique histopathological case report. Diabetologia 31, 46-50
53 Chapman, N.M., et al. (2008) 5' terminal deletions in the genome of a coxsackievirus B2 strain
occurred naturally in human heart. Virology 375, 480-491
54 Kim, K.S., et al. (2008) Replication of coxsackievirus B3 in primary cell cultures generates novel
viral genome deletions. J Virol 82, 2033-2037
55 Skog O, I.S., Korsgren O (2014) Evaluation of RT-PCR and immunohistochemistry as tools for
detection of enterovirus in the human pancreas and isolated islets of Langerhans. Journal of
Clinical Virology
56 Sane, F., et al. (2013) Coxsackievirus B4 can infect human pancreas ductal cells and persist in
ductal-like cell cultures which results in inhibition of Pdx1 expression and disturbed formation of
islet-like cell aggregates. Cellular and Molecular Life Sciences 70, 4169-4180
57 Lysy, P.A., et al. (2013) Making beta cells from adult cells within the pancreas. Curr Diab Rep
13, 695-703
58 Cunningham, L., et al. (1990) Persistence of enteroviral RNA in chronic fatigue syndrome is
associated with the abnormal production of equal amounts of positive and negative strands of
enteroviral RNA. J Gen Virol 71 ( Pt 6), 1399-1402
59 Klingel, K., et al. (1992) Ongoing enterovirus-induced myocarditis is associated with persistent
heart muscle infection: quantitative analysis of virus replication, tissue damage, and inflammation.
Proceedings of the National Academy of Sciences of the United States of America 89, 314-318
60 Tam, P.E. and Messner, R.P. (1999) Molecular mechanisms of coxsackievirus persistence in
chronic inflammatory myopathy: viral RNA persists through formation of a double-stranded
complex without associated genomic mutations or evolution. J Virol 73, 10113-10121
61 Hultcrantz, M., et al. (2007) Interferons induce an antiviral state in human pancreatic islet cells.
Virology 367, 92-101
62 Shibasaki, S., et al. (2010) Expression of Toll-like Receptors in the Pancreas of Recent-onset
Fulminant Type 1 Diabetes. Endocr J 57, 211-219
63 Aida, K., et al. (2011) RIG-I- and MDA5-initiated innate immunity linked with adaptive immunity
accelerates beta-cell death in fulminant type 1 diabetes. Diabetes 60, 884-889
64 Lind, K., et al. (2013) Induction of an Antiviral State and Attenuated Coxsackievirus Replication
in Type III Interferon-Treated Primary Human Pancreatic Islets. J Virol 87, 7646-7654
65 Pichlmair, A., et al. (2009) Activation of MDA5 requires higher-order RNA structures generated
during virus infection. J Virol 83, 10761-10769
66 Richardson, S.J., et al. (2011) Enteroviral capsid protein, VP1, co-localises with Protein Kinase
R in the islets of patients with recent-onset type 1 diabetes. Diabet Med 28, 13
67 Downes, K., et al. (2010) Reduced expression of IFIH1 is protective for type 1 diabetes. PLoS
One DOI: 10.1371/journal.pone.0012646 (http://www.ncbi.nlm.nih.gov/pubmed/20844740)
68 Winkler, C., et al. (2011) An interferon-induced helicase (IFIH1) gene polymorphism associates
with different rates of progression from autoimmunity to type 1 diabetes. Diabetes 60, 685-690
69 Nejentsev, S., et al. (2009) Rare variants of IFIH1, a gene implicated in antiviral responses,
protect against type 1 diabetes. Science (New York, N.Y 324, 387-389
70 Liu, S., et al. (2009) IFIH1 polymorphisms are significantly associated with type 1 diabetes and
IFIH1 gene expression in peripheral blood mononuclear cells. Hum Mol Genet 18, 358-365
16
71 Foulis, A.K., et al. (1987) Aberrant expression of class II major histocompatibility complex
molecules by B cells and hyperexpression of class I major histocompatibility complex molecules by
insulin containing islets in type 1 (insulin-dependent) diabetes mellitus. Diabetologia 30, 333-343
72 Bottazzo, G.F., et al. (1985) In situ characterization of autoimmune phenomena and expression
of HLA molecules in the pancreas in diabetic insulitis. The New England journal of medicine 313,
353-360
73 Richardson, S.J., et al. (2014) Pancreatic pathology in type 1 diabetes mellitus. Endocrine
pathology 25, 80-92
74 Foulis, A.K., et al. (1987) Immunoreactive alpha-interferon in insulin-secreting beta cells in type
1 diabetes mellitus. Lancet 2, 1423-1427
75 Huang, X., et al. (1995) Interferon expression in the pancreases of patients with type I diabetes.
Diabetes 44, 658-664
76 Coppieters, K.T., et al. (2012) Demonstration of islet-autoreactive CD8 T cells in insulitic lesions
from recent onset and long-term type 1 diabetes patients. The Journal of experimental medicine
209, 51-60
77 Quah, H.S., et al. (2014) Deficiency in Type I Interferon Signaling Prevents the Early InterferonInduced Gene Signature in Pancreatic Islets but Not Type 1 Diabetes in NOD Mice. Diabetes 63,
1032-1040
78 Ferreira, R.C., et al. (2014) A type I interferon transcriptional signature precedes autoimmunity
in children genetically at risk for type 1 diabetes. Diabetes 63, 2538-2550
79 Kallionpaa, H., et al. (2014) Innate Immune Activity Is Detected Prior to Seroconversion in
Children With HLA-Conferred Type 1 Diabetes Susceptibility. Diabetes 63, 2402-2414
80 Skog, O., et al. (2011) Modulation of innate immunity in human pancreatic islets infected with
enterovirus in vitro. J Med Virol 83, 658-664
81 Ylipaasto, P., et al. (2005) Global profiling of coxsackievirus- and cytokine-induced gene
expression in human pancreatic islets. Diabetologia 48, 1510-1522
82 Skaug, B. and Chen, Z.J. (2010) Emerging role of ISG15 in antiviral immunity. Cell 143, 187190
83 Levin, D., et al. (2014) Multifaceted activities of type I interferon are revealed by a receptor
antagonist. Science signaling 7, ra50
84 Flodstrom-Tullberg, M., et al. (2005) RNase L and double-stranded RNA-dependent protein
kinase exert complementary roles in islet cell defense during coxsackievirus infection. J Immunol
174, 1171-1177
85 Garcia, M.A., et al. (2006) Impact of protein kinase PKR in cell biology: from antiviral to
antiproliferative action. Microbiol Mol Biol Rev 70, 1032-1060
Glossary:
Coxsackievirus: a virus that belongs to the family Picornaviridae and the genus Enterovirus of
nonenveloped, linear, positive-sense ssRNA viruses.
Coxsackie-Adenovirus Receptor (CAR): a type I membrane receptor that is expressed in tissues
such as heart, brain, and in epithelial and endothial cells. It may function as a cell adhesion
molecule at tight junctions through its interactions with extracellular matrix glycoproteins.
17
Decay Accelerating Factor (DAF): is a membrane glycoprotein that regulates the complement
system on the cell surface. DAF is used as a receptor by some coxsackieviruses and enteroviruses.
Enterovirus: is a member of the Picornaviridae family, and the genus Enterovirus that includes a
diverse group of small RNA viruses characterized by a single positive-strand genomic RNA.
Enteroviruses, which include polioviruses, echoviruses and Coxsackie viruses, are often found in
the respiratory secretions and stool of infected persons and have been associated with several
human and mammalian diseases.
Fulminant diabetes: a form of insulin-dependent diabetes which has been seen most commonly in
Japanese populations and occurs by virtue of an acute, large-scale, destruction of islet cells,
possibly mediated by a lytic enteroviral infection of the islet cells.
Insulitis: is a term defining the influx of immune cells around the periphery or within the islets of
Langerhans in the pancreas. It is associated with the elaboration of pro-inflammatory cytokines
that play a role in mediating β-cell death.
Interferons: are signaling proteins synthesised and released by host cells in response to the
presence of pathogens, such as viruses. Release of interferons from infected cells induces
protective responses in neighbouring cells, thereby reducing the likelihood of the spread of
infection.
ISG15: is a protein induced by the release of interferons that becomes covalently linked to newly
synthesised proteins (including viral proteins in infected cells) targeting them for degradation. The
process of “ISG15ylation” may also inhibit the assembly of functional viral capsids in virally
infected cells.
Islet autoantibodies: are antibodies directed against one or more of an individual’s own islet cell
proteins. These are often produced as an early response to islet damage during the development
of type 1 diabetes.
18
Glutamate decarboxylase (GAD): is the principal islet autoantigen in T1D. It is an enzyme with a
molecular mass of approximately 64kDa which is expressed in pancreatic β-cells and serves as an
autoantigen in many patients with T1D. Production of autoantibodies to GAD can also be detected
frequently in the circulation in close relatives of patients with T1D who do not, themselves,
necessarily go on to develop the disease.
Mda5: is a pattern recognition receptor encoded by the IFIH1 gene, which has been associated
with a pre-disposition to type 1 diabetes by genome wide association studies. Mda5 is a helicase
enzyme which binds dsRNA to initiate antiviral responses in cells.
Myocarditis: defines the inflammation of heart muscle, frequently associated with an enteroviral
infection. In young children with an acute enteroviral infection, myocarditis can be fatal.
Non-obese diabetic (NOD) mouse: is an animal model of type 1 diabetes, used very widely as a
surrogate for the human disease. NOD mice exhibit a susceptibility to spontaneous development
of T1D, usually with a marked female bias. Beta cells are selectively destroyed by a process of
insulitis in NOD mice although the extent of insulitis is much greater than that typically seen in
humans with T1D.
Pattern recognition receptors (PRRs): are a class of molecules which detect and respond to
sequences common to many forms of pathogens.
Protein kinase R (PKR): is a protein kinase and a member of one class of PRRs which becomes
activated in response to the presence of dsRNA in cells (and, possibly, also by other stressors,
including endoplasmic reticulum stress). Its primary substrate is a eukaryotic initiation factor,
eIF2α, which, when phosphorylated by PKR, attenuates the rate of protein synthesis in cells.
19
Figure legends
Figure 1. Schematic representation of the sequence of events which derive from infection of
pancreatic β-cells with either a fully replicative enterovirus (left; red circles) or a replication
deficient enterovirus (right; black circles).
The sequence of events arising from infection of islets is summarised in each case via a flow
diagram with the key components highlighted in sequentially numbered circles and described in
the accompanying box.
Figure 2. Schematic representation of the possible sequence of events that might ensue following
enteroviral infection in humans
An initial respiratory or GI tract infection could spread to the pancreas and may then either be
resolved, lead to acute islet cells loss and fulminant diabetes (especially in Japanese populations)
or establish a persistent infection within β-cells. The cells may then either respond only modestly
leading to long-term persistence and, conceivably, no adverse consequences or, alternatively, they
may respond strongly in an attempt to resolve the infection. In the latter case we propose that the
relative “strength” of the response could be critical to the development of autoimmunity and T1D.
20
Download