Sei sulla pagina 1di 10

Endocrine

DOI 10.1007/s12020-012-9830-9

REVIEW

Pathophysiology of prediabetes and treatment implications


for the prevention of type 2 diabetes mellitus
Michael Bergman

Received: 1 October 2012 / Accepted: 23 October 2012


Ó Springer Science+Business Media New York 2012

Abstract Type 2 diabetes and other non-communicable KeyWords Prediabetes  Diabetes prevention 
diseases (NCD) are a growing public health challenge Impaired fasting glucose  Impaired glucose tolerance 
globally. An estimated 285 million people, corresponding Glucolipotoxicity  Incretin effect
to 6.4 % of the world’s adult population has diabetes. This
is expected to reach 552 million by 2030, 7.8 % of the
adult population, with the African region expected to Pathophysiology of prediabetes and treatment
experience the greatest increase. A much larger segment of implications for the prevention of type 2 diabetes
the world’s population, approximating 79 million individ- mellitus
uals in the US alone, has prediabetes. Multiple factors
including genetic predisposition, insulin resistance, Type 2 diabetes and other non-communicable diseases
increased insulin secretory demand, glucotoxicity, lipo- (NCD) are a growing public health challenge globally. An
toxicity, impaired incretin release/action, amylin accumu- estimated 285 million people, corresponding to 6.4 % of
lation, and decreased b-cell mass play a causative role in the world’s adult population has diabetes. This is expected
the progressive b-cell dysfunction characteristic of predi- to reach 552 million by 2030 corresponding to 7.8 % of the
abetes. Interventions preventing progression to type 2 adult population with the African region expected to
diabetes should therefore delay or prevent b-cell failure. experience the greatest increase [1].
This article will first review the principal pathophysiolog- A much larger segment of the world’s population,
ical mechanisms underlying prediabetes and subsequently approximating 79 million individuals in the US alone, has
address treatment considerations based on these in the prediabetes. Prediabetes, in which blood glucose concentra-
prevention of type 2 diabetes. In view of long-standing tions are higher than normal but not meeting the absolute
safety data with demonstrated efficacy and cost-effective- definition of diabetes, represents a high-risk state for diabetes
ness in the prevention of type 2 diabetes in high-risk development. According to World Health Organization
individuals, metformin should be considered as initial (WHO), individuals at risk have one or both prediabetic
therapy for those unable to comply with or lifestyle mod- conditions: impaired fasting glucose (IFG), defined as a
ification or where the latter has been ineffective in fasting plasma glucose (FPG) concentration C6.1 (110 mg/
decreasing progression to type 2 diabetes. dl) and \7.0 mmol/L (126 mg/dl), and/or impaired glucose
tolerance (IGT), defined by a FPG concentration \6.1 mmol/
L (110 mg/dl) and a 2 h post-load plasma glucose concen-
tration between C7.8 (140 mg/dl) and \11.1 mmol/L
(199 mg/dl) measured during a 75 g oral glucose tolerance
test (OGTT) [2]. The American Diabetes Association (ADA)
M. Bergman (&) applies the same post-load threshold values for IGT, but uses
Department of Medicine, Division of Endocrinology,
a lower cutoff value for IFG with a FPG 5.6–6.9 mmol/L
NYU School of Medicine, 345 East 37th Street, Suite 313,
New York, NY 10016, USA (100–125 mg/dl). Furthermore, the ADA recommends that a
e-mail: Michael.Bergman@nyumc.org glycated hemoglobin A1c (HbA1c) between 5.7 and 6.4 % as

123
Endocrine

another measure for diagnosing prediabetes. The ADA and abnormalities in glucose and insulin concentrations and
WHO both recognize a HbA1c level [6.5 % as indicative of dynamics occur continuously and insidiously over many
diabetes [2–7]. years [18, 19]. Trajectories of fasting and post-load glucose
IFG and IGT differ in their pathophysiologic mecha- levels as well as insulin sensitivity and insulin secretion
nisms which will be discussed below. Individuals with both (b-cell function) preceded the development of type 2 dia-
IFG and IGT have a more severe dysglycemic condition betes in the British Whitehall II study [10]. Increased
and are especially at high-risk for type 2 diabetes. glucose values were observed as early as 13 years before
[8].5–10 % of individuals with prediabetes develop dia- diagnosis, although these seemed to be tightly regulated
betes annually with up to 70 % eventually developing within the normal range until 2–6 years before diagnosis, at
diabetes [9, 10]. which time abrupt deterioration occurred. Insulin sensi-
A meta-analysis of randomized controlled clinical trials tivity was already reduced 13 years before onset of dia-
reported reductions in the risk of developing diabetes after betes, with a steeper fall noted 5 years before diagnosis.
lifestyle or drug-based interventions although it is unclear Insulin secretion (b-cell function) was steady throughout
whether in so-doing major cardiovascular events are affected the 13 year observation period and showed a substantial
[11]. Prediabetes can revert to normoglycemia as well. In the compensatory increase 3–4 years before diagnosis before
Diabetes Prevention Program Outcome Study, reversion to decreasing steeply before conversion to diabetes. These
normal glucose, even when transient, was associated with a results indicate that insulin resistance starts many years
significant 56 % reduced risk of future diabetes independent before diabetes development and that decreased b-cell
of previous treatment [12]. Diabetes risk reduction was function is already present in the prediabetic stage.
strongly associated with the number of times normal glucose Of note, the glucose levels that define IGT occurred
regulation was achieved and was highest in those who rather late during the lengthy observation period, just
remained with prediabetes despite lifestyle intervention. before conversion to diabetes. Hence, similar to FPG,
It is also vital to note that diabetes risk substantially postprandial concentrations also follow a continuum and it
increases in those with glucose values at the higher end of the is therefore plausible that by intervening much earlier,
defined normal range [13]; therefore risk prediction might be before achieving the defined absolute IGT thresholds,
more accurately determined if glucose parameters are treated could delay or prevent progression to diabetes. As Tirosh
as a continuous rather than categorical process [14]. Fur- et al. [13] pointed out that individuals with glucose levels
thermore, incorporation of post-load glucose into a model that approximating 94 mg/dl (5.2 mmol.l), considered as nor-
already includes FPG can improve prediction [15]. The mal and below the IFG threshold, are at increased risk for
combination of HbA1c and FPG levels has been shown to developing diabetes, it is conceivable that postprandial
improve risk prediction for developing diabetes [16]. In glucose levels below 140 mg/dl (7.8 mmol/l), the cut point
postpartum women, the OGTT was superior to FPG or HbA1c for defining IGT, confer increased risk for developing
in postpartum screening demonstrating the importance of diabetes. Therefore, individuals with even subtle abnor-
measuring a post-load glucose level in this population [17]. malities in either fasting or post-load glucose levels should
Multiple factors including genetic predisposition, insulin be identified as early as possible, well before achieving
resistance, increased insulin secretory demand, glucotox- critical thresholds for IFG or IGT especially if they have
icity, lipotoxicity, impaired incretin release/action, amylin associated risk factors for progression to prediabetes.
accumulation, and decreased b-cell mass play a causative
role in the progressive b-cell dysfunction [9]. Interventions Multistage model of diabetes development
preventing progression to type 2 diabetes should therefore
delay or prevent b-cell failure [9].This article will first Weir [19] described a multistage model of diabetes
review the principal pathophysiological mechanisms development that corresponds to progression of diabetes,
underlying prediabetes and subsequently address treatment each stage marked by changes in b-cell mass, phenotype
considerations based on these in the prevention of type 2 and function.The first stage is defined by a long period of
diabetes. insulin resistance accompanied by a compensatory
increased rate of insulin secretion and increased b-cell
mass. The second stage constitutes the stable adaptation
Pathophysiology of prediabetes period when b cells no longer fully compensate for
increased insulin resistance and is accompanied by changes
Trajectories of glycemic changes in prediabetes in b-cell phenotype demonstrated by changes in gene and
protein expression; thus, fasting and post-load glucose
Blood glucose levels are strictly regulated in healthy values are not completely maintained. Glucose levels rise
individuals. With the evolution toward type 2 diabetes, to 5.0–6.5 mmol/l (89–116 mg/dl).This period is usually

123
Endocrine

accompanied by a decrease in acute insulin secretion at implies that b-cell dysfunction is already present in the
FPG concentrations of 5.6 mmol/l (100 mg/dl). Acute prediabetic phase.
glucose stimulated insulin secretion drops off considerably b-cell function cannot be characterized solely on the
and disappears completely when fasting glucose levels basis of insulin secretion without considering underlying
increase to 6.3 mmol/l (114 mg/dl). The second phase of insulin resistance. b- cells respond to an increase in
insulin secretion is partially preserved. These observations glucose concentration with a rise in insulin secretion that
correspond to the concept of glucotoxicity by which is dependent on whole body insulin sensitivity. Accord-
modestly high-glucose levels creates an unfavorable envi- ingly, the relation between insulin secretion and insulin
ronment leading to alteration in b-cell function (see sensitivity is hyperbolic (‘‘Hyperbolic Law of Glucose
below). Tolerance’’) [23], and the ratio of incremental insulin to
During the unstable early decompensation period—the incremental glucose divided by insulin resistance is
third stage of diabetes development — b cells become described by a constant known as the disposition index
unable to compensate for insulin resistance when glucose (DI), the so-called ‘‘gold-standard’’ for b-cell function
levels approximate 7.3 mmol/l (130 mg/dl) and conse- [8]. This index, higher in healthy individuals and lower in
quently glucose concentrations start to increase rapidly, as prediabetes and diabetes, therefore represents the ability
was seen in Whitehall II. This period probably extends of the glucose regulating system to compensate for
from prediabetes to manifest diabetes during which the insulin resistance by increasing plasma insulin [8, 23].
subsequent two stages, stable and severe decompensation, When the plasma insulin response to oral glucose is
occur. related to the glycemic stimulus and severity of insulin
resistance, there is a progressive decline in b-cell func-
Glucose dysregulation tion that begins in NGT individuals (2–hour plasma
glucose [100 mg/dl (5.6 mmol/l) during a 75 gram
Glucose dysregulation has been reviewed by Vasudevan OGTT [24]. A reduced DI, representative of compro-
and Garber [20]. In the fasting state, hepatic glucose output mised b-cell function, is a harbinger of type 2 diabetes
(HGO) results from both glycogenolysis and gluconeo- [23]. Studies using different measures of b-cell function
genesis accounting for approximately 90 % of the glucose have reported severely abnormal (up to 80 % decreased)
released into the circulation. Conversely, in the postpran- insulin secretion in prediabetes, findings supported by
dial state, HGO is suppressed to help limit the rise in autopsy reports describing a 50 % decrease in b-cell
plasma glucose levels and the liver stores fuel by conver- volume in IFG.
sion to glycogen. These effects are mediated in the fasting
state by an increase in gluconeogenic substrate, a reduction Skeletal muscle glucose metabolism
in insulin concentration and an increase in other hormones
such as glucagon with the converse changes occurring Aside from impaired insulin action, kinetic defects in
postprandially. Insulin directly reduces HGO by inhibition insulin action in obesity have also been demonstrated
of gluconeogenic enzymes, such as phosphoenolpyruvate whereby the rate of activation of insulin’s effect to stim-
carboxykinase. Indirectly, insulin may reduce HGO via its ulate glucose disposal is decreased and the rate of deacti-
antilipolytic action, as a strong correlation exists between vation of insulin’s effect is increased [21]. In physiologic
plasma free fatty-acid (FFA) levels and HGO.FFA stimu- circumstances, insulin is secreted post-prandially in a
late gluconeogenesis by increasing ATP and NADH, gen- phasic rather than a steady-state manner. It is therefore
erated from their oxidation in the liver. Insulin inhibition of likely that kinetic defects in insulin action are of functional
HGO has been shown to correlate with suppression of importance and steady-state measurements of insulin
plasma FFA levels. action underestimate alterations in insulin sensitivity. Pul-
Endogenous glucose production is greatly suppressed in satility and/or rapid oscillation of insulin secretion is lost or
individuals with normal glucose tolerance postprandially disordered in first-degree relatives of individuals with type
by insulin release, whereas this is less pronounced in pre- 2 diabetes who show minimal glucose intolerance or NGT,
diabetes and diabetes. In the basal state, 30 % of glucose and in those with IGT [25–28]. A loss of coordinated
uptake is insulin mediated, whereas in the post-prandial insulin secretory responses to oscillatory glucose infusion
state, this increases to 85, 80–90 % of which occurs in is found in individuals with IGT, indicating a defect in the
skeletal muscle [21, 22]. In type 2 diabetes, total body ability of the b-cell to properly sense and respond to par-
glucose disposal is decreased, 85–90 % of which is related allel changes in the plasma glucose level [27]. These
to insulin resistance in muscle. If insulin secretion was able abnormalities in the dynamics of insulin secretion represent
to compensate for insulin resistance perfectly, no obser- an early manifestation of b-cell dysfunction that precedes
vable changes in glucose concentration would occur. This the development of type 2 diabetes.

123
Endocrine

Defects in muscle glycogen synthesis have been dem- there is a progressive, sustained plasma glucose rise during
onstrated in insulin resistant states, with a 50 % reduction OGTT and the 2 h level remains well-above the fasting
observed in type 2 diabetes. As glycogen synthesis is level [8]. This concept is supported by the finding that total
known to account for the majority of non-oxidative glucose body glucose disposal can gradually worsen from normal
metabolism, a defect in glucose incorporation into glyco- glucose tolerance to IFG to IGT and then to type 2
gen is an important manifestation of insulin resistance. The diabetes.
impairment in skeletal muscle glycogen synthesis has been Although individuals with IFG have 2-hour values
attributed to defects in glucose transport, hexokinase and during an OGTT comparable to those with NGT, the
glycogen synthase [21]. 1-hour level may nonetheless be elevated. Similarly, the
1-hour level in those with IGT exceeds those with NGT
Pancreatic b-cell function and is higher than the 2-hour values during an OGTT [32].
Abdul-Ghani et al. [33, 34] demonstrated that the 1 h
Chronic hyperglycemia impairs glucose-induced pancreatic glucose concentration exceeding 150 mg/dl (8.33 mmol/l)
insulin secretion and insulin gene expression through during the OGTT is a strong predictor of future risk for
mechanisms that impact glucose desensitization, ß-cell type 2 diabetes. In the Botnia [35] and San Antonio Heart
exhaustion, and glucotoxicity [29]. Glucose desensitization Studies [33, 34], a 1-hr plasma glucose of at least 155 mg/
is applied to the rapid and reversible refractoriness of dl was the best predictor of future type 2 diabetes in
pancreatic b-cell insulin exocytosis subsequent to sustained individuals with IGT. About 40 % of those, who develop
hyperglycemia, whereas b-cell exhaustion refers to deple- type 2 diabetes have normal glucose tolerance (NGT) at
tion of the intracellular insulin pool available for rapid baseline [34]. In one study [34], 16.7 % of those with NGT
release following prolonged exposure to a secretagogue. In having a1-hour glucose level [ 155 mg/dl (8.61 mmol/l)
contrast, glucotoxicity refers to the progressively irrevers- developed type 2 diabetes over 7–8 years. The group of
ible effects of chronic hyperglycemia on b-cell function. individuals with NGT having a 1-hour level [ 155 mg/dl
b-cell secretory defects are reversible up to a point and (8.61 mmol/l) fulfilling the ATP III criteria for the meta-
become irreversible thereafter, suggesting that there is a bolic syndrome had a 4.3 % annual risk for future type 2
continuum between b-cell exhaustion and glucotoxicity, diabetes [34]. The risk in the latter group for progression
with the latter occurring more predictably after sustained exceeds that of individuals with IFG or IGT and their odds
hyperglycemia. Chronic hyperglycemia also decreases ratio for developing diabetes is double that of IGT subjects
pancreatic b-cell mass by promoting islet cell apoptosis. with a 1-hour value \155 mg/dl (8.61 mmol/l). Each
b-cell mass may also be affected by accumulation of amyloid individual with combined glucose intolerance (IFG ?
plaques although this is unlikely to be a causal factor for IGT) had a 1-hour value [155 mg/dl (8.61 mmol/l). The
onset of hyperglycemia in type 2 diabetes in man [29, 30]. 1-hour plasma glucose concentration during the OGTT
correlates more strongly with insulin secretion, insulin
Insulin resistance in IFG and IGT resistance and insulin secretion/insulin resistance index
than the 2-hour plasma glucose concentration [33]. The
Individuals with isolated IFG differ from those with iso- 1-hour value of 155 mg/dl had greater sensitivity (75 %)
lated IGT in their fasting and 2 h post-load glucose values compared to the 2-hour level (51 %) as a threshold for
and by the shape of their glucose concentration curves predicting future type 2 diabetes although specificity was
during an OGTT [8, 10]. Both groups present with insulin greater with the 2-hour value (92 % vs 79 %). Thus, high-
resistance but the site of insulin resistance differing. Ele- risk individuals with IGT with additional risk factors for
vated hepatic insulin resistance is a typical finding in IFG, progression to type 2 diabetes, can be screened using the
with almost normal skeletal muscle sensitivity. Individuals 1-hour plasma glucose to further identify those for poten-
with IFG have moderate hepatic insulin resistance and tial pharmacologic therapy [8].
impaired early (1–30 min) insulin response exocytosis of When comparing HbA1c levels between 5.7–6.4 % with
insulin from secretory vesicles docked to the membrane the OGTT for diagnosing individuals at risk for developing
[30] during the OGTT [8]. Because the late-phase plasma diabetes, a 1-hour post-load glucose level of 155 mg/dl
insulin response is intact and muscle sensitivity is normal (8.61 mmol/l) could identify more individuals with early
or near-normal, the 2 h plasma glucose returns to the initial glucose abnormalities than the 2-hour post-load level
FPG level [8]. Those with IGT have moderate-severe of 140 mg/dl (7.8 mmol/l) (unpublished, Bergman). The
muscle insulin resistance with small changes in liver 1-hour post-load glucose level diagnosed dysglycemic
sensitivity [8] and impaired early and late insulin states as more severe than did the 2-hour level in both
(60–120 min) response (granule translocation and matura- low (5.7–5.9 %) and high (6.0–6.4 %) HbA1c groups.
tion) during OGTT [31]. Although FPG is not elevated, Approximately 60 % of individuals in the low and 70 %

123
Endocrine

in the high-HbA1c group had abnormal 1-hour glucose Fatty acids and glucose constitute the primary oxidative
readings. This decreased to about 40 % in both low and fuels that support muscle contractility and their relative
high HbA1c groups using the 2-hour post-load value utilization can be adjusted to match energy supply and
reflecting the increased sensitivity of the 1-hour level. demand. Metabolic fuel switching mediated the ability of
Although these findings need to be confirmed, individuals lipid and carbohydrate pathways to regulate each other
in the low HbA1c group may already be at risk for [22]. Isolated hyperglycemia or elevated circulating FFAs
developing diabetes based on the 1-hour post-load level may not be detrimental to the b cell. Elevated glucose
and might benefit from lifestyle intervention at this stage levels alone results in oxidation and isolated elevation in
when b –cell function is still relatively intact. FFAs are oxidized instead of glucose. However, when both
glucose and FFA levels are elevated, progressive tissue
toxicity may ensue [20]. FFAs impair b-cell function,
Glucolipotoxicity particularly in the setting of elevated glucose levels. FFAs
act via multiple, ultimately converging, pathways that
The relation between glucose and lipid toxicity with insulin include suppression of cellular proliferation, impairments
resistance and b-cell toxicity has been reviewed [22, 29]. in b-cell gene transcription, alterations in glycerolipid/free
Lipid accumulation in the liver appears to be a principal fatty-acid cycling, and elevations in reactive oxidative
mechanism associated with obesity-related insulin resis- species (ROS) production leading to a phenomenon known
tance and type 2 diabetes [36]. Altered metabolism of tri- as endoplasmic reticulum stress [38]. b-cells have limited
glyceride-rich lipoproteins is an integral part of the capacity to cope with oxidative stress, and given the central
atherogenic dyslipidemia in insulin resistant prediabetic role of the ER in the production of secreted proteins such as
individuals and in type 2 diabetes, and is characterized insulin, are therefore especially prone to ER stress.
typically by elevated serum triglyceride levels and Enhanced FFA flux, uptake and oxidation inhibit glycol-
decreased high-density lipoprotein cholesterol (HDL-c). ysis (intrahepatic substrate competitive cycle) and stimulate
Increased hepatic secretion and decreased clearance of gluconeogenesis [37].When both FFA and glucose levels are
VLDL and intestinally derived chylomicrons result in elevated, FFA-derived long-chain fatty acyl-CoA ester lev-
prolonged plasma retention of these particles and accu- els are high, yet they cannot be oxidized because glucose-
mulation of highly atherogenic partially lipolyzed choles- derived malonyl-CoA is elevated as well. Malonyl-CoA
terol-enriched intermediate density lipoprotein (IDL) regulates mitochondrial cytosolic lipid partitioning (the
remnants and small dense LDL particles. Excess triacyl- relative fluxes of FFA oxidation and esterification) through
glycerols beyond the oxidative needs of lean tissue (stea- its inhibitory action on carnitine palmitoyltransferase-1
tosis) leads to a ‘‘spill over’’ effect in liver, skeletal muscle, (CPT-1), which catalyzes the rate-limiting step leading to
cardiac muscle, and endocrine pancreas, resulting in tissue mitochondrial b-oxidation of fatty acids [21, 22]. An
dysfunction or lipotoxicity, largely due to potentially toxic increase in malonyl-CoA inhibits CPT-1 and hence FFA
end products of non-oxidative FFA metabolism. This oxidation which leads to their accumulation in the cytoplasm
eventually leads to lipoapoptosis or lipid-induced cell with subsequent deleterious effects on insulin action.
death. This tissue toxic effect has been attributed to the Alternatively, if LCFA-CoAs are not transported into the
generation of specific proapoptotic lipid species or signal- mitochondria they may be re-esterified via diacylglyercol
ing molecules such as reactive oxygen species generation, (DAG) to form triglycerides and phospholipids [20, 21].
de novo ceramide synthesis, nitric oxide generation, Palmitoyl-CoA may also be converted into ceramide which
decreases in phosphatidylinositol-3-kinase, and primary may be toxic and result in insulin resistance [22].
effects on mitochondrial structure or function. Long-chain Intramyocellular triglyceride content is increased in
fatty acids (LCFA) may also suppress antiapoptotic factors obesity and type 2 diabetes has been shown to be a strong
such as Bcl-2. predictor of insulin resistance. It is likely that increased in-
FFAs, resulting from hydrolysis of stored triacylglyce- tramyocellular triglyceride content may not in itself impair
rols, result in decreased glucose transport via inhibition of insulin signaling, but serves as a marker of increased intra-
key glucose transporters in insulin responsive tissues such cellular LCFA-CoAs and lipid intermediates. There are
as skeletal muscle, resulting in insulin resistance. FFA several mechanisms by which fatty-acid intermediates can
oxidation stimulates the activity of key gluconeogenic induce insulin resistance. Both LCFA-CoAs and DAG can
enzymes (pyruvate carboxylase, phosphoenolpyruvate activate protein kinase C (PKC), especially novel PKC iso-
carboxykinase, glucose-6-phodphastase). [37]. Thus, lipo- zymes such as PKC h leading in turn to serine phosphory-
toxicity results in pancreatic (insulin secretory) and lation of insulin receptor substrate-1(IRS-1), impairing its
peripheral (insulin resistance) defects eventually resulting ability to associate with the insulin receptor, and interfering
in sustained hyperglycemia. with PI3 K activation and insulin signaling [22, 29].

123
Endocrine

Altered fat distribution decrease in caloric intake likely secondary to centrally


mediated signaling [41]. Glucose-dependent insulinotropic
Body fat distribution is an additional factor that mitigates polypeptide (GIP) as well as GLP-1 potentiate glucose-
insulin resistance. Independent total body fat mass, accu- induced insulin release. Both GLP-1 and GIP mediate the
mulation of adipose tissue within the visceral/abdominal b-cell by activation of adenylate cyclase and ATP con-
region and liver accentuates insulin resistance. The latter version to cAMP amplifying the insulin secretory response
may be related to inflammatory changes in adipose depots activated by hyperglycemia. In the absence of hypergly-
with release of cytokines [37]. Intraperitoneal (visceral) cemia, neither incretin increases insulin secretion. They
adipose tissue may be particularly deleterious as it drains also have other important effects on the b-cell including
directly to the liver via the portal vein, therefore exposing increasing Pdx-1 gene expression and stimulate gene
the liver to high concentrations of FFA [21]. Furthermore, transcription [9] resulting in replenishment of insulin
visceral adipocytes appear to be more responsive to cate- stores, prevention b-cell exhaustion and preserving b-cell
cholamine-stimulated lipolysis and less so to suppression function. GLP-1 increases glucose transporter 2 and glu-
of lipolysis by insulin. This could lead to increased FFA cokinase expression, leading to improved b-cell sensitivity
flux into muscle and liver contributing to increased intra- to glucose.
myocellular and hepatic triglyceride content and insulin Both incretins have short half-lives as they are rapidly
resistance. cleared by several peptidases, particularly dipeptidyl pep-
Changes in glucose homeostasis have been linked to tidase IV (DPP-4). Sitagliptin, a DPP-4 inhibitor, augments
altered partitioning of fat in skeletal muscle and abdominal incretin levels and improves glycemic control by lowering
adipose tissue in obese children and adolescents with IGT the appearance of glucose (augmenting splanchnic reten-
[39].These children were found to have profound periph- tion in enterocytes or hepatocytes), postprandial endoge-
eral insulin resistance with major defects in the non-oxi- nous glucose release, and glucagon response and by
dative pathway of glucose metabolism, no compensatory improving insulin sensitivity and b-cell cell glucose sens-
increase in insulin secretion and low adiponectin concen- ing [27]. Enhanced insulin release, in concert with a
trations. Adiponectin, a possible mediator of triglyceride reduction in glucagon concentration, reduces the post-
accumulation, appears to have an important role in the prandial rise in glucose concentrations. Although it is still
genesis of insulin resistance. These children also displayed somewhat uncertain whether and to what extent the GLP-1
suppression of total body lipid oxidation, plasma fatty response is reduced in type 2 diabetes, the incretin effect,
acids, and glycerol turnover. defined as the potentiation of insulin release by oral versus
Altered distribution of fat between the abdominal sub- IV glucose underlying 60–70 % of insulin secretion in
cutaneous and visceral compartments is associated with normal healthy individuals [42], is compromised in type 2
IGT. The visceral-to-subcutaneous ratio appears to be diabetes. This correlates with reduced insulin secretion and
greater in those with IGT. Intramyocellular lipid accumu- severe b-cell resistance to the stimulatory effect of GLP-1
lation and increased visceral fat mass are related to insulin on insulin secretion [8]. Although the secretion of GLP-1 is
resistance in children with prediabetes supporting the view often reduced, the insulinotropic effect of both GLP-1 and
that increased lipid content in myocytes is a marker of GIP appears to be severely compromised.
impaired insulin action. Hence, intra-myocellular and intra-
abdominal fat accumulation is strongly related to post-glu-
cose hyperglycemia in obese prediabetic young individuals. Treatment implications for the prevention of type 2
As discussed above, over accumulation of long-chain fatty diabetes mellitus
acyl-CoA interferes directly with insulin signaling and glu-
cose transport. Selective depletion of intramyocellular fat Treatment paradigms for the treatment of prediabetes have
stores appears to restore normal insulin sensitivity despite been previously reviewed [8, 10, 43–46].It is assumed that
persistent excess of total body fat. in preventing type 2 diabetes, microvascular and macro-
vascular complications should consequentially be averted
[45]. This premise has recently been addressed in a recent
Impaired incretin effect meta-analysis [11] of diabetes prevention trials demon-
strating that all-cause mortality or cardiovascular mortality
Abnormalities in the incretin axis are important in pro- was not reduced which may have been impacted by the
gressive b-cell failure in type 2 diabetes [40]. Glucagon- sample size, low cardiovascular risk of the groups studied
like peptide-1 (GLP-1) is an incretin hormone that results or the presence of other risk factors in individuals with
in glucose-dependent insulin secretion, suppression of IGT. However, a borderline statistically significant reduc-
glucagon secretion, a delay in gastric emptying, and a tion in fatal and non-fatal stroke was observed. It should be

123
Endocrine

noted that none of the prevention studies were designed to concluded that metformin was cost-effective from the
assess the benefits on cardiovascular events [47]. Further perspective of a health system and society especially when
studies are therefore required in this area. using generic formulation [8].
1. Diet and Physical exercise 3. Thiazolidinediones (TZD)
Lifestyle modification, which improves insulin sensi- Thiazolidinedione agents in high-risk individuals are
tivity and b-cell function, should serve as the cornerstone capable of stabilizing b-cell function over a prolonged
of treatment focusing on diet, 7 % weight loss, and period [49]. TZD act through the peroxisome proliferator-
150 min per week of moderate physical activity [10]. In the activated receptor-c by increasing hepatic and peripheral
Finnish Diabetes Prevention Study and the US DPP [48], a insulin sensitivity and preserving insulin secretion. These
58 % risk reduction in progression to diabetes was agents mobilize fat from muscle, liver, and b-cell thereby
observed. These effects were sustained in long-term fol- improving lipotoxicity [40]. Improved muscle insulin
low-up although the initial benefit was reduced [47]. sensitivity is related to reduced FFA/intramyocellular lipid
Maintaining weight loss and physical activity is difficult to content, altered fat topography [8]. TZD have been shown
sustain over extended time periods [8] shown in the US to effectively reduce and maintain a durable reduction in
DPP where weight was regained at the termination of this HbA1c in type 2 diabetes in long-term studies implying
study. Similarly pharmacological induced weight loss is preservation of b-cell function. Rosiglitazone (8 mg)
also followed by weight regain. Even when weight loss is showed a 60 % reduction in incident diabetes but was
achieved, only 50–60 % will avert progression to type 2 associated with a significant increase in weight (approxi-
diabetes [8] indicating that lifestyle intervention alone is mately 2 kg) and increased risk of heart failure. Low dose
insufficient to prevent diabetes in a large number of (2 mg twice daily) rosiglitazone in combination with
individuals. metformin was studied to determine if side-effects would
When lifestyle modification is ineffective, pharmaco- be reduced. The risk of diabetes was reduced by 66 %, but
logic treatment should be considered although there are no diarrhea was more prevalent than in controls (16 % vs
approved pharmacologic treatments for prediabetes [49]. 6 %) [47].
The ‘‘key’’ to diabetes prevention resides in the preserva- Pioglitazone reduced the risk of diabetes in obese indi-
tion of b-cell function [8]. Pharmacological treatment viduals with IGT by more than 70 % with improved dia-
combined with diet and exercise that improve and preserve stolic blood pressure and HDL-cholesterol and a reduced
b-cell function and enhance insulin sensitivity may there- rate of carotid intima-media thickening. However, these
fore be appropriate for treating high-risk individuals [8]. benefits were associated with greater incidence of edema
and weight gain (about 3 kg). Pioglitazone has also been
2. Metformin
associated with congestive heart failure. As the fat weight
Metformin, a biguanide, has been shown to have bene- gain and fluid retention are dose-related, limiting the dose
ficial effects on body mass index (BMI) and lipid param- to 30 mg/d may provide most of the efficacy while mini-
eters [10] and is safe without serious side-effects although mizing side-effects [8]. An increased incidence of trauma-
contraindicated if the serum creatinine is [1.5 mg/dl in related fractures have been observed in postmenopausal
males and 1.4 mg/dl in females [8]. It reduces FPG and women treated with pioglitazone and should therefore be
HbA1c by inhibiting hepatic glucose production and used with caution in this population. A possible-link with
improving insulin sensitivity [8] and does not stimulate bladder cancer has recently been suggested and it was not
insulin secretion or preserve b-cell function. Although found to be more effective than lifestyle alone in the Indian
in vitro evidence exists for improved b-cell function and DPP-2 study [47]. Troglitazone was withdrawn in Europe
prevention of apoptosis, in vivo data from the UK Pro- due to hepatotoxicity and rosiglitazone removed because of
spective Diabetes Study (UKPDS) fail to support these concerns pertaining to increased cardiovascular risk.
observations [40]. Only minor gastrointestinal side-effects
4. a-glucosidase inhibitors
have been observed, with 10–15 % of individuals not tol-
erating metformin [8]. In the US DPP, 1750 mg daily of a-glucosidase inhibitors reduce the rate of polysaccha-
metformin reduced conversion to type 2 diabetes by 31 %, ride digestion from the proximal intestine and lower pri-
which was less effective than lifestyle with greater effect in marily postprandial glucose without causing hypoglycemia
those with higher baseline BMI and FPG. The ADA and have been evaluated in diabetes prevention trials [8,
Consensus Conference recommended that high-risk indi- 10, 50–53]. In the STOP-NIDDM study, acarbose resulted
viduals (HbA1c [6.0 %, BMI [30 kg/m2, age \60 years) in a 25 % relative-risk reduction for diabetes in individuals
with IGT or IFG be treated with metformin [50]. Further- with IGT with a suggested decrease in cardiovascular
more, data derived from the DPP Research Group disease and hypertension risk. However, about a third of

123
Endocrine

the acarbose-treated individuals could not complete the 7. Insulin secretagogues


trial because of gastrointestinal side-effects [51]. Vogli-
Although sulfonylureas (glyburide,glimepiride, and gli-
bose, another a-glucosidase inhibitor, showed a 40 %
clazide) are effective in lowering glucose levels associated
reduction in incident diabetes in a high-risk Japanese
with increased risk for hypoglycemia, they have not been
population [52]. a-glucosidase inhibitors have also been
studied in the prevention of diabetes. Nonetheless, based on
shown to augment incretin secretion as well as altering gut
evidence from the treatment of type 2 diabetes, they have
micobiota flora, which may partially explain their benefi-
been associated with a progressive decline in b-cell function
cial effects [53].
[40]. There is a consistent loss of glycemic control after
5. GLP-1 analogs 18 months of therapy demonstrating the lack of durability of
sulfonylureas and loss of glucose control is associated with
The GLP-1 analogs exenatide and liraglutide produced
progressive b-cell failure. Furthermore, nateglinide, a short-
weight loss in obese individuals and reversion from pre-
acting insulin secretagogue showed no benefit in diabetes
diabetes to normoglycemia which was accompanied by
prevention or cardiovascular outcomes in 9,000 individuals
nausea and vomiting [8, 10]. Liraglutide decreased con-
with IGT in the NAVIGATOR Study [54].
version to type 2 diabetes by 84–96 % in a 20 week study
of 564 obese, non-diabetic individuals, 31 % of whom had 8. Insulin
IGT [8]. Exenatide was found to improve b-cell function
When used to target normal fasting plasma glucose
and when continued for 3 years, a persistent beneficial
levels for more than 6 years, glargine had a neutral effect
effect was found even when it was discontinued for
on cardiovascular outcomes and cancers [55]. In the pre-
1 month. In vivo rodent and in vitro cultured human islet
diabetic cohort (12 % of the study population), glargine
cell studies have shown that exenatide can expand b-cell
slowed the progression to type 2 diabetes (assessed by
mass and prevent apoptosis of islets [41]. GLP-1 analogs
repeat OGTT 6 and 12 weeks after study end) representing
also impact liver function with reduced hepatic glucose
a 28 % reduction in risk. The implications of this finding,
production, a-cell function with a reduction in glucagon
however, remain questionable. Although reducing new-
secretion, the gut by improvement of the GLP-1 response,
onset diabetes, glargine also increased hypoglycemia and
and the brain by a reduction in appetite with weight loss.
modestly increased weight (1.6 kg). Whether the decrease
The insulin stimulatory effects wanes as normoglycemia is
in incident diabetes will affect future microvascular or
achieved thereby minimizing the risk for hypoglycemia
other outcomes remains unknown. These findings therefore
[41].
do not support changing standard therapies for early dys-
As GLP-1 analogs effectively reduce IGT conversion to
glycemia [55].
type 2 diabetes, improve b-cell function, promote weight
loss, improve cardiovascular risk factors and do not cause 9. Other medications
hypoglycemia, they may be ideally suited for treating IGT
Other medications often used to treat cardiovascular risk
[8]. The major side-effects of GLP-1 receptor agonists are
factors in those with prediabetes include angiotensin-con-
nausea and vomiting which occur in 20–30 % of individ-
verting enzyme (ACE) inhibitors, angiotensin receptor
uals and are generally mild and transient. Approximately
blockers (ARBs), thiazides, b-blockers and statins.
5 % of individuals cannot tolerate these agents. While
Hypertension is 2–3 times more common in those with
pancreatitis has been listed as a potential side-effect, an
prediabetes [56]. As the effectiveness of ACE inhibitors
increased incidence of the latter has not been substantiated
and ARBs is not limited to blood pressure reduction and
[8]. In considering their potential suitability for use in the
these agents do not only pejoratively glucose-control but
prevention of diabetes, the considerable costs of these
also have a specific effect on prevention of diabetic
agents requires cost effectiveness studies should they ulti-
nephropathy [57, 58], their primary use in prediabetes is
mately be approved for this indication.
recommended [58], whereas thiazide diuretics and
6. DPP-IV inhibitors b-blockers have been associated with a higher risk of
incident diabetes [59–61]. Finally, as prediabetes and dia-
Based on their mechanism of action discussed earlier,
betes are cardiovascular risk equivalents, treatment goals
agents in this class could in principle be beneficial in dia-
for LDL-C should be similar [58]. Although statin therapy
betes prevention although there are no long-term studies
has been associated with a small, increased risk of devel-
available examining the enhancement of insulin secretion
opment of diabetes [62], benefit in individuals at high
into preservation of b-cell function [42]. Hypoglycemia,
cardiovascular risk favors their use as part of a multifac-
weight loss or appetite suppression is not associated with
torial approach to risk factor management even when dia-
their use. Cost effectiveness of these agents need to be
betes is diagnosed [62–64].
taken into consideration as well.

123
Endocrine

Conclusion subjects with prediabetes: meta-analysis of randomized con-


trolled clinical trials. Eur. J. Cardiovasc. Prev. Rehabil. 18(6),
813–823 (2011)
As we have seen, certain therapeutic classes may be useful 12. L. Perreault, Q. Pan, K.J. Mather, K.E. Watson, R.F. Hammam,
for the treatment of prediabetes-based on their specific role S.E. Kahn, For the diabetes prevention program research group.
in the complex pathophysiologic mechanisms described Effect of regression from prediabetes to normal glucose regula-
earlier. However, in view of long-standing safety data with tion on long-term reduction in diabetes risk: results from the
diabetes prevention program outcomes study, Lancet published
demonstrated efficacy and cost-effectiveness in the pre- online June 9, (2012). (doi:10.1016/S0140-6736(12)60525-X
vention of type 2 diabetes in high-risk individuals, met- 13. A. Tirosh, I. Shai, D. Tekes-Manova, E. Israeli, D. Pereg, T.
formin should be considered as initial therapy for those Shochat, I. Kochba, A. Rudich, For the Israeli diabetes research
unable to comply with or lifestyle modification or where group, normal fasting plasma glucose levels and type 2 diabetes
in young men. N. Engl. J. Med. 353(10), 1454–1462 (2005)
the latter has been ineffective in decreasing progression to 14. M. Bergman, Inadequacies of absolute threshold levels for diag-
type 2 diabetes. The use of a-glucosidase inhibitors, nosing prediabetes. Diabetes Metab. Res. Rev. 26(1), 5–6 (2010)
despite their marked gastrointestinal intolerance, may 15. E. Ferrannini, M. Massari, M. Nannipieri, A. Natali, R.L. Rida-
present another reasonable option. At the present time, ura, C. Gonzales-Villalpando, Plasma glucose levels as predictors
of diabetes: the Mexico city diabetes study. Diabetologia 52(5),
these options would be preferable to other agents lacking 818–824 (2009)
long-term efficacy data, have the potential for side-effects 16. Y. Heianza, Y. Arase, K. Fujihara, H. Tsuji, K. Saito, S.D. Hsieh,
and where costs limit their usage in the prediabetic popu- S. Kodama, H. Shimano, N. Yamada, S. Hara, H. Sone, Screening
lation. First line therapies for reducing cardiovascular risk for pre-diabetes to predict future diabetes using various cut-off
points for HbA1c and impaired fasting glucose the Toranomon
factors in prediabetes include ACE inhibitors or ARBs for hospital health management center study 4 (TOPCS 4). Diabetes
hypertension and statins for hypercholesterolemia, with Med. 29, e279–e285 (2012)
therapeutic targets similar to those with established 17. M.J. Picon, M. Murri, A. Muñoz, J.C. Fernandez-Garcia, R.
diabetes. Gomez-Huelgas, F.J. Tinahones, Hemoglobin A1c versus oral
glucose tolerance test in postpartum diabetes screening. Diabetes
Care 35(8), 1648–1653 (2012)
Conflict of Interest The author has no conflicts to report. 18. C. Weyer, C. Bogardus, D.M. Mott, R.E. Pratley, The natural
history of insulin secretory dysfunction and insulin resistance in
the pathogenesis if type 2 diabetes mellitus. J. Clin. Invest.
104(6), 787–794 (1999)
References 19. G.C. Weir, S. Bonner-Weir, Five stages of evolving b-cell dys-
function during progression to diabetes, Diabetes 53(12, suppl 3),
1. IDF, Diabetes Atlas Fourth Edition. International Diabetes S16–S21 (2004)
Federation (Brussels, Belgium, 2009) 20. A. Vasudevan, A.J. Garber, Postprandial Hyperglycemia, in
2. The Expert Committee on the Diagnosis and Classification of Contemporary Endocrinology: Type 2 Diabetes Mellitus : An
Diabetes Mellitus, Report of the expert committee on the diag- Evidence-Based Approach to practical Management, ed. by M.N.
nosis and classification of diabetes mellitus. Diabetes Care 20(7), Feinglos, M.A. Bethel (Humana Press, Totowa, 2008),
1183–1197 (1997) pp. 97–113
3. The Expert Committee on the diagnosis and classification of 21. C.H. Courtney, J.M. Olefsky, Insulin Resistance, in Mechanisms
diabetes mellitus, Follow-up report on the diagnosis of diabetes of Insulin Action, ed. by A.R. Saltiel, J.E. Pessin (Landes Bio-
mellitus. Diabetes Care 26(11), 3160–3167 (2003) science and Springer Science ? Business Media, New York,
4. American Diabetes Association, Standards of medical care in 2007), pp. 185–209
diabetes-2012. Diabetes Care 35(1), S11–S63 (2012) 22. D.M. Muoio, T.R. Koves, J. An, C. Newgard, Metabolic Mech-
5. The International Expert Committee, International expert com- anisms of Muscle Insulin Resistance, in Contemporary Endo-
mittee report on the role of the A1c assay in the diagnosis of crinology: Type 2 Diabetes Mellitus : An Evidence-Based
diabetes. Diabetes Care 32(7), 1327–1334 (2009) Approach to Practical Management, ed. by M.N. Feinglos, M.A.
6. WHO, Use of glycated haemoglobin (HbA1c) in the diagnosis of Bethel (Humana Press, Totowa, 2008), pp. 97–113
diabetes mellitus. diabetes res. Clin. Pract. 93(3), 299–309 (2011) 23. R.N. Bergman, Banting Lecture 2006. Orchestration of glucose
7. Report of a WHO/IDF consultation: definition and diagnosis of homeostasis, from a small acorn to the California Oak. Diabetes
diabetes mellitus and intermediate hyperglycemia. WHO, Gen- 56(6), 1489–1501 (2007)
eva, (2006). http://whqlibdoc.who.int/publications/2006/924159 24. M.A. Abdul-Ghani, M.P. Stern, V. Lyssenko, T. Tuomi, L.
4934_eng.pdf Groop, R.A. DeFronzo, Minimal contribution of fasting hyper-
8. R.A. DeFronzo, M.A. Abdul-Ghani, Preservation of b-cell glycemia to the incidence of type 2 diabetes in subjects with
function: the key to diabetes prevention. J. Clin. Endocrinol. normal 2-h plasma glucose. Diabetes Care 33(3), 557–561 (2010)
Metab. 96(8), 2354–2366 (2011) 25. D.A. Lang, D.R. Matthews, M. Burnett, R.C. Turner, Brief,
9. J. Shaw, R.A. Sicree, P.Z. Zimmet, Global estimates of the irregular oscillations of basal plasma insulin and glucose con-
prevalence of diabetes for 2010 and 2030. Diabetes Res. Clin. centrations in diabetic man. Diabetes 30(5), 435–439 (1981)
Pract. 87(1), 4–14 (2010) 26. S. O’Rahilly, R.C. Turner, D.R. Matthews, Impaired pulsatile
10. A.G. Tábak, C. Herder, W. Rathmann, E.J. Brunner, M. Kivimäki secretion of insulin in relatives of patients with non-insulin-
Prediabetes: a high-risk state for diabetes development. Lancet Pub- dependent diabetes. N. Engl. J. Med. 318(19), 1225–1230 (1988)
lished online June 9, (2012) doi:10.1016/S0140-6736(12)60283-9 27. M.M. Byrne, J. Sturis, R.J. Sobel, K.S. Polonsky, Elevated
11. I. Hopper, B. Billah, M. Skiba, H. Krum, Prevention of diabetes plasma 2 h post challenge predicts defects in beta-cell function.
and reduction in major cardiovascular events in studies of Am. J. Physiol. 270(4 pt1), E572–E579 (1996)

123
Endocrine

28. S. Seino, T. Shibasaki, K. Minami, Dynamics of insulin secretion 48. J.P. Crandall, W.C. Knowler, S.E. Kahn, D. Marrero, J.C. Florez,
and the clinical implications for obesity and diabetes. J. Clin. G.A. Bray, S. M. Haffner, M. Hoskin, D.M. Nathan, For the
Invest. 121(6), 2118–2125 (2011) diabetes prevention program research group. Nat. Clin. Pract.,
29. J.L. Leahy, Pathogenesis of Type 2 diabetes mellitus, in Con- www.nature.com/clinicalpractice, published online 20 May
temporary Endocrinology: Type 2 Diabetes Mellitus : An Evi- (2008). doi:10.1038/ncpendmet0843
dence-Based Approach to Practical Management, ed. by M.N. 49. W.A. Hsueh, L. Orloski, K. Wyne, Prediabetes: the importance of
Feinglos, M.A. Bethel (Humana Press, Totowa, 2008), pp. 17–33 early identification and intervention. Postgrad. Med. 122(4),
30. A. Clark, Pancreatic Islet Pathology in Type 2 diabetes, in Pan- 129–142 (2010)
creatic Beta Cell in Health and Disease, ed. by S. Seino, G.J. Bell 50. D.M. Nathan, M.B. Davidson, R.A. DeFronzo, R.J. Heine, R.R.
(Springer, New York, 2008), pp. 381–398 Henry, R. Pratley, B. Zinman, Impaired fasting glucose and
31. A. Basu, M.G. Pedersen, C. Cobelli, Commentary. Prediabetes: impaired glucose tolerance: implications for care consensus
evaluation of b-cell function. Diabetes 61(2), 270–271 (2012) statement from American diabetes association. Diabetes Care 30,
32. M.A. Abdul-Ghani, D. Tripathy, R.A. Defronzo, Contributions of 753–759 (2007)
b-Cell dysfunction and insulin resistance to the pathogenesis of 51. J.L. Chiasson, R.G. Josse, R. Gomis, M. Hanefeld, A. Karasik, M.
Impaired glucose tolerance and impaired fasting glucose. Dia- Laakso, STOP-NIDDM trial research group, Acarbose for pre-
betes Care 29(5), 1130–1139 (2006) vention of type 2 diabetes: the STOP-NIDDM randomised trial.
33. M.A. Abdul-Ghani, K. Williams, R. DeFronzo, M. Stern, What is Lancet 359, 2072–2077 (2002)
the best predictor of future type 2 diabetes? Diabetes Care 30(6), 52. R. Kawamori, N. Tajima, Y. Iwamoto, A. Kashiwagi, K. Shi-
1544–1548 (2007) mamoto, K. Kaku, Voglibose for prevention of type 2 diabetes
34. M.A. Abdul-Ghani, T. Abdul-Ghani, N. Ali, R.A. DeFronzo, mellitus: a randomised, double-blind trial in Japanese individuals
One-hour plasma glucose concentration and the metabolic syn- with impaired glucose tolerance. Lancet 1607–1614 (2009)
drome identify subjects at high risk for future type 2 diabetes. 53. R. Moriya, T. Shirakura, J. Ito, S. Mashiko, T. Seo, Activation of
Diabetes Care 31(8), 1650–1655 (2008) sodium-glucose cotransporter 1 ameliorates hyperglycemia by
35. M.A. Abdul-Ghani, V. Lyssenko, T. Tuomi, R.A. DeFronzo, L. mediating incretin secretion in mice. Am. J. Physiol. Endocrinol.
Groop, Fasting versus postload plasma glucose concentration and Metab. 297, E1358–E1364 (2009)
the risk for future type 2 diabetes :results from the Botnia Study. 54. Effect of nateglinide on the incidence of diabetes and cardio-
Diabetes Care 32(2), 281–286 (2009) vascular events, Navigator study group. N. Engl. J. Med. 362(16),
36. J.M. Haus, T.P.J. Solomon, C.M. Marchetti, J.M. Edmison, F. 1463–1476 (2010)
Gonzalez, J.P. Kirwan, Free fatty acid-induced hepatic insulin 55. The ORIGIN Trial Investigators, Basal Insulin and Cardiovas-
resistance is attenuated following lifestyle intervention in obese cular and Other Outcomes in dysglycemia. N. Engl. J. Med.
individuals with impaired glucose tolerance. J. Clin. Endocrinol. 367(4), 319–328 (2012)
Metab. 95(1), 323–327 (2010) 56. J. Segura, L.M. Ruilope, Treatment of prehypertension in dia-
37. E. Ferrannini, A. Gastaldelli, P. Iozzo, Pathophysiology of Predi- betes and metabolic synderome: what are the pros? Diabetes Care
abetes, in Prediabetes and Diabetes Prevention, ed. by M. Berg- 32(11, suppl 2), S284–S289 (2009)
man (W.B. Saunders Company, Philadelphia, 2011), pp. 327–340 57. L. Niskanen, T. Hedner, L. Hansson, J. Lanke, A. Niklason, For
38. R.G. Mirmira, Saturated free fatty acids: islet b cell ‘‘stress ER’’. the CAPP study group. Diabetes Care 24(12), 2091–2096 (2001)
Endocrine 42(6), 1–2 (2012) 58. R. DeFronzo, M. Abdul-Ghani, Assessment and treatment of
39. R. Weiss, S. Dufour, S.E. Taksali, W.V. Tamborlane, K.F. Pet- cardiovascular risk in prediabetes: impaired glucose tolerance
ersen, R.C. Bonadonna, L. Boselli, G. Barbetta, K. Allen, F. Rife, and impaired fasting glucose. Am. J. Cardiol. 108(suppl), 3B–
M. savoye, J. Dziura, R. Sherwin, G.I. Shulman, S. Caprio, 24B (2011)
Prediabetes in obese youth: a syndrome of impaired glucose 59. E.N. Taylor, F.B. Hu, G.C. Curhan, Antihypertensive medica-
tolerance, severe insulin resistance, and altered myocellular and tions and the risk of incident type 2 diabetes. Diabetes Care 29(5),
abdominal fat partitioning. Lancet 362, 951–957 (2003) 1065–1070 (2006)
40. R.A. DeFronzo, From the triumvirate to the ominous octet: a new 60. C. Manrique, M. Johnson, J.R. Sowers, Thiazide diuretics alone
paradigm for the treatment of type 2 diabetes mellitus. Diabetes or with b-blockers impair glucose metabolism in hypertensive
58(4), 773–795 (2009) patients with abdominal obesity. Hypertension 55(1), 15–17
41. A. Vella, Mechanism of action of DPP-4 inhibitors-new insights. (2010)
J. Clin. Endocrinol. Metab. 97(8), 2626–2628 (2012) 61. W.J. Elliott, P.M. Meyer, Incident diabetes in clinical trials of
42. E. Muscelli, A. Casolaro, A. Gastaldelli, a Mari, G. Seghieri, B. antihypertensive drugs: a network meta-analysis. Lancet
Astiarraga, Y. Chen, M. alba, J. Holst, E. Ferrannini, Mechanisms 369(9557), 201–207 (2007)
for the antihyperglycemic effect of sitagliptin in patients with 62. N. Sattar, D. Preiss, H.M. Murray, P. Welsh, B.M. Buckley,
type 2 diabetes. J. Clin. Endocrinol. 97(8), 2818–2819 (2012) A.J.M. de Craen, S.R.K. Seshasai, J.J. McMurray, D.J. Freeman,
43. M. Bergman, Treatment of prediabetes. Louvain Med 131(3), J.W. Jukema, P.W. Macfarlane, C.J. Packard, D.J. Stott, R.G.
104–113 (2012) Westendorp, J. Shepherd, B.R. Davis, S.L. Pressel, R. Marchioli,
44. R.E. Ratner, A. Sathasivam, Treatment recommendations for R.M. Marfisi, A.P. Maggioni, L. Tavazzi, G. Tognoni, J. Kjekhus,
prediabetes, in Prediabetes and Diabetes Prevention, ed. by M. T.R. Perdersen, T.J. Cook, A.M. Gotto, M.B. Clearfield, J.R.
Bergman (W.B. Saunders Company, Philadelphia PA., 2011), Downs, H. Nakamura, Y. Ohashi, K. Mizuno, I. Ford, Statins and
pp. 385–397 risk of incident diabetes: a collaborative meta-analysis of ran-
45. J. Armato, R.A. DeFronzo, M. Abdul-Ghani, R. Ruby, Successful domized statin trials. Lancet 375(9716), 735–742 (2010)
treatment of prediabetes in clinical practice: targeting insulin resis- 63. P.M. Ridker, A. Pradhan, J.G. MacFadyen, P. Libby, R.J. Glynn,
tance and b-cell dysfunction. Endocr. Pract. 18(3), 342–350 (2012) Cardiovascular benefits and diabetes risks of statin therapy in
46. V. Aroda, R. Ratner, Approach to the patient with prediabetes. primary prevention: an analysis from the JUPITER trail. Lancet
J. Clin. Endcorinol. Metab. 93(9), 3259–3265 (2008) 380(9841), 565–571 (2012)
47. A. Ramachandran, C. Snehalatha, Diabetes Prevention Programs, 64. A. Goldfine, Statins: is it really time to reassess benefits and risk?
in Prediabetes and Diabetes Prevention, ed. by M. Bergman N. Engl. J. Med. 366(19), 1752–1755 (2012)
(W.B. Saunders Company, Philadelphia PA, 2011), pp. 353–372

123

Potrebbero piacerti anche