Advanced Glycation End Products and Biomarkers

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

REVIEWS

Circulating microRNAs as novel biomarkers


for diabetes mellitus
Claudiane Guay and Romano Regazzi
Abstract | Diabetes mellitus is characterized by insulin secretion from pancreatic β cells that is insufficient
to maintain blood glucose homeostasis. Autoimmune destruction of β cells results in type 1 diabetes mellitus,
whereas conditions that reduce insulin sensitivity and negatively affect β‑cell activities result in type 2 diabetes
mellitus. Without proper management, patients with diabetes mellitus develop serious complications that
reduce their quality of life and life expectancy. Biomarkers for early detection of the disease and identification
of individuals at risk of developing complications would greatly improve the care of these patients. Small non-
coding RNAs called microRNAs (miRNAs) control gene expression and participate in many physiopathological
processes. Hundreds of miRNAs are actively or passively released in the circulation and can be used to
evaluate health status and disease progression. Both type 1 diabetes mellitus and type 2 diabetes mellitus
are associated with distinct modifications in the profile of miRNAs in the blood, which are sometimes
detectable several years before the disease manifests. Moreover, circulating levels of certain miRNAs seem
to be predictive of long-term complications. Technical and scientific obstacles still exist that need to be
overcome, but circulating miRNAs might soon become part of the diagnostic arsenal to identify individuals
at risk of developing diabetes mellitus and its devastating complications.
Guay, C. & Regazzi, R. Nat. Rev. Endocrinol. 9, 513–521 (2013); published online 30 April 2013; doi:10.1038/nrendo.2013.86

Introduction
Diabetes mellitus affects >350 million people worldwide childhood or in young adults and accounts for 5–8% of
and makes a considerable contribution to morbidity and all cases of diabetes mellitus.4 The majority of the other
mortality globally.1 In industrialized countries, diabetes cases are attributable to T2DM. The pathogenesis of
mellitus is the leading cause of blindness, renal failure this disease is closely linked to genetic, environmental
and nontraumatic lower limb amputations. Patients and/or lifestyle factors, such as hypercaloric nutrition,
with diabetes mellitus also have an increased risk of lack of exercise and obesity. T2DM occurs when target
develop­ing cardiovascular disorders and having a stroke, tissues lose insulin sensitivity, including the liver, skeletal
which means that this disease is a heavy socioeconomic muscles and adipose tissues. This state of insulin resist‑
burden.2 Unfortunately, the prevalence of diabetes mel‑ ance can usually be compensated for by expansion of
litus is increasing at a dramatic pace both in children and the functional β‑cell mass and by an increase in insulin
in adults as a result of changes in lifestyle (reduced physi‑ secretion. However, in individuals who are genetically
cal activity and increased overnutrition and obesity), predisposed to develop T2DM, the β cells are unable to
but also as a consequence of population ageing. Indeed, sustain the increased demand for insulin, which leads
according to estimates from the International Diabetes to chronic hyperglycaemia and the onset of T2DM.5
Federation, 552 million people are expected to have Despite intensive research, the causes of T1DM and
d­iabetes mellitus in 2030.1 T2DM remain incompletely understood and a definitive
This Review focuses on type 1 diabetes mellitus cure is still not available. The efficacy of the current treat‑
(T1DM) and type 2 diabetes mellitus (T2DM), the two ments to delay progression of diabetes mellitus would
principal forms of the disease. T1DM is an autoimmune be drastically improved if they could be implemented
disorder in which pancreatic β cells are attacked and during the initial phases of the disease and targeted at
eliminated by the immune system. During the immune individuals with the highest probability of benefitting
response, leucocytes infiltrating the pancreatic islets from the therapeutic intervention. This goal can only
secrete proinflammatory cytokines that recruit cyto‑ be achieved by identifying new biomarkers for predict‑ University of Lausanne,
Department of
toxic T lymphocytes and contribute to β‑cell dysfunction ing and/or monitoring the progression of T1DM and Fundamental
and death.3 The immune reaction leads to progressive T2DM and their long-term complications. The aim of Neurosciences, Rue
du Bugnon 9, 1005
destruction of β cells, which results in severe or com‑ this Review is to summarize the weaknesses of the blood
Lausanne, Switzerland
plete insulin deficiency. T1DM generally develops during parameters and biomarkers that are currently used to (C. Guay, R. Regazzi).
detect people at risk of developing T1DM and T2DM and
Correspondence to:
Competing interests to discuss the potential use of circulating m­icroRNAs R. Regazzi
The authors declare no competing interests. (miRNAs) as a novel class of biomarkers. romano.regazzi@unil.ch

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 9  |  SEPTEMBER 2013  |  513


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points which provides a potentially long period of time in which


to identify and treat individuals at risk. In the past
■■ New biomarkers are needed to improve the identification of individuals at risk
2 years, progress has been made to preserve the function
of developing diabetes mellitus and its associated complications, monitor
disease progression and assess the efficacy of therapeutic interventions of residual β cells at the onset of T1DM using immuno­
■■ Circulating microRNAs (miRNAs) are attractive biomarker candidates as they suppressive medications.8,9 However, the efficacy of these
can be easily collected, are stable under different storage conditions and can treatments is currently limited, although considerable
be measured using assays that are specific, sensitive and reproducible improvements could probably be made if therapies could
■■ Pioneering studies have identified characteristic changes in blood levels of be initiated at earlier stages of the disease when many
miRNAs in samples from a range of cohorts of patients with diabetes mellitus β cells are still present.
■■ However, definitive miRNA signatures for type 1 diabetes mellitus, type 2 diabetes
Autoantibodies against islet antigens are often used
mellitus or their associated complications remain to be defined and agreed upon
■■ Although measuring circulating miRNAs is a promising approach in individuals
as biomarkers for T1DM, as their presence in the blood
at risk of developing diabetes mellitus, several key issues still need to is characteristic of the disease. Several autoanti­bodies
be addressed, including the determination of the most appropriate blood have been described, but those directed against islet
sampling protocols cells, insulin, tyrosine phosphatase IA‑2 and IA‑2β,
glutamate decarboxylase and zinc transporter 8 are the
most reliable for identifying individuals at risk of devel‑
Box 1 | Blood parameters and biomarkers for diabetes oping T1DM (Box 1).7,10 However, the use of islet auto­
T1DM and T2DM antibodies as biomarkers has some important limitations.
Blood parameters Firstly, autoantibodies appear fairly late in the course
■■ Glycaemia of T1DM, so cannot be used to initiate treatment early
■■ HbA1c
in the disease course. Secondly, although most indivi­
T1DM duals at the onset of T1DM are positive for at least some
Autoantigens
autoantibodie­s, many autoantibody-positive individuals
■■ Glutamate decarboxylase (GADA)
■■ Insulin (IAA)
will never develop the disease. Thirdly, autoantibodies are
■■ Islet cells (ICA) not suitable for monitor­ing therapeutic outcomes because
■■ Tyrosine phosphatases (IA‑2 and IA‑2β) they are not immediately removed from the circulation
■■ Zinc transporter 8 (ZnT8) if the autoimmune reaction is stopped. 10 Additional
T2DM biomarkers for T1DM are therefore needed to comple‑
Traditional biomarkers ment the information obtained from the presence of
■■ Cholesterol* autoantibodie­s and other risk factors (such as age, family
■■ Creatinine* history, susceptibilit­y genes and environmental triggers).
■■ Free fatty acids (FAA)*
■■ α-hydroxybutyrate (α-HB)
Biomarkers for T2DM
■■ Lipoproteins (HDL)*
■■ Triacylglycerol (Tg)*
Individuals at risk of developing T2DM are currently
Novel biomarkers identified by a combination of easily accessible serum
■■ Adipokines* parameters (including levels of glucose, triacylglycerol,
■■ C-reactive protein (CRP)* cholesterol, lipoproteins and HbA1c), physical charac­
■■ Ferritin* teristics (BMI, waist-to-hip ratio, blood pressure and
■■ Incretins (e.g. glucagon-like peptide 1) sex) and lifestyle factors (food consumption, physical
■■ Linoleoylglycero-phosphocholine (L-GPC)* inactivity and smoking). By combining all these classic
*These biomarkers are not specific to T2DM but are also
predictive of other metabolic disorders. Abbreviations: T1DM,
biomarkers and risk factors, the probability of predict‑
type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus. ing the development of the disease ranges from 0.85 to
0.90 in a period of 5–10 years before the onset of T2DM.11
Other molecules have emerged as potentially useful bio­
Classic diabetes mellitus biomarkers markers (Box 1), including incretins such as g­lucagon-like
According to the WHO, a diagnosis of diabetes peptide 1, cytokines, adipokines, ferritin and C‑reactive
mellitu­s should be based on measurements of blood protein.12 Individually, none of these so-called novel
levels of glucose in the fasted state and following an biomarkers can predict the manifestation of T2DM effi‑
oral glucose tolerance test.6 A diabetic state is defined ciently, but in combination they can achieve predictive
by glucose levels >7.0 mmol/l (126 mg/dl) in the fasted values similar to those possible with classic biomarkers.13
state and >11.1 mmol/l (200 mg/dl) after an oral glucose All these serum parameters can predict the develop‑
tolerance test.6 Other serum parameters such as levels of ment of T2DM a few years in advance of disease mani‑
HbA1c or residual C‑peptide can also be helpful in the festation in individuals already displaying metabolic
diagnosis of diabetes mellitus. alterations. However, the biomarkers are not specific for
diabetes mellitus and cannot be used to assess disease
Biomarkers for T1DM susceptibility in the general population. Genotype analy‑
T1DM is generally diagnosed when >80–90% of the sis could potentially complement the use of these bio‑
pancreatic β cells have been destroyed by the immune markers for the identification of individuals susceptible
system.7 The progression of the disease is slow (it can take to developing T2DM later in life. However, the predic‑
months to years for the patient to become sympto­matic), tive values of genotypic traits has yet to exceed 0.60. 12

514  |  SEPTEMBER 2013  |  VOLUME 9  www.nature.com/nrendo


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Thus, early and lifestyle-independent predictive factors Box 2 | MicroRNA biogenesis


are currently required to enable physicians to recognize
MicroRNAs (miRNAs) are generated from intergenic genomic sequences or
individuals at risk of developing T2DM. from intronic regions of protein-coding genes. Precursors of miRNAs generated from
intronic regions of protein-coding genes are therefore cotranscribed with their
miRNAs hosting gene.91 Most mammalian miRNAs are transcribed by RNA polymerase II
miRNAs are small non-coding RNA molecules of 21–23 as long precursor molecules that contain a characteristic stem-loop structure.92
nucleotides that function as translational repressors by par‑ These primary transcripts are cleaved by the ribonuclease III enzyme Drosha to
tially pairing to the 3' untranslated region of target mRNAs produce hairpin-structured pre-miRNAs of ~70 nucleotides. These molecules are
(Box 2). These regulators of gene expression were first dis‑ then transported via an Exportin‑5-dependent process into the cytoplasm, where
they are further cleaved by the endoribonuclease Dicer to generate imperfect
covered in Caenorhabditis elegans,14,15 and then in verte‑
duplexes of ~22 nucleotides consisting of a guide strand (miRNA) and a passenger
brates and plants.16 According to the latest estimates, the strand (miRNA*). The guide miRNA strand is incorporated together with Argonaute
human genome encodes >1,600 miRNA precursors, gener‑ proteins into the RNA-induced silencing complex (RISC). The passenger miRNA*
ating up to 2,237 mature miRNAs,17 each of which has the strand is usually degraded but can sometimes also be loaded into the RISC
potential to control hundreds of targets. Now, miRNAs are complex and be functional. The majority of the miRNAs are generated by this
universally recognized as major regulators of gene expres‑ canonical pathway, but alternative pathways have now been described.93,94
sion and key controllers of several biologic and pathologic Mature miRNAs exert their action by guiding the RISC complex to complementary
processes.18 They are produced from stem-loop precursor sequences within the 3' untranslated region of target mRNAs, leading to
translational repression and/or transcript degradation.16 Target recognition is
RNAs that are generated from independent transcriptional
mainly determined by miRNA complementarity with the so-called seed sequences
units or from introns of genes that encode proteins (Box 2). (that is, residues 2–8) of target mRNAs.16,95 Interestingly, a single miRNA can
These primary transcripts (pri-miRNAs) are initially pro‑ potentially bind and regulate the expression of hundreds of targets, whereas
cessed to produce short RNA molecules (pre-­miRNAs) a single 3' untranslated region can be targeted by numerous different miRNAs.95
and are then exported to the cytosol where they are
further cleaved to generate the mature forms of miRNAs
(Figure 1). The mature miRNAs can either be included in Drosha
the RNA-induced silencing complex to guide translational 1 2
Exportin-5
repression of target mRNAs or be released by the cells.
If they are to be released, the miRNAs become attached to pri-miRNA pre-miRNA
Dicer
proteins or lipoproteins or are loaded inside vesicles that 3
Nucleus
are released into the extracellular space during plasma
membrane blebbing or after fusion of m­ulti­vesicular bodies Multivesicular bodies

with the plasma membrane (Figure 1). RISC


4 8
Role of miRNAs in diabetes pathogenesis
Pancreatic β cells and the tissues targeted by insulin mRNA target miRNA
express a well-defined set of miRNAs. Most of the
miRNAs are not cell-specific, but are widely distributed
throughout the tissues of the human body. A notable 5 6 7
exception is miR‑375, a miRNA highly enriched in
pancreatic islets that regulates the expression of genes
involved in hormone secretion and in β‑cell mass expan‑ Argonaute-2
sion in response to insulin resistance.19,20 The miRNA HDL

expression profile of β cells and tissues targeted by insulin


Protein complex Lipoproteins Microvesicles Exosomes
is altered in patients with T1DM and T2DM, which prob‑
ably contributes to the impaired function of these tissues Figure 1 | Biogenesis and release of miRNAs. Pre-miRNAs are generated in the
under disease states.21–23 Indeed, the islets of prediabetic nucleus by the ribonuclease III enzyme Drosha after cleavage of pri-miRNAs (1).
nonobese diabetic (NOD) mice, a model of T1DM, The pre-miRNAs are then transported in the cytoplasm through a process involving
contain increased levels of several miRNAs, including Exportin‑5 and the GTP-binding protein Ran (2) and further cleaved by Dicer to yield
miR‑21, miR‑34a, miR‑29 and miR‑146a, which have 21–23 nucleotide duplexes (3). One strand of the miRNA duplex can either
deleterious effects on β‑cell function.24,25 The expres‑ associate to the RISC complex and guide translational repression of target mRNAs
(4) or be released by the cells. In the latter case, the mature miRNA binds to
sion of most of these miRNAs, as well as that of many
RNA‑binding proteins such as Argonaute‑2 (5) or to lipoproteins (6). Alternatively,
other miRNAs, is also altered in the islets of ob/ob and the miRNAs can be loaded in microvesicles formed by plasma membrane blebbing
db/db mice, which are models of obesity and T2DM.26,27 (7) or in exosomes that are released in the extracellular space upon exocytic
Interestingly, the expression of miR‑29 and miR‑34a is fusion of multivesicular bodies with the plasma membrane (8). Abbreviations:
also increased in tissues targeted by insulin in these mouse miRNA, microRNA; pre-miRNA, miRNA precursor; pri-miRNA, primary miRNA
models, which possibly contributes to insulin resist‑ transcript; RISC, RNA-induced silencing complex.
ance.28,29 Other miRNAs that are dysregulated in tissues
targeted by insulin in ob/ob mice, dietary mouse models of a contribution of these miRNAs to the developmen­t of
obesity and diabetic Goto-Kakizaki rats include miR‑143, insulin resistance in these obesity models.29–31
miR‑802 and two closely related miRNAs, miR‑103 and Changes in the miRNA profile that are related to diabete­s
miR‑107.28–31 Strong experimental evidence indicates mellitus have also been reported in human tissues. More

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 9  |  SEPTEMBER 2013  |  515


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

miRNAs are involved in the early phases of the disease


RNA-binding protein process. The expression of some of these miRNAs is con‑
Lipoprotein trolled by insulin, but this regulatory mechanism seems to
Exosome Blood be impaired in patients with diabetes mellitus.33
vessel
As well as the changes in tissues targeted by insulin that
are described above, diabetes mellitus results in consider‑
able modifications in miRNA expression in blood vessels,
heart, retina and kidneys. This finding indicates that
these non-coding RNAs are involved in the development
of long-term complications of diabetes mellitus.22,34,35

A functional role for circulating miRNAs?


In addition to regulating gene expression inside the
cells that produce them, several miRNAs are found in
1 blood and other body fluids in association with proteins,
HDL microvesicles and/or lipoprotein complexes (Figure 2).36–38
The function of circulating miRNAs remains to be estab‑
lished, but in vitro studies indicate that miRNAs trans‑
Receiving cells Fusion ported by exosomes (Box 3) or HDL can be transferred
Receptors in an active form to recipient cells.38,39 This observation
Endocytosis 2 raises the intriguing possibility of an involvement of
miRNAs in a novel cell-to-cell communication mode.
Circulating miRNAs are very stable and resistant to treat‑
Figure 2 | Blood and other body fluids contain active miRNAs. miRNAs can ment with ribonucleases, freezing/thawing cycles and
be released or shed by cells and are found in a stable form in body fluids. other drastic experimental conditions.40,41 Consequently,
The miRNAs present in the blood are associated with protein complexes such serum or plasma samples can be stored at –20 °C or
as Argonaute‑2 or with HDL particles, or are transported inside membrane-bound
–80 °C for up to several months without notable degra‑
vesicles such as exosomes (1). Evidence suggests that circulating miRNAs can be
taken up in active form through different mechanisms, including receptor-mediated dation of miRNAs,42 which suggests that these small RNA
capture, endocytosis or fusion of exosomes with the plasma membrane of molecules are sufficiently robust to serve as biomarkers.
receiving cells (2). Transfer of miRNAs between distantly located cells constitutes Circulating miRNAs have several other advantages as
a potentially new communication mode. Abbreviation: miRNA, microRNA. potential biomarkers: they are found not only in blood
but also in other easily accessible biologic fluids (such as
Box 3 | Exosomes urine, saliva, amniotic fluid and breast milk),43 they can be
detected by highly sensitive and specific quantitative real-
Exosomes are microvesicles of 30–120 nm that were first observed to be
released by reticulocytes in the 1980s.96–98 Since then, exosomes have been
time PCR, and most of them are evolutionarily conserved,
detected in the culture media of most cell types and are abundant in several which facilitates the translation of results obtained from
body fluids, such as blood, urine, saliva and breast milk.43 The concentration in vivo animal studies to human health care. Moreover,
of exosomes in the blood has been estimated to be ~3 million per μl.99 These profiles of the miRNAs in serum of healthy donors are
vesicles display a characteristic size, form and lipid bilayer structure that can be fairly homogeneous and constant over 24 h and miRNAs
visualized by electron microscopy. Exosome markers, such as the tetraspanins can be measured in both serum and plasma.41,44,45
CD63 and CD81 and the Escort proteins Alix and Tsg101, are also routinely used
to distinguish exosomes from other vesicles secreted by cells. Detailed analysis
miRNAs as diabetes mellitus biomarkers
of the exosome content revealed that they can carry proteins and nucleic acids
(such as mRNAs, microRNAs and long non-coding RNAs).39,100 The interest in The idea of using miRNAs found in blood as bio­markers
exosomes has increased dramatically in the past 5 years, following the discovery is fairly new and was first proposed for detecting various
that their cargo can be transferred in active form to recipient cells,39,101,102 which forms of cancer,41,46,47 autoimmune diseases48 and sepsis.49
led to the exciting concept of exosomes as messengers of a new cell-to-cell Studies have now also analysed the miRNA profile in
communication mode. However, the precise mechanisms leading to exosome serum, plasma or blood cells in an attempt to develop
release and/or uptake remain to be elucidated. Exosomes are produced inside new approaches to predict the development and pro‑
cells via the multivesicular endosomal pathway,97,98 but very little is known about
gression of diabetes mellitus (Table 1). Zampetaki and
the selective packaging of nucleic acids and proteins composing the exosome
colleagues50 were the first to identify a characteristic
cargo. An improved understanding of these mechanisms might reveal new biologic
roles for exosomes in the healthy state, but also in pathophysiological conditions. expression profile of miRNAs in blood that was related
to T2DM. In their prospective study, they analysed blood
samples from >800 individuals randomly selected from
than 60 differentially expressed miRNAs were detected in the Bruneck population (Bolzano Province, Italy) and
human skeletal muscle biopsy samples from patients with identified a subset of five miRNAs (miR‑15a, miR‑28-3p,
T2DM, including miR‑143, which is upregulated, and two miR‑29b, miR‑126 and miR‑223) that displayed a charac­
muscle-specific miRNAs, miR‑206 and miR‑133a, which teristic deregulation in 80 participants with either pre­
are downregulated.32 Interestingly, the levels of about 15% diabetes or T2DM. Importantly, the levels of these
of these miRNAs were already modified in individuals miRNAs were already modified 5–10 years before
with impaired glucose tolerance, which suggests that the the onset of the disease, providing initial evidence for

516  |  SEPTEMBER 2013  |  VOLUME 9  www.nature.com/nrendo


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Blood miRNA changes associated with diabetes mellitus


Study Study design Source Method of analysis miRNAs identified Observations
T2DM
Zampetaki 800 individuals from Plasma Microarray profiling miR‑15a, miR‑28-3p, First study to suggest
et al. the Bruneck cohort (confirmed by qPCR) miR‑29b, miR‑126 and a blood miRNA signature
(2010)50 miR‑223 for T2DM
Kong et al. 56 individuals with Serum qPCR on specific miR‑9, miR‑29a, miR‑30d, Deregulated in T2DM
(2011)51 health conditions miRNAs miR‑34a, miR‑124a,
related to diabetes miR‑146a and miR‑375
mellitus
Karolina 265 individuals with Blood Microarray profiling miR‑150, miR‑192, Correlation between
et al. health conditions (confirmed by qPCR) miR‑27a, miR‑320a raised levels of fasting
(2012)52 related to the and miR‑375 glucose and altered levels
metabolic syndrome of miR‑27a and miR‑320a
T1DM
Nielsen Danish and Hvidoere Serum Microarray profiling miR‑24, miR‑25, miR‑25 negatively
et al. cohorts of newly (confirmed by qPCR) miR‑26a, miR‑27a, correlated with residual
(2012)53 diagnosed children miR‑27b, miR‑29a, β‑cell function
miR-30a-5p, miR‑148a,
miR‑152, miR‑181a,
miR‑200a and miR‑210
Sebastiani 20 patients newly Serum Microarray profiling miR‑9, miR‑31, miR‑34a, Deregulated in T1DM
et al. diagnosed with (confirmed by qPCR) miR‑146a, miR‑155,
(2012)54 T1DM miR‑181a and miR‑199a
Salas-Perez 20 patients with PBMCs qPCR on specific miR‑21a and miR‑93 Underexpressed in T1DM
et al. T1DM miRNAs
(2012)56
Sebastiani 19 patients with Lymphocytes qPCR on specific miR‑326 Correlation with islet
et al. T1DM miRNAs autoimmune attack
(2011)57
Abbreviations: T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; miRNA, microRNA; PBMC, peripheral blood mononuclear cell; qPCR, quantitative
real-time PCR.

the usefulness of circulating miRNAs as early predictors in T1DM. Nielsen and colleagues compared two cohorts
of T2DM and its vascular complications. (Danish and Hvidoere) of patients newly diagnosed with
The miRNA content of serum from patients with pre‑ T1DM with an age-matched control group. 53 Global
diabetes and/or who are newly diagnosed with T2DM miRNA sequencing, followed by quantitative real-time
has also been analysed by other groups. Kong and co- PCR verification and regression analysis to adjust for age,
workers51 detected an increase in the expression of seven sex and multiple testing, highlighted a group of miRNAs
diabetes-related miRNAs (miR‑9, miR‑29a, miR‑30d, (miR‑24, miR‑25, miR‑26a, miR‑27a, miR‑27b, miR‑29a,
miR‑34a, miR‑124a, miR‑146a and miR‑375) in patients miR‑30a-5p, miR‑148a, miR‑152, miR‑181a, miR‑200a
with T2DM compared with patients who had prediabetes and miR‑210) that were differentially expressed in cohorts
or were susceptible to T2DM. However, no differences of patients with T1DM compared with control groups.
were observed between individuals with normal glucose Several of these miRNAs modulate the expression of genes
tolerance and those with prediabetes, which indicates involved in apoptosis and/or important β‑cell regulatory
that the level of these miRNAs in serum is not suitable networks.53 Moreover, levels of miR‑25 were found to cor‑
for predicting susceptibility to T2DM. In a study pub‑ relate with residual β‑cell function (determined by levels
lished in 2012, Karolina et al.52 measured the miRNAs of C‑peptide) and adequate glycaemic control (measured
present in the blood and exosomes of 265 patients with by levels of HbA1c) 3 months after disease onset in the
different health conditions associated with the metabolic Danish cohort. However, this correlation was not observed
syndrome. They detected an upregulation of miR‑27a, in the Hvidoere cohort, in which glycaemic control was
miR‑150, miR‑192, miR‑320a and miR‑375 in patients evaluated 12 months after the diagnosis of T1DM, possibly
with T2DM and observed a strong correlation between because of the loss of residual β cells.
raised fasting levels of glucose and the increase in levels In a study presented at the 2012 meeting of the European
of miR‑27a and miR‑320a. These pioneering studies Association for the Study of Diabetes, Sebastiani and co-
demonstrate the potential of miRNAs as biomarkers for workers54 compared the profile of miRNAs in the blood
T2DM. However, the heterogeneity of the results obtained of 20 patients newly diagnosed with T1DM with that of
underscores the need for large prospective studies to healthy control individuals. Of 206 miRNAs detected in
i­dentify reliable miRNA signatures for diagnosing T2DM. the serum of both groups, 64 were found to be differently
A similar approach was used to identify new bio­markers expressed in the patients with T1DM. Interestingly, some
to predict destruction or regeneration of re­sidual β cells of these miRNAs regulate the functions of immune cells

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 9  |  SEPTEMBER 2013  |  517


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

(miR‑31, miR‑146a, miR‑155, miR‑181a and miR‑199a) diseases. Interestingly, another study also reported down‑
or of β cells (miR‑9 and miR‑34a). A miRNA abundantly regulation of miR‑126 in blood samples obtained from
expressed in the islets of Langerhans, miR‑375, has been patients with coronary artery disease.59 This miRNA is
proposed as a suitable biomarker to detect β‑cell death and highly enriched in endothelial cells, where it has important
to predict the development of T1DM in animal models.55 roles in cell homeostasis and vascular integrity in differ‑
Indeed, massive β‑cell loss elicited by administration of ent animal models.60,61 Moreover, the levels of miR‑126
streptozocin caused a dramatic rise in circulating levels released in apoptotic bodies are reduced by chronic expo‑
of this miRNA.55 Moreover, plasma levels of miR‑375 were sure of endothelial cells to raised blood levels of glucose,50
considerably increased in NOD mice 2 weeks before the which makes this miRNA an ideal candidate biomarker
onset of T1DM.55 The changes in levels of miR‑375 con‑ for monitoring diabetic vascular complications. Another
sequent to β‑cell death were short-lived (<1 week). Thus, miRNA in endothelial cells that deserves further attention
these promising findings obtained in mice will need to is miR‑503. The levels of this miRNA are upregulated in
be verified in humans, as the decline in β‑cell mass takes muscle biopsy samples and in peripheral blood-derived
much longer in humans than in NOD mice. plasma of patients with diabetes mellitus who have limb
Instead of analysing plasma samples, other studies have ischaemia.62 Interestingly, local inhibition of miR‑503
focused their attention on blood cells and measured the in a mouse model of limb ischaemia improved vascula­r
expression of specific miRNAs that are thought to have healing and blood flow recovery. 62 Other circulating
important roles in the immune reaction that leads to T1DM. miRNAs have also been suggested as diagnostic markers
Salas-Pérez et al.56 observed diminished expression of for various cardiovascular diseases,63,64 but their use in
miR‑21a and miR‑93 in peripheral blood mono­nuclear cells predicting or monitoring cardiovascular complications
(PBMC) of patients with T1DM compared with healthy in patients with diabetes mellitus has yet to be investigated.
control individuals. The reduction of miR‑21a (but not Kidney disease affects 30% and 50% of patients with
of miR‑93) expression could be reproduced by incubating T1DM and T2DM, respectively.65–67 Microalbuminuria has
PBMC from control individuals in a medium containing been proposed as a biomarker that could be used to predict
25 mmol/l of glucose, suggesting that the reduced levels of the occurrence of this important complication; however,
miR‑21a might be the consequence of chronic hypergly‑ studies published in the past decade have revealed that
caemia. Finally, Sebastiani and colleagues analysed miRNA a noteworthy proportion of patients with diabetes mellitus
expression in blood lymphocytes from patients with T1DM undergo renal failure before microalbuminuria is detect‑
and observed a rise in levels of miR‑326 that correlated with able, or even before it occurs.68–70 Circulating miRNAs
the timing of the islet autoimmune attack.57 The primary represent a viable alternative for monitoring renal failure
goal of the latter two studies was to identify miRNAs that in patients with diabetes mellitus. They are not elimi‑
could be involved in the development of T1DM. However, nated by haemodialysis71 and have already been tested
as a large proportion of miRNAs in serum are released by in different renal diseases with promising results in both
blood cells, it is possible that the miRNA changes in PBMC animal models and human patients.72–74 Indeed, a corre‑
and/or lymphocytes observed in these two studies might lation was observed between levels of certain circulating
yield detectable differences in plasma levels that would miRNAs, such as miR-16, miR-21, miR-210 and miR-638,
enable the autoimmune reaction to be monitored. and glomerular filtration rate, a well-known parameter of
the progression of kidney disease.72 Large-scale prospec‑
Prediction of diabetes mellitus complications tive studies focusing on patients with diabetes m­ellitus
T1DM and T2DM are both associated with long-term undergoing renal failure will be necessary to identify
microvascular and macrovascular complications that a specific miRNA profile in plasma or urine that can be
can have a devastating effect on quality of life and life used to predict the appearance of this complication. Urine
expectancy. The discovery of biomarkers that could represents an ideal source of miRNAs as it can be easily
be used to identify individuals at risk of experiencing collected noninvasively and in large amounts. Moreover,
serious complications such as retinopathy, nephropathy urinary exosomes originate from various cell types that
or cardiovascular disorders would enable clinicians to span the entire urinary track and would be ideally suited
tailor therapeutic approaches and minimize the expected for use in monitoring the progression of renal diseases.75–78
effects of the disease. However, reliable biomarkers for Indeed, profiles of miRNAs in urine have been reported
these long-term complications are still lacking. to differ across the stages of diabetic nephropathy,79 sug‑
Cardiovascular complications are a major concern in gesting that they could be used as tools to follow the pro‑
this group of patients as they account for up to 80% of gressive alteration of the renal processes in patients with
premature mortality in patients with diabetes mellitus.58 diabetes mellitus.
Prevention, or even a delay, of these complications would To our knowledge, no reports have been published to
be a huge advancement in the treatment of diabetes date about the use of circulating miRNAs to predict the
m­ellitus. As discussed previously, Zampetaki and col‑ occurrence of diabetic retinopathy. However, an involve‑
leagues50 identified a unique plasma miRNA signature in ment of some specific miRNAs, such as miR‑29b and
patients with T2DM. Among the miRNA-related char‑ miR‑200b, in the development of this complication has
acteristic changes observed, reduced levels of miR‑126 been demonstrated.35 These findings might open the door
showed the strongest association with T2DM, and cor‑ to future investigations aimed at assessing the potential
related with the occurrence of subclinical and overt artery use of miRNAs as predictors of this debilitating condition.

518  |  SEPTEMBER 2013  |  VOLUME 9  www.nature.com/nrendo


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Gestational diabetes mellitus urgently needed to facilitate comparisons between differ‑


Circulating miRNAs could in principle also be used ent studies and to reach a consensus about the miRNAs
to identify women at increased risk of developing most suitable to function as early biomarkers of diabetes
­gestational diabetes mellitus. Most screening protocols mellitus. In fact, the level of certain miRNAs, such as
are currently based on a glucose challenge test performed miR‑15b and miR‑16, in plasma is greatly influenced by
around weeks 24–28 of gestation. Therefore, interven‑ the degree of haemolysis and by cellular contamination,
tions such as diet, exercise or medication are sometimes making them less suitable as clinical biomarkers.82
started as late as 32 weeks of gestation. In a multistage Diabetes mellitus is a complex disorder that involves
retrospective study, the miRNAs in serum of women at major metabolic alterations and important adaptations
16–19 weeks of gestation were screened. Three miRNAs in the activity of several organs. miRNAs are being pro‑
(miR‑29a, miR‑122 and miR‑132) were identified that posed as potential biomarkers for an increasing number
were deregulated in women developing gestational dia‑ of diseases. Therefore, it will be essential to determine
betes mellitus before changes in blood levels of glucose whether the detected miRNA changes are exclusively
became detectable.80 Placental-specific miRNAs can also indicative of a prediabetic or diabetic condition or if they
be detected in maternal serum.44,81 Thus, it is possible are also observed in other physiological or pathological
that gestational diabetes mellitus might lead to changes situations,45 such as in response to modifications in the
in the levels of miRNAs in blood that differ from those nutritional state, inflammation, autoimmunity or cancer.
of T1DM or T2DM. At present, the origin of miRNAs in the blood is largely
unknown and their levels do not directly mirror changes
Future directions in pancreatic β cells or tissues targeted by insulin that
The findings described in this article confirm that occur in diabetes mellitus. miRNAs can be actively or
miRNAs are attractive novel biomarkers for diabetes passively released by a variety of cells and can be carried
mellitus. Indeed, changes in the levels of a subset of these by membrane-bound vesicles, protein complexes or lipo‑
small RNA molecules in body fluids promise to provide protein particles (Figures 1 and 2). So far, the majority of
new clues for early identification of individuals at risk of studies have measured levels of miRNAs directly in the
developing diabetes mellitus and the complications asso‑ plasma (or serum). This approach is obviously conveni‑
ciated with this disorder, for following disease progression ent and straightforward. However, modifications in the
and for assessing the efficacy of therapeutic interventions. level of miRNAs originating from specific groups of cells
However, major scientific and technical advances need to that are not in direct contact with the blood or are not
be made before these goals can be achieved. very abundant are unlikely to have a notable effect on
On the basis of the current data, circulating miRNAs the pool of miRNAs in the plasma and will probably go
should be able to be used to substitute or complement undetected. Although technically demanding, protocols
other routine measurements in the future. Thus, their enabling a specific assessment of the miRNAs carried by
efficacy in predicting the occurrence of diabetes m­ellitus exosomes, protein complexes or lipoproteins will proba‑
or its complications needs to be systematically compared bly provide more detailed information about the physio­
with biomarkers that are already available. In particu‑ logical or pathological status of the organs of interest
lar, it will be essential to scrutinize whether miRNAs than measuring serum levels of miRNAs. Furthermore,
are indeed able to provide earlier and/or more precise it might be possible to affinity-purify membrane-bound
detection of individuals at risk of developing the disease vesicles originating from a specific group of cells, taking
or its long-term complications than current biomarkers. advantage of the presence of characteristic proteins on
The situation will hopefully evolve in the future; however, the vesicle surface.12 This approach will be particularly
there is currently no obvious advantage to replacing appropriate for estimating the residual β‑cell mass in
other traditional biomarkers with measurement­s of levels patients newly diagnosed with T1DM. Indeed, except
of c­irculating miRNAs. in the case of a sudden loss of a large number of β‑cells,
Different papers have reported changes in levels of the miRNAs released by insulin-secreting cells represent
miRNAs that are associated with diabetes mellitus or its only a negligible fraction of all non-coding RNAs circu‑
complications but, for various reasons, we are still very lating in the blood. This problem might be partially over‑
far from a consensus about the most relevant miRNAs come by searching for miRNAs that are highly expressed
to be measured. This lack of agreement can in part in β cells, such as miR‑375.19,55 However, the expression
be attributed to the heterogeneity of the approaches of this particular miRNA is modified in a variety of
selected by investigators. Some of the published studies tumorous cells and several authors have suggested using
carried out systematic profiling of a large number of circulating levels of miR‑375 for the diagnosis of differ‑
miRNAs,50,52,53 whereas others focused on a small group ent types of cancer.83–86 Several insulin-secreting cell lines
of selected miRNAs that were supposedly most likely release notable amounts of exosomes87–89 and we have
to be affected in patients with diabetes mellitus.51,55–57 initial evidence indicating that this is also the case for
These approaches yielded a large number of potentially rodent and human islet cells.90 Protocols permitting an
interesting candidate biomarkers, but most of them still enrichment in exosomes derived from islet cells would
await confirmation by independent researchers and/or probably improve the detection of specific changes in
in larger cohorts. Standardized protocols for sample insulin-secreting cells and provide a better evaluation
preparation, RNA extraction and miRNA analysis are of the functional β‑cell mass.

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 9  |  SEPTEMBER 2013  |  519


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Conclusions Review criteria


miRNAs are emerging as important regulators of gene
We systematically searched PubMed using different
expression and central players in many physiological and
combinations of the following words: “diabetes”, “type 1
pathological processes. Their presence in extra­cellular
diabetes”, “type 2 diabetes”, “gestational diabetes”,
fluids in astonishingly stable forms has led to the idea “diabetes complications”, “diabetic retinopathy”,
of using them as biomarkers for a variety of diseases. “diabetic nephropathy”, “cardiovascular complications”,
These properties have also attracted the attention of “biomarkers”, “microRNAs”, “circulating miRNAs”
diabet­ologists who have initiated the search for miRNAs and “exosomes”. We also scrutinized the appropriate
that enable early detection of T1DM and T2DM and references cited in the selected papers. Two abstracts
their associated complications. A number of scientific from the 2012 European Association for the Study of
and methodological issues need to be addressed before Diabetes meeting held in Berlin were also included. Owing
ci­rculating miRNAs can attain the status of biomarkers of to space limitations, not all original articles on the subject
diabetes mellitus, but the available data suggest that they could be listed in the present review. Whenever possible,
will soon serve as valuable new blood parameters that will the most recent and complete reviews on the topic were
chosen. All selected articles are in English.
help physicians to refine their therapeutic interventions.

1. Whiting, D. R., Guariguata, L., Weil, C. & Shaw, J. 16. Bartel, D. P. MicroRNAs: genomics, biogenesis, 32. Gallagher, I. J. et al. Integration of microRNA
IDF diabetes atlas: global estimates of the mechanism, and function. Cell 116, 281–297 changes in vivo identifies novel molecular
prevalence of diabetes for 2011 and 2030. (2004). features of muscle insulin resistance in type 2
Diabetes Res. Clin. Pract. 94, 311–321 (2011). 17. miRBase. The microRNA database [online], diabetes. Genome Med. 2, 9 (2010).
2. Li, R., Zhang, P., Barker, L. E., Chowdhury, F. M. www.mirbase.org (2013). 33. Granjon, A. et al. The microRNA signature
& Zhang, X. Cost-effectiveness of interventions 18. Flynt, A. S. & Lai, E. C. Biological principles of in response to insulin reveals its implication
to prevent and control diabetes mellitus: microRNA-mediated regulation: shared themes in the transcriptional action of insulin in human
a systematic review. Diabetes Care 33, amid diversity. Nat. Rev. Genet. 9, 831–842 skeletal muscle and the role of a sterol
1872–1894 (2010). (2008). regulatory element-binding protein 1c/myocyte
3. Pirot, P., Cardozo, A. K. & Eizirik, D. L. Mediators 19. Poy, M. N. et al. A pancreatic islet-specific enhancer factor 2C pathway. Diabetes 58,
and mechanisms of pancreatic beta-cell death microRNA regulates insulin secretion. 2555–2564 (2009).
in type 1 diabetes. Arq. Bras. Endocrinol. Nature 432, 226–230 (2004). 34. Kantharidis, P., Wang, B., Carew, R. M.
Metabol. 52, 156–165 (2008). 20. Poy, M. N. et al. miR‑375 maintains normal & Lan, H. Y. Diabetes complications: the microRNA
4. American Diabetes Association. Diagnosis and pancreatic α- and β-cell mass. Proc. Natl Acad. perspective. Diabetes 60, 1832–1837 (2011).
classification of diabetes mellitus. Diabetes Care Sci. USA 106, 5813–5818 (2009). 35. Natarajan, R., Putta, S. & Kato, M. MicroRNAs
28 (Suppl. 1), S37–S42 (2005). 21. Kumar, M., Nath, S., Prasad, H. K., Sharma, G. D. and diabetic complications. J. Cardiovasc. Transl.
5. Prentki, M. & Nolan, C. J. Islet β cell failure in & Li, Y. MicroRNAs: a new ray of hope for diabetes Res. 5, 413–422 (2012).
type 2 diabetes. J. Clin. Invest. 116, 1802–1812 mellitus. Protein Cell 3, 726–738 (2012). 36. Arroyo, J. D. et al. Argonaute2 complexes carry a
(2006). 22. Shantikumar, S., Caporali, A. & Emanueli, C. population of circulating microRNAs independent
6. Stumvoll, M., Goldstein, B. J. & van Haeften, T. W. Role of microRNAs in diabetes and its of vesicles in human plasma. Proc. Natl Acad.
Type 2 diabetes: principles of pathogenesis and cardiovascular complications. Cardiovasc. Sci. USA 108, 5003–5008 (2011).
therapy. Lancet 365, 1333–1346 (2005). Res. 93, 583–593 (2012). 37. Gibbings, D. J., Ciaudo, C., Erhardt, M.
7. Winter, W. E., Harris, N. & Schatz, D. Type 1 23. Guay, C., Roggli, E., Nesca, V., Jacovetti, C. & Voinnet, O. Multivesicular bodies associate
diabetes islet autoantibody markers. Diabetes & Regazzi, R. Diabetes mellitus, a microRNA- with components of miRNA effector complexes
Technol. Ther. 4, 817–839 (2002). related disease? Transl. Res. 157, 253–264 and modulate miRNA activity. Nat. Cell Biol. 11,
8. Orban, T. et al. Co-stimulation modulation (2011). 1143–1149 (2009).
with abatacept in patients with recent-onset 24. Roggli, E. et al. Involvement of microRNAs in 38. Vickers, K. C., Palmisano, B. T., Shoucri, B. M.,
type 1 diabetes: a randomised, double-blind, the cytotoxic effects exerted by proinflammatory Shamburek, R. D. & Remaley, A. T. MicroRNAs
placebo-controlled trial. Lancet 378, 412–419 cytokines on pancreatic β-cells. Diabetes 59, are transported in plasma and delivered to
(2011). 978–986 (2010). recipient cells by high-density lipoproteins.
9. Sherry, N. et al. Teplizumab for treatment of 25. Roggli, E. et al. Changes in microRNA expression Nat. Cell Biol. 13, 423–433 (2011).
type 1 diabetes (Protégé study): 1‑year results contribute to pancreatic β-cell dysfunction in 39. Valadi, H. et al. Exosome-mediated transfer of
from a randomised, placebo-controlled trial. prediabetic NOD mice. Diabetes 61, 1742–1751 mRNAs and microRNAs is a novel mechanism
Lancet 378, 487–497 (2011). (2012). of genetic exchange between cells. Nat. Cell Biol.
10. Purohit, S. & She, J. X. Biomarkers for type 1 26. Lovis, P. et al. Alterations in microRNA 9, 654–659 (2007).
diabetes. Int. J. Clin. Exp. Med. 1, 98–116 (2008). expression contribute to fatty acid-induced 40. Kroh, E. M., Parkin, R. K., Mitchell, P. S.
11. Schulze, M. B. et al. Use of multiple metabolic pancreatic β-cell dysfunction. Diabetes 57, & Tewari, M. Analysis of circulating microRNA
and genetic markers to improve the prediction 2728–2736 (2008). biomarkers in plasma and serum using
of type 2 diabetes: the EPIC-Potsdam Study. 27. Zhao, E. et al. Obesity and genetics regulate quantitative reverse transcription-PCR (qRT‑PCR).
Diabetes Care 32, 2116–2119 (2009). microRNAs in islets, liver, and adipose of diabetic Methods 50, 298–301 (2010).
12. Muller, G. Microvesicles/exosomes as potential mice. Mamm. Genome 20, 476–485 (2009). 41. Mitchell, P. S. et al. Circulating microRNAs
novel biomarkers of metabolic diseases. Diabetes 28. Herrera, B. M. et al. Global microRNA as stable blood-based markers for cancer
Metab. Syndr. Obes. 5, 247–282 (2012). expression profiles in insulin target tissues detection. Proc. Natl Acad. Sci. USA 105,
13. Kolberg, J. A. et al. Development of a type 2 in a spontaneous rat model of type 2 diabetes. 10513–10518 (2008).
diabetes risk model from a panel of serum Diabetologia 53, 1099–1109 (2010). 42. Mraz, M., Malinova, K., Mayer, J. & Pospisilova, S.
biomarkers from the Inter99 cohort. Diabetes 29. Trajkovski, M. et al. MicroRNAs 103 and 107 MicroRNA isolation and stability in stored RNA
Care 32, 1207–1212 (2009). regulate insulin sensitivity. Nature 474, samples. Biochem. Biophys. Res. Commun. 390,
14. Lee, R. C., Feinbaum, R. L. & Ambros, V. 649–653 (2011). 1–4 (2009).
The C. elegans heterochronic gene lin‑4 encodes 30. Jordan, S. D. et al. Obesity-induced 43. Weber, J. A. et al. The microRNA spectrum
small RNAs with antisense complementarity overexpression of miRNA‑143 inhibits insulin- in 12 body fluids. Clin. Chem. 56, 1733–1741
to lin‑14. Cell 75, 843–854 (1993). stimulated AKT activation and impairs glucose (2010).
15. Wightman, B., Ha, I. & Ruvkun, G. metabolism. Nat. Cell Biol. 13, 434–446 (2011). 44. Gilad, S. et al. Serum microRNAs are promising
Posttranscriptional regulation of the 31. Kornfeld, J. W. et al. Obesity-induced novel biomarkers. PLoS ONE 3, e3148 (2008).
heterochronic gene lin‑14 by lin‑4 mediates overexpression of miR‑802 impairs glucose 45. Keller, A. et al. Toward the blood-borne miRNome
temporal pattern formation in C. elegans. metabolism through silencing of Hnf1b. Nature of human diseases. Nat. Methods 8, 841–843
Cell 75, 855–862 (1993). 494, 111–115 (2013). (2011).

520  |  SEPTEMBER 2013  |  VOLUME 9  www.nature.com/nrendo


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

46. Chen, X. et al. Characterization of microRNAs 66. Parving, H. H., Lewis, J. B., Ravid, M., prognostic markers in metastatic breast cancer.
in serum: a novel class of biomarkers for Remuzzi, G. & Hunsicker, L. G. Prevalence and Clin. Cancer Res. 18, 5972–5982 (2012).
diagnosis of cancer and other diseases. risk factors for microalbuminuria in a referred 87. Lee, H. S., Jeong, J. & Lee, K. J. Characterization
Cell Res. 18, 997–1006 (2008). cohort of type II diabetic patients: a global of vesicles secreted from insulinoma NIT‑1 cells.
47. Lawrie, C. H. et al. Detection of elevated levels perspective. Kidney Int. 69, 2057–2063 (2006). J. Proteome Res. 8, 2851–2862 (2009).
of tumour-associated microRNAs in serum of 67. Thomas, M. C., Groop, P. H. & Tryggvason, K. 88. Palmisano, G. et al. Characterization of membrane-
patients with diffuse large B‑cell lymphoma. Towards understanding the inherited shed microvesicles from cytokine-stimulated
Br. J. Haematol. 141, 672–675 (2008). susceptibility for nephropathy in diabetes. Curr. β-cells using proteomics strategies. Mol. Cell
48. Alevizos, I. & Illei, G. G. MicroRNAs as Opin. Nephrol. Hypertens. 21, 195–202 (2012). Proteomics 11, 230–243 (2012).
biomarkers in rheumatic diseases. Nat. Rev. 68. Macisaac, R. J. & Jerums, G. Diabetic kidney 89. Sheng, H. et al. Insulinoma-released exosomes
Rheumatol. 6, 391–398 (2010). disease with and without albuminuria. Curr. Opin. or microparticles are immunostimulatory and
49. Wang, J. F. et al. Serum miR‑146a and miR‑223 Nephrol. Hypertens. 20, 246–257 (2011). can activate autoreactive T cells spontaneously
as potential new biomarkers for sepsis. 69. Perkins, B. A. et al. Microalbuminuria and the developed in nonobese diabetic mice.
Biochem. Biophys. Res. Commun. 394, 184–188 risk for early progressive renal function decline J. Immunol. 187, 1591–1600 (2011).
(2010). in type 1 diabetes. J. Am. Soc. Nephrol. 18, 90. Guay, C., Menoud, V., Gattesco, S. & Regazzi, R.
50. Zampetaki, A. et al. Plasma microRNA profiling 1353–1361 (2007). MicroRNA transfer as a new cell‑to‑cell
reveals loss of endothelial miR‑126 and other 70. Perkins, B. A. et al. Regression of communication mode between pancreatic β
microRNAs in type 2 diabetes. Circ. Res. 107, microalbuminuria in type 1 diabetes. N. Engl. cells. Diabetologia 55, S95 (2012).
810–817 (2010). J. Med. 348, 2285–2293 (2003). 91. Rodriguez, A., Griffiths-Jones, S., Ashurst, J. L.
51. Kong, L. et al. Significance of serum microRNAs 71. Martino, F. et al. Circulating microRNAs are & Bradley, A. Identification of mammalian
in pre-diabetes and newly diagnosed type 2 not eliminated by hemodialysis. PLoS ONE microRNA host genes and transcription units.
diabetes: a clinical study. Acta Diabetol. 48, 7, e38269 (2012). Genome Res. 14, 1902–1910 (2004).
61–69 (2011). 72. Neal, C. S. et al. Circulating microRNA 92. Lee, Y. et al. MicroRNA genes are transcribed
52. Karolina, D. S. et al. Circulating miRNA profiles expression is reduced in chronic kidney by RNA polymerase II. EMBO J. 23, 4051–4060
in patients with metabolic syndrome. J. Clin. disease. Nephrol. Dial. Transplant 26, (2004).
Endocrinol. Metab. 97, E2271–E2276 (2012). 3794–3802 (2011). 93. Cifuentes, D. et al. A novel miRNA processing
53. Nielsen, L. B. et al. Circulating levels of 73. Wang, G. et al. Elevated levels of miR‑146a pathway independent of Dicer requires
microRNA from children with newly diagnosed and miR‑155 in kidney biopsy and urine from Argonaute2 catalytic activity. Science 328,
type 1 diabetes and healthy controls: evidence patients with IgA nephropathy. Dis. Markers 30, 1694–1698 (2010).
that miR‑25 associates to residual beta-cell 171–179 (2011). 94. Ruby, J. G., Jan, C. H. & Bartel, D. P. Intronic
function and glycaemic control during disease 74. Wang, N. et al. Urinary microRNA‑10a and microRNA precursors that bypass Drosha
progression. Exp. Diabetes Res. 2012, 896362 microRNA‑30d serve as novel, sensitive and processing. Nature 448, 83–86 (2007).
(2012). specific biomarkers for kidney injury. PLoS ONE 95. Doench, J. G. & Sharp, P. A. Specificity of
54. Sebastiani, G. et al. MicroRNA expression 7, e51140 (2012). microRNA target selection in translational
fingerprint in serum of type 1 diabetic patients. 75. Alvarez, M. L. & Distefano, J. K. The role of non- repression. Genes Dev. 18, 504–511 (2004).
Diabetologia 55, S48 (2012). coding RNAs in diabetic nephropathy: potential 96. Trams, E. G., Lauter, C. J., Salem, N. Jr & Heine, U.
55. Erener, S., Mojibian, M., Fox, J. K., Denroche, H. C. applications as biomarkers for disease Exfoliation of membrane ecto-enzymes in the
& Kieffer, T. J. Circulating miR‑375 as a biomarker development and progression. Diabetes Res. form of micro-vesicles. Biochim. Biophys. Acta
of β-cell death and diabetes in mice. Endocrinology Clin. Pract. 99, 1–11 (2013). 645, 63–70 (1981).
154, 603–608 (2013). 76. Miranda, K. C. et al. Nucleic acids within 97. Pan, B. T., Teng, K., Wu, C., Adam, M.
56. Salas-Perez, F. et al. MicroRNAs miR‑21a and urinary exosomes/microvesicles are potential & Johnstone, R. M. Electron microscopic
miR‑93 are down regulated in peripheral blood biomarkers for renal disease. Kidney Int. 78, evidence for externalization of the transferrin
mononuclear cells (PBMCs) from patients with 191–199 (2010). receptor in vesicular form in sheep reticulocytes.
type 1 diabetes. Immunobiology 218, 733–737 77. van Balkom, B. W., Pisitkun, T., Verhaar, M. C. J. Cell Biol. 101, 942–948 (1985).
(2013). & Knepper, M. A. Exosomes and the kidney: 98. Harding, C., Heuser, J. & Stahl, P. Receptor-
57. Sebastiani, G. et al. Increased expression of prospects for diagnosis and therapy of renal mediated endocytosis of transferrin and
microRNA miR‑326 in type 1 diabetic patients diseases. Kidney Int. 80, 1138–1145 (2011). recycling of the transferrin receptor in rat
with ongoing islet autoimmunity. Diabetes Metab. 78. Beltrami, C., Clayton, A., Phillips, A. O., reticulocytes. J. Cell Biol. 97, 329–339 (1983).
Res. Rev. 27, 862–866 (2011). Fraser, D. J. & Bowen, T. Analysis of urinary 99. Vlassov, A. V., Magdaleno, S., Setterquist, R.
58. Winer, N. & Sowers, J. R. Epidemiology of microRNAs in chronic kidney disease. Biochem. & Conrad, R. Exosomes: current knowledge
diabetes. J. Clin. Pharmacol. 44, 397–405 Soc. Trans. 40, 875–879 (2012). of their composition, biological functions, and
(2004). 79. Argyropoulos, C. et al. Urinary microRNA profiling diagnostic and therapeutic potentials. Biochim.
59. Fichtlscherer, S. et al. Circulating microRNAs in in the nephropathy of type 1 diabetes. PLoS ONE Biophys. Acta 1820, 940–948 (2012).
patients with coronary artery disease. Circ. Res. 8, e54662 (2013). 100. Johnstone, R. M., Adam, M., Hammond, J. R.,
107, 677–684 (2010). 80. Zhao, C. et al. Early second-trimester serum Orr, L. & Turbide, C. Vesicle formation during
60. Fish, J. E. et al. miR‑126 regulates angiogenic miRNA profiling predicts gestational diabetes reticulocyte maturation. Association of plasma
signaling and vascular integrity. Dev. Cell 15, mellitus. PLoS ONE 6, e23925 (2011). membrane activities with released vesicles
272–284 (2008). 81. Chim, S. S. et al. Detection and characterization (exosomes). J. Biol. Chem. 262, 9412–9420
61. Wang, S. et al. The endothelial-specific microRNA of placental microRNAs in maternal plasma. (1987).
miR‑126 governs vascular integrity and Clin. Chem. 54, 482–490 (2008). 101. Kosaka, N. et al. Secretory mechanisms and
angiogenesis. Dev. Cell 15, 261–271 (2008). 82. McDonald, J. S., Milosevic, D., Reddi, H. V., intercellular transfer of microRNAs in living cells.
62. Caporali, A. et al. Deregulation of microRNA‑503 Grebe, S. K. & Algeciras-Schimnich, A. J. Biol. Chem. 285, 17442–17452 (2010).
contributes to diabetes mellitus-induced Analysis of circulating microRNA: preanalytical 102. Zhang, Y. et al. Secreted monocytic miR‑150
impairment of endothelial function and and analytical challenges. Clin. Chem. 57, enhances targeted endothelial cell migration.
reparative angiogenesis after limb ischemia. 833–840 (2011). Mol. Cell 39, 133–144 (2010).
Circulation 123, 282–291 (2011). 83. Bryant, R. J. et al. Changes in circulating
63. Creemers, E. E., Tijsen, A. J. & Pinto, Y. M. microRNA levels associated with prostate Acknowledgements
Circulating microRNAs: novel biomarkers and cancer. Br. J. Cancer 106, 768–774 (2012). The authors are supported by grants from the Swiss
extracellular communicators in cardiovascular 84. Komatsu, S. et al. Circulating microRNAs in National Science Foundation, from the European
disease? Circ. Res. 110, 483–495 (2012). plasma of patients with oesophageal squamous Foundation for the Study of Diabetes and from
64. van Empel, V. P., De Windt, L. J. cell carcinoma. Br. J. Cancer 105, 104–111 the Société Francophone du Diabète (SFD)-Servier.
& da Costa Martins, P. A. Circulating miRNAs: (2011). C. Guay is supported by a fellowship from Fonds
reflecting or affecting cardiovascular disease? 85. Li, L. M. et al. Serum microRNA profiles serve de la Recherche en Santé du Québec, the SFD
Curr. Hypertens. Rep. 14, 498–509 (2012). as novel biomarkers for HBV infection and and the Canadian Diabetes Association.
65. Pambianco, G. et al. The 30-year natural history diagnosis of HBV-positive hepatocarcinoma.
of type 1 diabetes complications: the Pittsburgh Cancer Res. 70, 9798–9807 (2010). Author contributions
Epidemiology of Diabetes Complications Study 86. Madhavan, D. et al. Circulating miRNAs as Both authors contributed equally to all aspects
experience. Diabetes 55, 1463–1469 (2006). surrogate markers for circulating tumor cells and of this article.

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 9  |  SEPTEMBER 2013  |  521


© 2013 Macmillan Publishers Limited. All rights reserved

You might also like