ArticlePDF AvailableLiterature Review

CLINICAL REVIEW: Type 1 Diabetes and Latent Autoimmune Diabetes in Adults: One End of the Rainbow

Authors:

Abstract

The aim of this review was to explore the pathogenic and clinical spectrum of type 1 diabetes, which includes a form of adult onset autoimmune diabetes usually referred to as latent autoimmune diabetes in adults (LADA). We looked at this entire range of forms of autoimmune diabetes as a spectrum of genetic and nongenetic environmental influences, diabetes-associated immune responses, and metabolic changes. We assessed epidemiological, genetic, immunological, and clinical data from major articles on autoimmune diabetes, including LADA and type 1 diabetes, published since 1992. Data analysis of autoimmune diabetes indi-cates that type 1 diabetes and LADA occupy different poles of the same spectrum. Evidence is presented that LADA represents one end of a rainbow encompassing type 1 diabetes. The clinical nature and management of autoimmune diabetes poses important therapeutic questions regarding conventional therapy for hyperglycemia as well as therapy aiming to protect residual beta-cell function. Limiting loss of endogenous insulin secretion using immunomodulation could be valuable, not only for LADA but also for type 1 diabetes.
CLINICAL REVIEW: Type 1 Diabetes and Latent
Autoimmune Diabetes in Adults: One End of
the Rainbow
R. David G. Leslie, Rhys Williams, and Paolo Pozzilli
Institute of Cell and Molecular Science (R.D.G.L., P.P.), Queen Mary College, University of London, London E1 4NS, United
Kingdom; University of Wales School of Medicine (R.W.), Swansea SA2 8PP, United Kingdom; and Department of
Endocrinology and Diabetes (P.P.), University Campus Bio-Medico, 83-00155 Rome, Italy
Context: The aim of this review was to explore the pathogenic and
clinical spectrum of type 1 diabetes, which includes a form of adult
onset autoimmune diabetes usually referred to as latent autoimmune
diabetes in adults (LADA). We looked at this entire range of forms of
autoimmune diabetes as a spectrum of genetic and nongenetic envi-
ronmental influences, diabetes-associated immune responses, and
metabolic changes.
Evidence Acquisition: We assessed epidemiological, genetic, im-
munological, and clinical data from major articles on autoimmune
diabetes, including LADA and type 1 diabetes, published since 1992.
Evidence Synthesis: Data analysis of autoimmune diabetes indi-
cates that type 1 diabetes and LADA occupy different poles of the
same spectrum.
Conclusion: Evidence is presented that LADA represents one end of
a rainbow encompassing type 1 diabetes. The clinical nature and
management of autoimmune diabetes poses important therapeutic
questions regarding conventional therapy for hyperglycemia as well
as therapy aiming to protect residual
-cell function. Limiting loss of
endogenous insulin secretion using immunomodulation could be valu-
able, not only for LADA but also for type 1 diabetes. (J Clin Endo-
crinol Metab 91: 1654 –1659, 2006)
TYPE 1 DIABETES RESULTS from the destruction of the
insulin-secreting islet cells by an immune mediated
process. This adverse immune response is induced and pro-
moted by the interaction of genetic and environmental fac-
tors and is one of a group of autoimmune diseases that affect
about 10% of the population in the developed world. Type
1 diabetes used to be defined in terms of the absolute need
for insulin therapy (insulin-dependent diabetes) or, before
that, the age at onset of the disease (juvenile onset diabetes).
These defining features were then abandoned in favor of the
term type 1 diabetes (1) when it became apparent that not
everyone with autoimmune diabetes is either a juvenile or
necessarily exhibits an absolute insulin requirement.
Individuals diagnosed with autoimmune diabetes, i.e. di-
abetes associated with diabetes-associated autoantibodies,
when they are adults may not initially require insulin treat-
ment and have been classified as having latent autoimmune
diabetes of adults (LADA) (2, 3), latent because without
testing for diabetes-associated autoantibodies it would not be
possible to identify these patients as having autoimmune
diabetes and adult because at that time it was suggested that
this form of diabetes was not present in juveniles. This form
of diabetes has also been called slowly progressing insulin-
dependent diabetes (4) or type 1.5 diabetes (5). The aim of this
article was to explore the clinical and pathogenic spectrum
of autoimmune diabetes that extends into and includes
LADA.
LADA is defined by three features including: adult age at
diagnosis, the presence of diabetes-associated autoantibod-
ies, and delay from diagnosis in the need for insulin therapy
to manage hyperglycemia. However, the first and last are not
categorical traits, being dependent on the mode of ascertain-
ment and decision making by physicians. The second feature
lacks disease specificity because it is based on positivity for
autoantibodies found in type 1 diabetes mellitus. In a recent
review (6), it was suggested that LADA patients should be
diagnosed with non-insulin-requiring diabetes at age 30 yr or
older and that age (range 30–70 yr) was also used in a major
European Union initiative (www.actionlada.org); in addi-
tion, both defined LADA to include patients who had 6
months without insulin treatment after diagnosis (6). Other
large studies of autoimmune non-insulin-requiring patients
have included selected cases, cases not taking any pharma-
cological agent, or avoided a definition entirely (7–9). Diffi-
culties with the performance of islet cell and insulin auto-
antibody assays precluded them from being used routinely
in defining LADA. Because insulinoma-associated antigen
(IA)-2 autoantibodies are usually found with glutamic acid
decarboxylase (GAD) autoantibodies but rarely in LADA,
this condition is broadly defined by the presence of GAD
autoantibodies. However, GAD autoantibodies are also
found in type 1 diabetes mellitus, so it follows that using
them in the definition of LADA lacks disease specificity (10).
The epidemiology of LADA is also influenced by geography,
First Published Online February 14, 2006
Abbreviations: GAD, Glutamic acid decarboxylase; HLA, human leu-
kocyte antigen; IA, insulinoma-associated antigen; IAA, insulin auto-
antibody; ICA, islet cell autoantibody; LADA, latent autoimmune dia-
betes in adults.
JCEM is published monthly by The Endocrine Society (http://www.
endo-society.org), the foremost professional society serving the en-
docrine community.
0021-972X/06/$15.00/0 The Journal of Clinical Endocrinology & Metabolism 91(5):1654 –1659
Printed in U.S.A. Copyright © 2006 by The Endocrine Society
doi: 10.1210/jc.2005-1623
1654
on May 9, 2006 jcem.endojournals.orgDownloaded from
genetic susceptibility, environmental factors, gender, and
age at diagnosis. In Northern Europe and North America,
about 5–10% of newly diagnosed non-insulin-requiring di-
abetes patients have LADA, according to the mode of as-
certainment, the sourced population, the age of the patient
(frequency is higher in younger age groups), and the defi-
nition of the disease (5, 6, 10).
LADA at One End of a Spectrum of Genetic
Susceptibility
The epidemiology of autoimmune diabetes including type
1 diabetes is influenced by genetic susceptibility, which mod-
ifies age at onset (6). There is evidence in autoimmune di-
abetes for a continuum of genetic susceptibility, which ex-
tends from a marked effect in childhood-onset type 1
diabetes to the relatively limited effect detected in LADA
(Table 1). Survival analysis estimates that nondiabetic iden-
tical twins of probands diagnosed with type 1 diabetes under
25 yr of age have a 38% probability of developing diabetes,
compared with only 6% for twins of probands diagnosed
later (11–13) (Table 2). Such a remarkably low twin concor-
dance for adult-onset type 1 diabetes implies that the genetic
impact in adult-onset diabetes is limited (14, 15), favoring a
substantial impact of environmental factors. There is an age-
related continuum in diminishing twin concordance, sug-
gesting that the decline in genetic influence is gradual, con-
sistent with a phased influence on a single disease, type 1
diabetes, rather than an age-related step-wise effect resulting
from two distinct diseases. There are, as yet, no twin studies
of LADA. Of genes implicated in the genetic susceptibility to
type 1 diabetes and LADA, the most important, for both, are
in the histocompatibility [human leukocyte antigen (HLA)]
region of chromosome 6 (14).
HLA alleles associated with type 1 diabetes susceptibility
include HLA DR3, DQB1*0201, and DR4, DQB1*0302,
whereas others are associated with disease protection, e.g.
HLA DR2, DQB1*0602 (16 –19). Children with type 1 diabetes
show an increased prevalence of the heterozygous alleles
HLA DR3, DQB1*0201, and DR4, DQB1*0302, the proportion
of heterozygotes declining with age at diagnosis (19). Chil-
dren with the diabetes-protective HLA DR2, DQB1*0602, are
unlikely to develop diabetes (20), whereas in type 1 diabetes
of adult-onset and LADA, the same alleles carry less pro-
tection (21, 22) (Table 1). Nevertheless, both the latter and
LADA show HLA genetic susceptibility with little or no HLA
genetic protection (19–21).
Strikingly, even adults with non-insulin-requiring diabe-
tes without the diabetes-associated autoantibody to GAD
have an excess of diabetes-associated HLA alleles and are
relatively young and lean (7, 23–25). Age-related genetic
factors also influence the risk of type 1 diabetes. Not only is
the age incidence of type 1 diabetes lower in adults than in
children, the range of incidence across European countries is
also reduced in adults (26). Furthermore, there is a male
excess in incidence that becomes evident during puberty and
is most striking in the age group 25–29 yr (26).
A recent, albeit small, genetic study (8) found similar HLA
susceptibility genes in both type 1 diabetes and LADA. Other
genes have been linked to type 1 diabetes and these genes,
including TNF
, TNF
, IL-10, IL-6 gene polymorphisms, and
IL-18 gene promoter polymorphism, but they have yet to be
studied comprehensively in LADA (27, 28). Other gene poly-
morphisms within the CTLA4, PTPN22, IRS-1, ICOS, and
SUMO4 genes confer a substantial risk to type 1 diabetes with
odds ratios between 1.8 and 2.5 but have not been studied in
LADA (29).
In the light of these observations, it remains possible that
LADA represents one end of a rainbow of autoimmune di-
abetes, which is distinguished from classic type 1 diabetes
only because it is diagnosed in adulthood and presents with
some clinical, anthropometric, and metabolic features more
commonly associated with type 2 diabetes.
LADA in a Spectrum of Nongenetic Influences
Nongenetic factors play a major role in causing type 1
diabetes as shown by studies of populations that have mi-
grated, populations with changing disease incidence, and
twins. We know little of the current incidence of autoimmune
diabetes in adults and LADA. The incidence of a range of
autoimmune diseases, including diabetes, has increased no-
tably over the last three decades (30). The current low se-
lection density and relative stability of HLA gene polymor-
phisms indicates that this increasing incidence cannot be due
to genetic selection pressures and is most likely the result of
nongenetic factors (10, 15).
Unfortunately, population studies are of limited value in
TABLE 1. Genetic, immunological, and metabolic differences between childhood-onset and adult-onset type 1 diabetes and LADA
Children T1DM Adults T1DM LADA
Age at diagnosis Childhood Adulthood Adulthood
Identical twin concordance rate Moderate (e.g. 38%) Very low (e.g. 6%) ?
HLA-DR3/ DR4 Moderate (e.g. 37%) Low (e.g. 13%) Low-moderate (e.g. 22%)
Protective HLA genotype (HLA-DR2) Very low (e.g. 9%) Low (e.g. 15%) Low-moderate (e.g. 22%)
Autoantibodies IAA GAD IA-2 GAD IA-2 GAD IA-2
Plasma insulin Very low Low Low
Note that the children, compared with the others, have a higher identical twin concordance rate, frequency of HLA genetic susceptibility
heterozygosity and insulin autoantibodies, and lower serum insulin levels. HLA DR3/4 is found in about 6% of North American and European
control populations. Data compiled from different sources (6, 11, 17, 19–22). T1DM, Type 1 diabetes mellitus; ?, rate unknown.
TABLE 2. Concordance for type 1 diabetes in identical twins
according to age at clinical onset in the index twin
Young-onset, % (yr) Older-onset, % (yr)
UK/US (Ref. 11) 38 (25) 6 (25)
US (Ref. 12) 44 (15) 13 (15)
Finland (Ref. 13) 50 (10) 23 (10)
Note the substantially lower concordance rates in the older-onset
twins consistent with a marked nongenetically determined effect
causing diabetes in them.
Leslie et al. Type 1 Diabetes and Latent Autoimmune Diabetes J Clin Endocrinol Metab, May 2006, 91(5):1654 –1659 1655
on May 9, 2006 jcem.endojournals.orgDownloaded from
identifying the impact of nongenetic factors because it is
difficult to segregate genetic from environmental influences.
However, changes in disease incidence within a genetically
stable population are important when disease incidence rises
rapidly or changes abruptly as in migrants (31, 32). Migrant
studies support a role for environmental factors influencing
disease incidence (32, 33). Type 1 diabetes incidence in Asian
children in families who have migrated to Britain increased
from 3.1 per 100,000 per year in 1978 –1981 to 11.7 per 100,000
per year in 1988–1990, much higher than in their native
Karachi (1 per 100,000 per year) (29, 30). However, Sardinian
migrants moving to continental Italy retained the high inci-
dence of the ancestry region, suggesting that it is the genetic
susceptibility that determines the prevalence of the disease
in response to the environmental factors (34). There are no
comparable migration studies of adults with type 1 diabetes
or of LADA patients. On the other hand, the identical twin
concordance for adult-onset type 1 diabetes is low, implying
that the genetic impact on this form of diabetes is limited,
which in turn suggests a major impact of environmental
factors (10). The declining identical twin concordance rate for
type 1 diabetes with increasing age appears to be a contin-
uum and not a categorical phenomenon, in line with an
age-related spectrum of environmental impact on the etiol-
ogy of autoimmune diabetes. However, there are no twin
studies in LADA, so it is unclear whether this spectrum
extends into that form of autoimmune diabetes.
LADA at One End of a Spectrum of Diabetes-
Associated Immune Responses
At birth, children of mothers with diabetes may have islet
cell autoantibodies (ICAs), insulin autoantibodies (IAAs),
and GAD autoantibodies. But these autoantibodies can also
be found in the maternal serum and are probably placentally
transferred to the child because autoantibody specificities are
similar in mother and cord blood and are not usually de-
tected in the infants of mothers without such autoantibodies
(35–37). Passively acquired maternal autoantibodies disap-
pear after birth as expected but can subsequently be replaced
by the infant’s own autoantibodies.
Diabetes-associated autoantibodies can appear at a later
stage. In one study, three of 58 infants of diabetic mothers
developed IAAs, ICAs, and GAD de novo by 2 yr of age, and
only then were autoantibodies associated with diabetes risk
(35), and in another study,137 children with ICAs from a
prospective Finnish study of 4590 consecutive newborns
with the disease-risk HLA-DQB1, IAAs, and GAD autoan-
tibodies usually appeared in childhood before ICAs, whereas
IA-2 autoantibodies usually appeared later when IAAs are
uncommon (38). In contrast to LADA and adult-onset type
1 diabetes, children often have IAAs at diagnosis and, in
them, IAA is highly predictive of the disease (39). Because
seroconversion continued up to at least age 10 yr of age, it
follows that the induction event with activation of immune
response to produce diabetes-associated autoantibodies is
not confined to early childhood. It remains unclear whether
the age at clinical diagnosis is in part dependent on the age
at which an environmental event activates the immune re-
sponse. If this is the case, then the immune process that leads
to adult-onset type 1 diabetes and LADA would be induced
later in them than in childhood-onset type 1 diabetes.
Taken together these observations suggest that activation
of the diabetes-associated immune process can occur in early
childhood when it is more likely to be associated with IAAs
in those who progress to childhood-onset type 1 diabetes. But
the induction of diabetes-associated autoantibodies is not
confined to early childhood. Currently we have no clear
evidence identifying the age of induction of diabetes-asso-
ciated autoantibodies in those subjects who develop either
adult-onset type 1 diabetes or LADA. However, we know
that these diabetes-associated autoantibodies, when detected
in adult life, are predictive of an ongoing
-cell destructive
process.
In this respect the prevalence of autoantibodies to protein
tyrosine phosphatase isoforms IA-2 and IA-2
/phogrin has
been recently examined in a cohort of adult U.K. Prospective
Diabetes Study patients thought to have type 2 diabetes to
determine whether these autoantibodies predict a require-
ment for insulin therapy (37). In this cohort the presence of
IA-2A was infrequent (about 2%), associated with the HLA-
DR4 haplotype as is the case in classic type 1 diabetes and
highly predictive of insulin therapy (positive predictive
value 60%). The measurement of IA-2
A does not provide
additional information (40).
LADA at One End of a Spectrum of
Metabolic Changes
There is evidence in autoimmune diabetes for a continuum
of metabolic changes, predominantly decreased insulin se-
cretory capacity, but also insensitivity to insulin. These ex-
tend from the severe changes seen in childhood-onset type
1 diabetes to the relatively minor changes initially detected
in LADA.
Some individuals pass through a prediabetic stage of im-
paired glucose tolerance or even non-insulin-requiring dia-
betes before becoming frankly insulin dependent (41). Dia-
betes Prevention Trial of Type 1 Diabetes detected 585
relatives of type 1 diabetic patients who had ICAs plus either
IAAs or low first-phase insulin response to iv glucose (42).
Of these, 427 had normal glucose tolerance, 87 impaired
glucose tolerance, and 61 were diabetic, yet asymptomatic
(39). Of the latter, those with impaired fasting glucose were
significantly older (mean age 21 yr) than those with normal
fasting glucose (mean age 12 yr). These subjects with asymp-
tomatic autoimmune diabetes resemble LADA, but their age
is less than 30 yr precluding the diagnosis. It follows that
some patients with autoimmune diabetes pass through a
phase of altered glucose levels including non-insulin-requir-
ing diabetes before becoming insulin dependent, and the
frequency of this phase, to a degree, is age dependent. It
remains to be determined whether all children with diabetes-
associated autoantibodies will progress to diabetes, let alone
insulin-dependent diabetes. The rate of progression to clin-
ical diabetes is more rapid in patients presenting younger
than 5 yr of age than in those presenting much later (43).
Histological evidence supports this contention: islet
-cells
tend to be absent within 12 months of diagnosis in patients
aged younger than 7 yr but detected for longer periods in
1656 J Clin Endocrinol Metab, May 2006, 91(5):1654 –1659 Leslie et al. Type 1 Diabetes and Latent Autoimmune Diabetes
on May 9, 2006 jcem.endojournals.orgDownloaded from
older patients (44). Variability in progression to clinical di-
abetes can even be detected in very young children; for
example, of children identified between 1 and 5 yr of age with
diabetes-associated autoantibodies and subnormal insulin
responses, half of them progress rapidly to diabetes, whereas
the remainder are free from diabetes up to 4 yr later (45).
Other studies have noted such variable progression to type
1 diabetes, which is more rapid in obese than lean children
(46) and in children than adults (47–50). From these obser-
vations it follows that there is a spectrum in the rate of
metabolic decompensation during the prediabetic period in
autoimmune type 1 diabetes, but no data are available, as yet,
in LADA.
Insulin secretory capacity is less in children than adults at
the onset of type 1 diabetes and after diagnosis deteriorates
more rapidly. A study of 235 consecutive cases with newly
diagnosed type 1 diabetes found that those aged younger
than 7 yr had the lowest baseline residual insulin secretion
and required the highest insulin dose for optimal control,
whereas the older the age at diagnosis, the higher was the
basal C-peptide level (51). Patients with LADA also have
reduced fasting and stimulated C-peptide at diagnosis, al-
though the levels of C-peptide are higher than those found
in children and similar to those found in adult-onset type 1
diabetes (8). However, after diagnosis, the C-peptide levels
have been reported to fall more rapidly in childhood-onset
type 1 diabetes than in adult-onset type 1 diabetes and in the
latter more rapidly than in LADA (52–55). Furthermore, per-
sistent C-peptide secretion, implying less aggressive disease,
is detected in more adults than adolescents after diagnosis of
type 1 diabetes and in more adolescents than prepubertal
children with diabetes (52–54). Other studies report a quite
rapid loss of C-peptide even in LADA, which argues against
a chronic destructive process in that condition (6, 55, 56). In
summary, there is a continuous spectrum of loss of insulin
secretory capacity, the severity of which can be age related,
being more severe in children than adults with type 1 dia-
betes and more severe in the latter than in LADA subjects,
although some patients with LADA may show a rapid loss
of insulin secretory capacity.
The metabolic decompensation that leads to frank diabetes
could result from either increased linear growth, which has
been linked to diabetes risk, or increased childhood obesity,
which has been correlated with age at presentation (10, 46).
People with LADA may well have more severe loss of insulin
sensitivity than in childhood-onset type 1 diabetes, but there
are only two small studies (57, 58) considering insulin sen-
sitivity in LADA, and both used the homeostasis model of
assessment, whereas no studies have used the gold standard
euglycemic hyperinsulinemic clamp. Certainly in LADA the
frequency of the metabolic syndrome, usually associated
with insulin resistance, although less prevalent than in type
2 diabetic patients of similar age, is more prevalent than in
the general population (9). In a recent report, the metabolic
syndrome, which is found in approximately 22% of the North
American population was identified in 74% of those with
LADA but in significantly more subjects with type 2 diabetes
(84%) (9).
It is likely, therefore, that within autoimmune diabetes,
including both type 1 diabetes and LADA, there is an age-
related spectrum of decreasing insulin secretory capacity and
increasing insulin insensitivity associated with the metabolic
syndrome. The distinction between LADA and adult-onset
classical type 1 diabetes is a matter of debate. It is possible
that the distinction between the two is one of degree, with the
classical type 1 diabetes being at one end of the spectrum and
LADA, when it remains insulin independent, being at the
other end.
A Spectrum of Clinical Management in LADA
Type 1 diabetes progresses to insulin dependence usually
within 2 yr of the clinical diagnosis as noted in the preinsulin
era. Before 2 yr some patients may have a partial or complete
remission when insulin therapy is not required (59). Of
LADA patients, in one study (7), 94% required insulin treat-
ment by 6 yr as compared with only 14% in those initially
non-insulin-requiring diabetes patients without either GAD
autoantibodies or ICAs. Progression to insulin dependence
in LADA patients was more rapid in those aged younger
than 45 yr than in older cases (7). It follows that patients with
autoimmune diabetes, including both type 1 diabetes and
LADA, are at high risk of progression to insulin dependence,
but that risk declines with age at diagnosis.
It is well established that insulin is the treatment of choice
for type 1 diabetes, but there is no established management
strategy for patients diagnosed with LADA (60 65). The
European Union, therefore, funded a major initiative
(ACTION-LADA) to study the characteristics of LADA and
report on how to treat it. In considering how to treat LADA,
some important questions arise as to our broad management
of autoimmune diabetes. Because the predominant defect in
autoimmune diabetes is loss of insulin secretion, should we
treat the disease with insulin irrespective of the level of
dependency on insulin? Autoantibody positive, initially non-
insulin-requiring, diabetic patients initially treated with sul-
fonylureas have been found to require insulin earlier than
autoantibody-negative patients, but sulfonylureas did not
have an impact on the need for insulin treatment or the time
to progression to insulin therapy (41).
Metformin is routinely offered to patients with non-insu-
lin-requiring diabetes, but its specific role in LADA is un-
clear, and the drug may be contraindicated in those with
LADA because there is a theoretical risk of severe metabolic
disturbance in individuals who progress to insulin depen-
dency while on it. Intriguingly, however, there is limited
evidence that metformin could be of value, even in patients
with type 1 diabetes. For example, in one study, adolescent
type 1 insulin-dependent diabetes patients given metformin
subsequently showed a significantly lower hemoglobin A1c
and reduced insulin requirements, compared with those not
taking metformin (62).
Thiazoledinediones also might theoretically be of value
because they not only improve insulin sensitivity but also
have an antiinflammatory effect and protect nonobese dia-
betic mice, a well-established model of autoimmune diabe-
tes, from developing diabetes (63). In a small study of LADA
patients in China (64), there was a significant improvement
in C-peptide but not hemoglobin A1c in patients receiving
rosiglitazone plus insulin, compared with insulin alone.
Leslie et al. Type 1 Diabetes and Latent Autoimmune Diabetes J Clin Endocrinol Metab, May 2006, 91(5):1654 –1659 1657
on May 9, 2006 jcem.endojournals.orgDownloaded from
Because the primary defect in autoimmune diabetes is loss
of insulin secretion, treatment should aim to restore islet
insulin secretion. Therapy to prevent progression toward
insulin dependency could include insulin or immunomodu-
lation, given the inflammatory nature of the disease process
thought to cause insulin secretory cell destruction. The op-
timal insulin regimen is unclear; given the broad loss of
insulin secretory capacity, it might be argued that the early
introduction of a long-acting insulin could be beneficial. Al-
ternatively, the loss of rapid insulin release in LADA patients
suggests that replacement with a fast-acting insulin would be
more valuable.
One study in Japan of patients with LADA compared early
treatment with insulin given as multiple injections with sul-
fonylureas (61). Although of limited power, this study did
show a statistically significant persistence of C-peptide in the
insulin-treated group as compared with the sulfonylurea
group with the proviso that the insulin-treated group had
preserved insulin secretory capacity and a high titer of GAD
autoantibodies at the start of the study (61). An alternative
interpretation of this study is that sulfonylureas are disad-
vantageous, in support of which sulfonylureas could theo-
retically promote apoptosis, apoptosis being one mechanism
whereby insulin-secreting cells could be destroyed in auto-
immune diabetes.
A pilot phase 2 trial in LADA patients (65) found that a
tolerance induction plan using alum-formulated whole GAD
(Diamyd) had a significant effect on the C-peptide response
to a mixed meal consistent with modulation of the aggressive
process. Another phase 2 trial in LADA patients using the
peptide analog of heat shock protein 60 (Diapep 277) has
been completed after initial positive results in protecting
residual
-cell function in adult-onset type 1 diabetes pa-
tients (66) and in experimental models of the disease (67).
These immunomodulatory studies, although small and pre-
liminary, pioneer a novel approach toward the maintenance
of islet cell function, itself a new field in the management of
autoimmune diabetes.
In conclusion, LADA, whether viewed genetically, immu-
nologically, metabolically, or clinically, occupies one end of
a rainbow of features associated with autoimmune diabetes.
The management and prevention of LADA need to be in-
vestigated to define the best strategy for treating this most
prevalent form of autoimmune diabetes.
Acknowledgments
We thank all our collaborators, in particular Drs. Mohammed Hawa,
Huriya Beyan, Chiara Guglielmi, Marta Vadacca, Sinead Brophy, and
Mark Airey, for their contribution to the work in our laboratories and
in the field.
Received July 21, 2005. Accepted February 7, 2006.
Address all correspondence and requests for reprints to: Professor
David Leslie, Department of Diabetes, St. Bartholomew’s Hospital, West
Smithfield, London EC1A 7BE, United Kingdom. E-mail: r.d.g.leslie@
qmul.ac.uk.
The work in London, Swansea, and Rome was supported by grants
from the Juvenile Diabetes Research Foundation, Develogen, Diabetes
UK, Diabetes Twin Research Trust, and the European Union.
The authors have nothing to declare.
References
1. The Expert Committee on the Diagnosis and Classification of Diabetes
Mellitus 1998 Report of the Expert Committee on the Diagnosis and Classi-
fication of Diabetes Mellitus. Diabetes Care 21:S3–S519
2. Tuomi T, Groop LC, Zimmet PZ, Rowley MJ, Knowles W, Mackay IR 1993
Antibodies to glutamic acid decarboxylase reveal latent autoimmune diabetes
mellitus in adults with a non-insulin-dependent onset of disease. Diabetes
42:359–362
3. Zimmet PZ, Elliot RB, Mackay IR, Rowley MJ, Pilcher CC, Knowles WJ 1994
Autoantibodies to glutamic acid decarboxylase and insulin in islet cell anti-
body positive presymptomatic type 1 diabetes mellitus: frequency and seg-
regation by age and gender. Diabet Med 11:866–871
4. Kobayashi T, Tamemoto K, Nakanishi K, Kato N, Okubo M, Kajio H,
Sugimoto T, Murase T, Kosaka K 1993 Immunogenetic and clinical charac-
terization of slowly progressive IDDM. Diabetes Care 16:780–788
5. Naik RG, Palmer JP 2003 Latent autoimmune diabetes in adults (LADA). Rev
Endocr Metab Disord 4:233–241
6. Fourlanos S, Dotta F, Greenbaum CJ, Palmer JP, Rolandsson O, Colman PG,
Harrison LC 2005 Latent autoimmune diabetes in adults (LADA) should be
less latent. Diabetologia 48:2206–2212
7. Turner R, Stratton I, Horton V, Manley S, Zimmet P, Mackay IR, Shattock
M, Bottazzo GF, Holman R 1997 UKPDS 25: autoantibodies to islet-cell cy-
toplasm and glutamic acid decarboxylase for prediction of insulin requirement
in type 2 diabetes. U.K. Prospective Diabetes Study Group. Lancet 350:1288
1293
8. Hosszufalusi N, Vatay A, Rajczy K, Prohaszka Z, Pozsonyi E, Horvath L,
Grosz A, Gero L, Madacsy L, Romics L, Karadi I, Fust G, Panczel P 2003
Similar genetic features and different islet cell autoantibody pattern of latent
autoimmune diabetes in adults (LADA) compared with adult-onset type 1
diabetes with rapid progression. Diabetes Care 26:452–457
9. Zinman B, Kahn SE, Haffner SM, O’Neill MC, Heise MA, Freed MI; ADOPT
Study Group 2004 Phenotypic characteristics of GAD antibody-positive re-
cently diagnosed patients with type 2 diabetes in North America and Europe.
Diabetes 53:3193–3200
10. Leslie RD, Delli Castelli M 2004 Age-dependent influences on the origins of
autoimmune diabetes: evidence and implications. Diabetes 53:3033–3040
11. Redondo MJ, Yu L, Hawa M, Mackenzie T, Pyke DA, Eisenbarth GS, Leslie
RDG 2001 Heterogeneity of type I diabetes: analysis of monozygotic twins in
Great Britain and the United States. Diabetologia 44:354–362
12. Kumar D, Gemayel NS, Deapen D, Kapadia D, Yamashita PH, Lee M, Dwyer
JH, Roy-Burman P, Bray GA, Mach TM 1993 North-American twins with
IDDM: genetic, etiological, and clinical significance of disease concordance
according to age, zygosity, and the interval after diagnosis in first twin. Di-
abetes 42:1351–1363
13. Hyttinen V, Kaprio J, Kinnunen L, Koskenvuo M, Tuomilehto J 2003 Genetic
liability of type 1 diabetes and the onset age among 22,650 young Finnish twin
pairs: a nationwide follow-up study. Diabetes 52:1052–1055
14. Field LL 2002 Genetic linkage and association studies of type 1 diabetes:
challenges and rewards. Diabetologia 45:21–35
15. Salvetti M, Ristori G, Bomprezzi R, Pozzilli P, Leslie RDG 2000 Twins:
mirrors of the immune system. Immunology Today 21:342–347
16. Lambert AP, Gillespie KM, Thomson G, Cordell HJ, Todd JA, Gale EA,
Bingley PJ 2004 Absolute risk of childhood-onset type 1 diabetes defined by
human leukocyte antigen class II genotype: a population-based study in the
United Kingdom. J Clin Endocrinol Metab 89:4037–4043
17. Greenbaum CJ, Schatz DA, Cuthbertson D, Zeidler A, Eisenbarth GS,
Krischer JP 2000 Islet cell antibody-positive relatives with human leukocyte
antigen DQA1* 0102, DQB1*0602: identification by the Diabetes Prevention
Trial-type 1. J Clin Endocrinol Metab 85:1255–1260
18. Sabbah E, Savola K, Ebeling T, Kulmala P, Vahasalo P, Ilonen J, Salmela PI,
Knip M 2000 Genetic, autoimmune, and clinical characteristics of childhood-
and adult-onset type 1 diabetes. Diabetes Care 23:1326–1332
19. Vanderwalle CL, Cecraene T, Schuit FC, De Leeuw IH, Pipeleers DG, Gorus
FK, the Belgian Diabetes Register 1993 Insulin antibodies and high titre islet
cell antibodies are preferentially associated with the HLA DQA1*03100-
DQB1*0302 haplotype at clinical onset of type 1 (insulin-dependent) diabetes
mellitus before age 10 years but not at onset between age 10 and 40 years.
Diabetologia 36:1155–1162
20. Petrone A, Bugawan TL, Mesturino CA, Nistico` L, Galgani A, Giorgi G,
Cascino I, Erlich HA, Di Mario U, Buzzetti R 2001 The distribution of HLA
class II susceptible/protective haplotypes could partially explain the low in-
cidence of type 1 diabetes in continental Italy (Lazio region). Tissue Antigens
58:385–394
21. Vatay A, Rajczy K, Pozsonyi E, Hosszufalusi N, Prohaszka Z, Fust G, Karadi
I, Szalai C, Grosz A, Bartfai Z, Panczel P 2002 Differences in the genetic
background of latent autoimmune diabetes in adults (LADA) and type 1
diabetes mellitus. Immunol Lett 84:109–115
22. Cerna M, Novota P, Kolostova K, Cejkova P, Zdarsky E, Novakova D, Kucera
P, Novak J, Andel M 2003 HLA in Czech adult patients with autoimmune
diabetes mellitus: comparison with Czech children with type 1 diabetes and
patients with type 2 diabetes. Eur J Immunogenet 30:401–407
1658 J Clin Endocrinol Metab, May 2006, 91(5):1654 –1659 Leslie et al. Type 1 Diabetes and Latent Autoimmune Diabetes
on May 9, 2006 jcem.endojournals.orgDownloaded from
23. Horton V, Stratton I, Bottazzo GF, Shattock M, Mackay I, Zimmet P, Manley
S, Holman R, Turner R, for the U.K. Prospective Diabetes Study (UKPDS)
Group 1999 Genetic heterogeneity of autoimmune diabetes: age at presenta-
tion in adults is influenced by HLA DRB1 and DQB1 genotypes (UKPDS 43).
Diabetologia 42:608–616
24. Pozzilli P, Di Mario U 2001 Autoimmune diabetes not requiring insulin at di-
agnosis (latent autoimmune diabetes of the adult). Diabetes Care 24:1460–1467
25. Weets I, Siraux V, Daubresse JC, De Leeuw IH, Fery F, Keymeulen B,
Krzentowski G, Letiexhe M, Mathieu C, Nobels F, Rottiers R, Scheen A, Van
Gaal L, Schuit FC, Van der Auwera B, Rui M, De Pauw P, Kaufman L, Gorus
FK 2002 Belgian Diabetes Registry: relation between disease phenotype and
HLA-DQ genotype in diabetic patients diagnosed in early adulthood. J Clin
Endocrinol Metab 87:2597–2605
26. Kyvik KO, Nystrom L, Gorus F, Songini M, Oestman J, Castell C, Green A,
Guyrus E, Ionescu-Tirgoviste C, Mckinney PA, Michalkova D, Ostrauskas
R, Raymond NT 2004 The epidemiology of type 1 diabetes mellitus is not the
same in young adults as in children. Diabetologia 47:377–384
27. Tsiavou A, Hatziagelaki E, Chaidaroglou A, Manginas A, Koniavitou K,
Degiannis D, Raptis SA 2004 TNF-
, TGF-
1, IL-10, IL-6, gene polymor-
phisms in latent autoimmune diabetes of adults (LADA) and type 2 diabetes
mellitus. J Clin Immunol 26:591–599
28. Novota P, Kolostova K, Pinterova D, Novak J, Treslova L, Andel M, Cerna
M2005 Interleukin-18 gene promoter polymorphisms in adult patients with
type 1 diabetes mellitus and latent autoimmune diabetes in adults. Immunol
Lett 96:247–251
29. Hornum L, Markholst H 2004 New autoimmune genes and the pathogenesis
of type 1 diabetes. Curr Diabet Rep 4:135–142
30. Bach JF 2002 The effect of infections on susceptibility to autoimmune and
allergic diseases. N Engl J Med 347:911–920
31. Serrano-Rı`os M, Goday A, Martinez Larrad T 1999 Migrant populations and
the incidence of type 1 diabetes mellitus: an overview of the literature with a
focus on the Spanish-heritage countries in Latin America. Diabetes Metab Res
Rev 15:113–132
32. Bodansky HJ, Saines A, Stephenson C, Haigh D, Cartwright R 1992 Evidence
for an environmental effect in the aetiology of insulin dependent diabetes in
a transmigratory population. Br Med J 304:1020–1022
33. Staines A, Hanif S, Ahmed S, Mckinney PA, Shera S, Bodansky HJ 1997
Incidence of insulin-dependent diabetes mellitus in Karachi, Pakistan. Arch
Dis Child 76:121–123
34. Muntoni S, Fonte MT, Stoduto S, Marietti G, Bizzarri C, Crino A, Ciampalini
P, Multari G, Suppa MA, Matteoli MC, Lucentini L, Sebastiani LM, Visalli
N, Pozzilli P, Boscherini B, Muntoni S 1997 Incidence of insulin-dependent
diabetes mellitus among Sardinian-heritage children born in Lazio region,
Italy. Lancet 349:160–162
35. Ziegler AG, Hummel M, Schenker M, Bonifacio E 1999 Autoantibody ap-
pearance and risk for development of childhood diabetes in offspring of
parents with type 1 diabetes: the 2-year analysis of the German BABYDIAB
study. Diabetes 48:460–468
36. Lindberg B, Ivarsson S-A, Landin-Olsson M, Sundkvist G, Svanberg L,
Lernmark A 1999 Islet autoantibodies in cord blood from children who de-
veloped type 1 (insulin-dependent) diabetes mellitus before 15 years of age.
Diabetologia 42:181–187
37. Hamalanian A-M, Savola K, Kulmala PK, Koskela P, Akerblom HK, Knip
M, The Finnish TRIGR Study Group 2001 Disease-associated autoantibodies
during pregnancy and at birth in families affected by type 1 diabetes. Clin Exp
Immunol 126:231–236
38. Kupila A, Keskinen P, Simell T, Erkkila, Arvilommi P, Korhonen S, Kimpi-
maki T, Sjoroos M, Ronkainen M, Ilonen J, Knip M, Simell O 2002 Genetic
risk determines the emergence of diabetes-associated autoantibodies in young
children. Diabetes 51:646–651
39. Kukko M, Kimpimaki T, Korhonen S, Kupila A, Simell S, Veijola R, Simell T,
Ilonen J, Simell O, Knip M 2005 Dynamics of diabetes-associated autoantibodies
in young children with human leukocyte antigen-conferred risk of type 1 diabetes
recruited from the general population. J Clin Endocrinol Metab 90:2012–2717
40. Bottazzo GF, Bosi E, Cull CA, Bonifacio E, Locatelli M, Zimmet P, Mackay
IR, Holman RR 2005 IA-2 antibody prevalence and risk assessment of early
insulin requirement in subjects presenting with type 2 diabetes (UKPDS 71).
Diabetologia 48:703–708
41. Davis TM, Wright AD, Mehta ZM, Cull CA, Stratton IM, Bottazzo GF, Bosi
E, Mackay IR, Holman RR 2005 Islet autoantibodies in clinically diagnosed
type 2 diabetes: prevalence and relationship with metabolic control (UKPDS
70). Diabetologia 48:695–702
42. Greenbaum CJ, Cuthbertson D, Krischer JP, Disease Prevention Trial of
Type I Diabetes Study Group 2001 Type I diabetes manifested solely by 2-h
oral glucose tolerance test criteria. Diabetes 50:470–476
43. Eisenbarth GS 2004 Prediction of type 1 diabetes: the natural history of the
prediabetic period. Adv Exp Med Biol 552:268–290
44. Pipeleers D, Ling Z 1992 Pancreatic
cells in insulin-dependent diabetes.
Diabetes Metab Rev 8:209–227
45. Keskinen P, Korhonen S, Kupila A, Veijola R, Erkkila S, Savolainen H,
Arvilommi P, Simell T, Ilonen J, Knip M, Simell O 2002 First-phase insulin
response in young healthy children at genetic and immunological risk for type
I diabetes. Diabetologia 45:1639–1648
46. Kibirige M, Metcalf B, Renuka R, Wilkin TJ 2003 Testing the accelerator
hypothesis: the relationship between body mass and age at diagnosis of type
1 diabetes. Diabetes Care 26:2865–2870
47. Bingley PJ, Bonifacio E, Gale EA 1993 Can we really predict IDDM? Diabetes
42:213–220
48. Gorus FK 1997 Diabetes registries and early biological markers of insulin-
dependent diabetes mellitus. Belgian Diabetes Registry. Diabetes Metab Rev
13:247–274
49. Barker JM, Barriga KJ, Yu L, Miao D, Erlich HA, Norris JM, Eisenbarth GS,
Rewers M, Diabetes Autoimmunity Study in Young 2004 Prediction of au-
toantibody positivity and progression to type 1 diabetes: Diabetes Autoim-
munity Study in the Young (DAISY). J Clin Endocrinol Metab 89:3896–3902
50. Pozzilli P, Visalli N, Buzzetti R, Cavallo MG, Marietti G, Hawa M, Leslie
RD 1998 Metabolic and immune parameters at clinical onset of insulin-de-
pendent diabetes: a population-based study. IMDIAB Study Group. Metab-
olism 47:1205–1210
51. The DCCT Research Group 1987 Effects of age, duration and treatment of
insulin-dependent diabetes mellitus on residual
-cell function: observations
during eligibility testing for the Diabetes Control and Complications Trial
(DCCT). J Clin Endocrinol Metab 65:30–36
52. Bonfanti R, Bazzigaluppi E, Calori G, Riva MC, Viscardi M, Bognetti E,
Meschi F, Bosi E, Chiumello G, Bonifacio E 1998 Parameters associated with
residual insulin secretion during the first year of disease in children and
adolescents with type 1 diabetes mellitus. Diabet Med 15:844–850
53. Martin S, Pawlowski B, Greulich B, Ziegler AG, Mandrup-Poulsen T, Ma-
hon J 1992 Natural course of remission in IDDM during 1st yr after diagnosis.
Diabetes Care 15:66–74
54. Palmer JP, Fleming GA, Greenbaum CJ, Herold KC, Jansa LD, Kolb H,
Lachin JM, Polonsky KS, Pozzilli P, Skyler JS, Steffes MW 2004 C-peptide
is the appropriate outcome measure for type 1 diabetes clinical trials to pre-
serve
-cell function. Diabetes 53:250–264
55. Gottsater A, Landin-Olsson M, Fernlund P, Lernmark A, Sundkvist G 1993
-Cell function in relation to islet cell antibodies during the first 3 yr after clinical
diagnosis of diabetes in type II diabetic patients. Diabetes Care16:902–910
56. Borg H, Gottsater A, Landin-Olsson M, Fernlund P, Sundkvist G 2001 High
levels of antigen-specific islet antibodies predict future
-cell failure in patients
with onset of diabetes in adult age. J Clin Endocrinol Metab 86:3032–3038
57. Carlsson A, Sundkvist G, Groop L, Tuomi T 2000 Insulin and glucagon
secretion in patients with slowly progressing autoimmune diabetes (LADA).
J Clin Endocrinol Metab 85:76–80
58. Behme MT, Dupre J, Harris SB, Hramiak IM, Mahon JL 2003 Insulin resistance
in latent autoimmune diabetes of adulthood. Ann NY Acad Sci 1005:374–377
59. Baroni M, Vialettes B, Pozzilli P 1997 The Glucose Evaluation Trial for
Remission (GETREM): a European effort to evaluate insulin-dependent dia-
betes mellitus in the first year after diagnosis. Diabetes Metab 23:264–265
60. Palmer J 2000 Therapeutic importance of subset of type 2 diabetes? Diabetes
Care 23:574–575
61. Moruyama T, Shimada A, Kanatsuka A, Kasuga A, Takei I, Yokoyama J,
Kobayashi T 2003 Multicenter prevention trials of slowly progressive type 1
diabetes with small dose of insulin (the Kyoto study): preliminary report. Ann
NY Acad Sci 1005:362–369
62. Hamilton J, Cummings E, Zdravkovic V, Finegood D, Daneman D 2003
Metformin as an adjunct therapy in adolescents with type 1 diabetes and
insulin resistance: a randomized controlled trial. Diabetes Care 26:138–143
63. Beales PE, Pozzilli P 2002 Thiazolidinediones for the prevention of diabetes
in the non-obese diabetic (NOD) mouse: implications for human type 1 dia-
betes. Diabetes Metab Res Rev 18:114–117
64. Zhou Z, Li X, Huang G, Peng J, Yang L, Yan X, Wang J 2005 Rosiglitazone
combined with insulin preserves islet
cell function in adult-onset latent
autoimmune diabetes (LADA). Diabetes Metab Res Rev 21:203–208
65. Agardh CD, Cilio CM, Lethagen A, Lynch K, Leslie RD, Palmer M, Harris
RA, Robertson JA, Lernmark A 2005 Clinical evidence for the safety of GAD65
immunomodulation in adult-onset autoimmune diabetes. J Diabetes Compli-
cations 19:238–246
66. Raz I, Elias D, Avron A, Tamir M, Metzger M, Cohen IR 2001
-cell function
in new-onset type 1 diabetes and immunomodulation with a heat-shock pro-
tein peptide (diaPep277): a randomised, double-blind, phase two trial. Lancet
358:1749–1753
67. Brugman S, Klatter FA, Visser J, Bos NA, Elias D, Rozing J 2004 Neonatal oral
administration of DiaPep277, combined with hydrolysed casein diet, protects
against type 1 diabetes in BB-DP rats. An experimental study. Diabetologia
47:1331–1333
JCEM is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the
endocrine community.
Leslie et al. Type 1 Diabetes and Latent Autoimmune Diabetes J Clin Endocrinol Metab, May 2006, 91(5):1654 –1659 1659
on May 9, 2006 jcem.endojournals.orgDownloaded from
... The eggs and sperms were placed in the IVF culture, transferred to an incubator waiting for fertilization, and then embryo transfer (E.T.). Incubation in a culture media continues for (16)(17)(18)(19)(20)(21)(22)(23)(24) hours. E.T. is usually done by choosing one or more good embryos to be returned to the wife's uterus. ...
... Genetic features, immunological aspects and environmental factors have a different weight in determining the onset of DM type 1depending on the age at the diagnosis. The genetic susceptibility extends from a marked effect in childhood-onset DM type 1 to the relatively limited effect detected in LADA (Latent Autoimmune Diabetes of the Adult) (Leslie RD., et al. 2006), as shown, for example, by the age-dependent diminishing twin concordance. It is clear, therefore, that there is a continuum in the pathological factors that lead to an age-related increased influence of immunological and environmental components despite the reduction in genetics weight. ...
Chapter
Full-text available
The SJUST, Scientific Journal of University of Science and Technology has three major issues: Medical, Engineering and Administrative Sciences. The current one is the Medical Sciences Issue No. 1 of Vol 1 of the Six Issue from the University Scientific Journal Issue No, 6
... Beta-cell destruction in adults appears to occur at a much slower rate than in young T1DM cases, often delaying the need for insulin therapy after diagnosis. 35 Younger adolescents in particular appear to have a more robust and aggressive autoimmune process with more rapid progression to disease and a shorter remission (or honeymoon) phase. 36 Not surprisingly, there have been suggestions of e cacy differences in younger vs. older subjects in clinical studies of anti-CD3 and anti-CD20 mAbs, with higher e cacy trends observed in younger cohorts. ...
Preprint
Full-text available
This protocol details a prospective randomized placebo-controlled double blind clinical trial to evaluate the safety and efficacy of CLBS03 (autologous ex vivo expanded polyclonal CD4+CD25+CD127lo/-FOXP3+ regulatory T-cells [Tregs]) in adolescents with recent onset Type 1 Diabetes Mellitus.
... A special subgroup of type 1 DM is LADA (late autoimmune diabetes in adults) that is characterized by beta-cell failure and high titers of anti-islet and anti-GAD (glutamic acid decarboxylase) antibodies. LADA leads to the profound impairment of glucose-induced insulin secretion and could sometimes be found in nonobese older patients incorrectly treated as T2DM [41]. ...
Article
Full-text available
Type 2 diabetes is a frequent chronic disease. Given its strong positive association with older age, it is a significant public health issue in elderly populations. Furthermore, the aging of the population, driven by increasing life expectancy in high and middle-income countries leads to an increasing prevalence of diabetes. Although the same diagnostic criteria apply to the elderly and to younger people, there are unique aspects to the care for elderly type 2 diabetes patients. Both treatment goals and preferred medications, as well as non-pharmacological approaches should be adjusted in the elderly. For example, increasing the amount of physical activity may encounter difficulties, while introducing an appropriate diet may be more challenging. The patients' therapeutic adherence requires special attention due to cognitive and physical limitations. The most important treatment goal is to avoid hypoglycemia. Frailty, social and economic issues, comorbidities and the consequent polypharmacy frequently causing drug-drug interactions, as well as the increased danger of drug toxicity due to renal failure are only some of the problems that make the health care for old diabetes patients extremely difficult. Adequate care requires cooperation from a multidisciplinary team of health care professionals. Acute diabetes complications have a higher mortality in the elderly, thus close attention must be paid to avoid them. Family members should be involved in the care of elderly diabetes patients, and it is recommended to educate them on clinical signs of complications. Regular care for the patients including feedback on quality of life and early signs of health issues are essential.
... Increased expression of vascular endothelial growth factor and angiogenesis in diabetes-associated AC has also been observed [27]. Previous reports also suggest an immunological pathogenesis of AC and DM [24,[28][29][30][31]. It is possible that these two conditions are related. ...
Article
Full-text available
Background This study aimed to investigate the association between type 2 diabetes mellitus (T2DM) and shoulder adhesive capsulitis (AC) using a large-scale, nationwide, population-based cohort in the Republic of Korea. Methods A total of 3,471,745 subjects aged over 20 years who underwent a National Health Insurance Service medical checkup between 2009 and 2010 were included in this study, and followed from the date of their medical checkup to the end of 2018. Subjects were classified into the following four groups based on the presence of dysglycemia and history of diabetes medication: normal, prediabetes, newly diagnosed T2DM (new-T2DM), and T2DM (claim history for antidiabetic medication). The endpoint was new-onset AC during follow-up. The incidence rates (IRs) in 1,000 person-years and hazard ratios (HRs) of AC for each group were analyzed using Cox proportional hazard regression models. Results The IRs of AC were 9.453 (normal), 11.912 (prediabetes), 14.933 (new-T2DM), and 24.3761 (T2DM). The adjusted HRs of AC in the prediabetes, new-T2DM, and T2DM groups were 1.084 (95% confidence interval [CI], 1.075 to 1.094), 1.312 (95% CI, 1.287 to 1.337), and 1.473 (95% CI, 1.452 to 1.494) compared to the normal group, respectively. This secular trend of the HRs of AC according to T2DM status was statistically significant (P<0.0001). Conclusion This large-scale, longitudinal, nationwide, population-based cohort study of 3,471,745 subjects confirmed that the risk of AC increases in prediabetic subjects and is associated with T2DM status.
... Type 1 diabetes (T1D) results from the destruction of insulin-secreting islet cells by an immune-mediated process, which may be induced or promoted by genetic or environmental factors [1]. ...
Article
Full-text available
Type 1 diabetes (T1D) is typically diagnosed in young people; however, it can appear at any age. Its incidence in adulthood is not as well-known as in childhood, particularly if it is diagnosed in geriatric age. T1D diagnosed in adulthood can be explained by the development of antibodies in adulthood or also by the existence of slow-disease progressors. A 71-year-old normal-weight woman presented to the Emergency Department complaining of polyuria, polydipsia, and tiredness. She was identified with hyperglycemia (450mg/dL) and high blood and urine ketone bodies. Her arterial gasometry revealed mild metabolic ketoacidosis. Further laboratory work-up was remarkable for positive anti-GAD and anti-ICA antibodies and her HbA1c was 14.1%. The diagnosis of T1D was established. A urinary infection was also identified. The patient’s symptoms in association with metabolic ketoacidosis, in the presence of high titers of more than one positive T1D-related antibody, have helped us to diagnose T1D in this elderly woman. A prompt diagnosis enabled us to establish adequate diabetes treatment. The urinary infection was probably a trigger to the symptomatic phase of diabetes. T1D can be diagnosed at any age, even in elderly patients. A prompt T1D diagnosis can avoid the misdiagnosis of type 2 diabetes (T2D), enabling the beginning of correct medication earlier.
Chapter
Nanotechnology has shown immense promise for advancing blood glucose control. This technology offers the potential to safeguard pancreatic cells from autoimmune destruction by driving the creation of innovative therapeutic agents that can be delivered to specific targets. In this book, you will find a comprehensive exploration of diabetes and its approved medical treatments. The book delves into how nanotechnology can amplify the efficacy of current treatment modalities, potentially paving the way for a gene therapy solution to combat this disease. Starting with the history of diabetes treatment, the book explains treatment challenges for diabetes before getting into the three ways nanoscience is helping in diabetes treatment: insulin delivery, drug delivery and nucleotide delivery. Each chapter is contributed by accomplished experts in their respective fields, who strive to offer a thorough, yet accessible discussion of the subject.
Article
Type 1 diabetes mellitus is a condition caused by autoimmune damage to insulin–producing beta cells of the pancreatic islets, leading to endogenous insulin deficiency. Despite the sufficient knowledge of the disease and the availability of clinical recommendations for substitution therapy, the number of patients with this pathology is growing worldwide. At the same time, their cohort is very heterogeneous, including amid different etiology, concomitant genetic background, variations in the manifestation of the disease and severity. In this regard, traditional ideas about type 1 diabetes mellitus are being questioned, which requires special attention when managing patients with a clinical picture of the disease that differs from the traditional one. The article presents a clinical case of type 1 diabetes mellitus in a young patient, which demonstrates the importance of a personalized approach to the diagnosis and treatment of diabetic patients with a "non-classical" history.
Book
Full-text available
ABD, Kanada ve Avrupa’daki diyabet vakalarının %90’ından fazlasından Tip 2 diyabet sorumlu olup; %5-10’undan Tip 1 diyabet, geri kalanından ise özel etiyolojik ve patofizyolojik etkenler sorumludur. Diyabetin bilinen monogenik nedenleri (örn. gençlerin erişkin tipi diyabetine [MODY, mature onset diabetes of the young], ya da neonatal diyabete sebep olan nedenler), vakaların ancak ufak bir kısmını oluşturmaktadır. Tip 1 ve tip 2 diyabetin yaygın formlarının genetik temelleri halen karmaşıktır ve yaygın gen varyantları tek başlarına ancak düşük derecede risk ya da koruma sunmaktadır. Patofizyolojisi kesin olmayıp, tip 1 ve tip 2 kategorilerine net bir biçimde uymayan geniş yelpazede fenotipik diyabet türü ortaya çıkmış ve toplu olarak ‘atipik diyabet’ şeklinde adlandırılmaya başlanmıştır. Dahası, dünya çapındaki fazla kiloluluk ve obezite salgını da tip 2 diyabet patofizyolojisinin diğer türlere göre görünürlüğünü artırmıştır. Bu bölümde, diabetes mellitus sınıflandırması, diyabetin yeni ortaya çıkan formlarının kısa tanımlarıyla beraber gözden geçirilecektir.
Article
Full-text available
Introduction: This study was aimed to assess the prevalence of islet cell autoantibodies (ICAs) in type 2 diabetes mellitus (T2DM) patients and to compare some clinical and metabolic markers between ICA-negative and ICA- positive groups in South Khorasan province, East of Iran. Methods: In this cross-sectional study, 384 T2DM patients, all over 30 years old, were selected. None of the patients needed insulin therapy for at least 6 months following diagnosis. Demographic data including gender, age, age at diagnosis, disease duration, health status, family history of diabetes, body mass index (BMI) and treatment type were collected from all subjects. ICA assays were conducted using commercial enzyme-linked immunosorbent assay (ELISA) kits. In addition, blood pressure, as well as cholesterol, serum C-peptide and HbA1c levels were compared between ICA-negative and ICA-positive groups. Data were analyzed using SPSS software and P < 0.05 was considered as statistically significant. Results: ICA was detected in 1.30% of all diabetic patients. Compared with ICA-negative patients, ICA-positive subjects had significantly lower median age (P = 0.000), lower median disease duration (P = 0.001), lower C-peptide level (P = 0.006) and worse glycaemic control (HbA1c, P = 0.040). Furthermore, autoantibody-positive patients had more need for insulin therapy (P = 0.022). No significant correlation was found between ICA prevalence and family history of diabetes (P = 0.262), mean fasting blood glucose (FBG) (P = 0.282), BMI (P = 0.054), blood pressure (P = 0.070) as well as cholesterol levels (P = 0.888). Conclusion: Autoantibody screening is recommended in T2DM patients, especially non-obese patients under 40 years old, showing worse glycaemic control
Article
Full-text available
To describe the natural course of clinical remission in insulin-dependent diabetes mellitus (IDDM) when insulin dose is minimized without loss of target glycemia and to identify factors that predict clinical remission. Ninety-five patients, who were placebo-treated control subjects in the Canadian-European multicenter randomized trial of cyclosporin A in recent-onset IDDM, were studied. The mean insulin dose decreased during the first months after diagnosis, with a nadir at 3 mo, when 27% of the patients did not require insulin to maintain target glycemia. At 1 yr, 10% of patients still did not need insulin. Patients not receiving insulin who had glycosylated hemoglobin within the normal range were called remitters. Mean basal and glucagon-stimulated C-peptide values were significantly (P less than 0.025) higher in remitters than nonremitters at the start of the study. Therefore, all patients were divided into those with values above the mean stimulated C-peptide (0.4 nM) and those with values below the mean at entry. The probability of entering a remission with a stimulated C-peptide greater than 0.4 nM was 10 times as high (P less than 0.05) as for those with a stimulated C-peptide below this level. Surprisingly, the beginning and end of the remission were associated with neither major changes in C-peptide levels nor islet cell antibody and insulin-antibody titer. A more rapid loss of stimulated C-peptide occurred in patients who lacked HLA-DR3 and -DR4 (P less than 0.05 at mo 9). This study shows a higher spontaneous clinical remission rate than expected during the 1st yr after diagnosis. Preserved beta-cell function at entry predicts a greater chance of entering a remission, and a more rapid loss of beta-cell function was seen in patients without HLA-DR3 and -DR4.
Article
Full-text available
To examine the clinical and immunogenetic heterogeneity of IDDM. We divided 207 IDDM patients into groups based on the interval from clinical onset to initiation of insulin therapy: group A (< 3 mo, acute clinical-onset group, n = 134), group B (3-12 mo, intermediate group, n = 31), and group C (> or = 13 mo, slowly progressive group, n = 42). Immunogenetic and clinical markers were compared between group A and group C. The mode age of onset was higher in group C (52 yr) than group A (10 yr). Group C had a higher prevalence of islet cell antibodies (42.9%, 18 of 42) than group A (25.4%, 34 of 134, P = 0.05). Serum C-peptide immunoreactivity assayed by radioimmunoassay in response to a 100-g oral glucose tolerance test was significantly higher in group C than in group A. Group C patients were also more likely to have a family history of NIDDM (26.1%, 11 of 42) among their first-degree relatives than group A patients (11.2%, 15 of 134, P = 0.039). The prevalences of family history of IDDM and endocrine autoimmune diseases were not different between groups C and A. The frequency of complications of endocrine autoimmune disease was not different between group A (6.7%, 9 of 134) and group C (2.3%, 1 of 42). Significant associations with two class I major histocompatibility complex antigens (HLA-A24 and -Bw54) and one class II antigen (HLA-DR4) were observed in group A. Group A patients were associated with three diabetogenic HLA-DQ haplotypes including DQA1*0301-DQB1*0401, DQA1*0301-DQB1*0302, and DQA1*0301-DQB1*0303. In contrast, group C lacked the association with class I antigens, although HLA-DR4 and HLA-DQA1*0301-DQB1*0401 were more common in this group than in control subjects. These results indicate that the clinical subtype with slowly progressive course (slowly progressive IDDM) has distinct findings including late-age onset, high prevalence of islet cell antibodies, preserved beta-cell function, and high family history of NIDDM. An additive effect of class I and class II major histocompatibility complex antigens is suggested as an explanation for the acute clinical manifestations and more severe beta-cell destruction in group A patients.
Article
We studied the pattern of type 1 diabetes-associated autoantibodies during pregnancy and the transplacental transfer of these autoantibodies to the fetal circulation and searched for possible signs of prenatal induction of β-cell autoimmunity in newborn infants. The population comprised 208 mothers and their newborn infants. Seventy-four of the mothers (36%) had type 1 diabetes and 134 (64%) of the infants had an affected father or sibling. Blood samples were obtained from the mother at the end of the first trimester and at delivery, and from the cord blood of the newborn infant. Close to 40% of the mothers with type 1 diabetes had antibodies to islet cells (ICA), 55% to glutamic acid decarboxylase (GADA) and 54% to the IA-2 protein (IA-2A) in early pregnancy, whereas the corresponding frequencies in the nonaffected mothers were 5·2%, 5·2% and 3·0%. No significant changes could be seen in autoantibody levels during pregnancy, and there was a close correlation between the two maternal samples. One third of the infants of mothers with type 1 diabetes tested positive for ICA, 50% for GADA and 51% for IA-2A. Six percent of the infants of nondiabetic mothers had ICA, 2·2% GADA and none had IA-2A. None of the infants of the antibody negative mothers had antibodies in their cord blood. These observations indicate that the immunomodulatory effect of pregnancy on signs of β-cell autoimmunity is weak, but if diabetes-associated autoantibodies are present in the mother, most of them are transferred to the fetal circulation. Our data do not provide any support for fetal induction of β-cell autoimmunity.
Article
The age at diagnosis of insulin-dependent diabetes mellitus (type I DM) varies between childhood and adulthood. The aim of this study was to define the immunologic and metabolic characteristics of the disease according to the age at which it is diagnosed. We evaluated the residual β-cell function (basal and stimulated C-peptide) and frequency of two major islet cell—related autoantibodies, glutamic acid decarboxylase (GAD) and tyrosine phosphatase-like molecule (IA-2ic), at the onset of type I DM. A population-based study was performed with 235 consecutive cases of recent-onset (<4 weeks) type I DM (ages 5 to 45 years) diagnosed in the Lazio region of central Italy. Five age groups were considered: patients diagnosed between ages 5 and 7 years (n = 10), 7 and 10 years (n = 38), 10 and 17 years (n = 94), 17 and 20 years (n = 17), and 20 and 45 years (n = 76). Patients diagnosed before puberty had significantly reduced C-peptide secretion compared with patients diagnosed at a later age (P < .02). Glycosylated hemoglobin (HbA1c) did not differ at diagnosis between the different age groups. Patients diagnosed at puberty or after required significantly less insulin compared with younger patients (P < .04). GAD antibodies were found in 65% and IA-2ic antibodies in 59% of patients. GAD antibodies tended to be more frequent in patients diagnosed after age 17 compared with younger patients (P = .05), while IA-2ic antibodies were not age-related. These data suggest that (1) the extent of β-cell damage differs between patients diagnosed before and after puberty, the process being more destructive in children less than 7 years of age, when C-peptide levels are the lowest; and (2) residual β-cell function at diagnosis is not influenced by the presence or absence of islet cell—related antibodies. These findings have implications for trials in type I DM diagnosis aimed at protecting β cells from end-stage destruction and in attempts to prevent the disease in susceptible individuals.
Article
To examine whether children of families moving from an area of low incidence of childhood diabetes to one which is higher show a corresponding rise in disease incidence. Disease incidence study over 12 years. Bradford District Metropolitan Council area. All subjects aged 0-16 years resident within the study area. The incidences of childhood diabetes in Asian and non-Asian families. The incidence of diabetes in Asian children increased from 3.1/100,000 per year in 1978-81 to 11.7/100,000 per year in 1988-90 (chi 2 for trend = 4.95, df = 1, p = 0.026) whereas that for other children remained constant at 10.5/100,000 per year. Over the entire study period rates were lower in Asian females (4.9/100,000 per year) than in Asian males (8.8/100,000 per year) whereas the reverse was true for other children (males 9.2/100,000 per year; females 12.0/100,000 per year) (test for common odds ratio: chi 2 = 3.81, df = 1, p = 0.052). Offspring of this transmigratory population had a rising incidence of childhood diabetes which was approaching that of the indigenous population. The data provide strong evidence for an environmental effect in the aetiology of insulin dependent diabetes.
Article
The frequency of antibodies to glutamic acid decarboxylase (GAD) and insulin (IAA) in presymptomatic Type 1 diabetes mellitus with a positive test for antibodies to islet cell antigen (ICA) was examined. Thirty-two persons positive for ICA (> 10 JDF units) were tested 2 to 48 months before their ascertained onset of Type 1 diabetes. ICA was quantitated by immunofluorescence as JDF units, anti-GAD by radioimmunoprecipitation and anti-insulin by radioimmunoassay. There was a positive test for anti-GAD in 25 (78%), and for IAA in 23 (72%), of the 32 prediabetic ICA-positive subjects. Stratification according to age at the onset of diabetes showed differing frequencies of anti-GAD and IAA in the prediabetic stage. Thus the positivity rate for anti-GAD for 18 subjects older than 10 years at onset of diabetes was 83%, and for 14 aged 10 or younger at onset was 71%; conversely, the rate for IAA for 18 subjects older than 10 at onset was 56% and for 14 aged 10 or less at time of onset was 93% (p = 0.01). The frequency of anti-GAD was higher in females (88%) than males (71%) whereas the frequency of IAA was higher in males (82%) than in females (60%). Since autoantibodies to GAD and insulin occur in presymptomatic Type 1 diabetes with differences in frequencies by age and gender, the stimuli to autoimmunity may operate differently at different ages, and may also be gender-related.
Article
Demographic and biological data were collected from all Caucasian Type 1 diabetic patients (n = 279) who were recruited at clinical onset by the Belgian Diabetes Registry over 34 months. The male/female ratio was significantly higher for onset between age 20 and 40 years (2.4) than before age 20 years (1.0); no age-or sex-differences were noticed in serum fructosamine concentration. Total and high concentrations of insulin autoantibodies and islet cell antibodies were preferentially associated with the HLA DQA1*0301-DQB1*0302 susceptibility haplotype. The occurrence of both types of antibodies was also correlated, irrespective of haplotype. At onset before age 10 years, the high risk genotype DQA1*0301-DQB1*0302/DQA1*0501-DQB1*0201 was more prevalent than all other DQA1-DQB1 genotypes taken together, leading to a higher prevalence of the DQA1*0301-DQB1*0302 haplotype in this age group (75%) than in the 10-39 years age group (54%). Under age 10 years, the presence of DQA1*0301-DQB1*0302 was strongly associated with insulin autoantibodies (90%) and islet cell autoantibodies (92% with 85% of high titre), whereas patients without this haplotype were less frequently positive for insulin autoantibodies (31%) or islet cell autoantibodies (38% high titre). In the group with onset at age 10-39 years, the DQA1*0301-DQB1*0302 haplotype presented a lower association with insulin autoantibodies (approximately 40%) and islet cell autoantibodies (50 to 65% high titre), prevalences which no longer differed from those in subjects lacking this haplotype.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
To determine the effects of islet cell antibodies on beta-cell function during the first 3 yr after diagnosis in type II diabetic patients. beta-cell function in type II diabetic patients with (n = 11, 50 +/- 5 yr of age) and without (n = 10, 52 +/- 4 yr of age) ICA was followed prospectively and compared with beta-cell function in type I adult diabetic patients (n = 17, 37 +/- 5 yr of age) and in healthy control subjects (n = 34, age 45 +/- 3 yr). beta-cell function was evaluated as fasting C-peptide, 1 + 3 min C-peptide after intravenous glucose, and delta C-peptide after glucagon. Fasting C-peptide was equal in type II diabetic patients with ICA (0.30 +/- 0.03 nM) and type I diabetic patients (0.24 +/- 0.03 nM) at diagnosis, and decreased (P < 0.05) during 3 yr in these groups but not in type II diabetic patients without ICA. At diagnosis, type II diabetic patients with ICA showed a 1 + 3 min C-peptide (0.92 +/- 0.17 nM) lower (P < 0.001) than control subjects but higher (P < 0.05) than type I diabetic patients (0.53 +/- 0.11 nM). After 1 yr, 1 + 3 min C-peptide in type II diabetic patients with ICA had decreased (P < 0.05) to 0.18 +/- 0.11 nM and was equal to type I diabetic patients (0.38 +/- 0.10 nM). delta C-peptide after glucagon was equally impaired in type II diabetic patients with ICA (0.38 +/- 0.06 nM) and type I diabetic patients (0.35 +/- 0.11 nM) at diagnosis. After 3 yr, type II diabetic patients with ICA had fasting C-peptide of 0.09 +/- 0.04 nM, 1 + 3 min C-peptide of 0.18 +/- 0.10 nM, and delta C-peptide after glucagon of 0.20 +/- 0.09 nM, values equal to type I diabetic patients but lower (P < 0.01) than in type II diabetic patients without ICA, whose values remained unchanged; fasting C-peptide of 0.97 +/- 0.17 nM, 1 + 3 min C-peptide of 2.31 +/- 0.50 nM, and delta C-peptide after glucagon of 1.76 +/- 0.28 nM. In patients considered type II diabetic with ICA, beta-cell function progressively decreased after diagnosis, and after 3 yr was similar to type I diabetic patients, whereas beta-cell function in type II diabetic patients without ICA was unchanged.