ArticlePDF Available

Prospective Relation of C-Reactive Protein With Type 2 Diabetes: Response to Han et al

Authors:

Figures

No caption available
… 
Content may be subject to copyright.
OBSERVATIONS
Analysis of the
Agreement Between
the World Health
Organization
Criteria and the
National Cholesterol
Education Program-
III Definition of the
Metabolic Syndrome
Results from a population-based
survey
O
ur objective was to evaluate the di-
agnostic proficiency of the World
Health Organization (WHO) and
the National Cholesterol Education Pro-
gram (NCEP)-III definitions (1,2) for the
metabolic syndrome in a Mexican nation-
wide, population-based survey. Details of
the sampling procedures have been pre-
viously described (3). The population was
composed of 2,158 men and women aged
2069 years sampled after a 9- to 12-h
fasting period. For the WHO criteria, in-
sulin resistance was diagnosed if a non-
diabetic case had fasting insulin concen-
trations 126 pmol/l (21 U/ml) (75th
percentile in Mexican adults). The age-
adjusted prevalence was 13.61% for the
WHO criteria (n268) and 26.6% for
the NCEP-III definition (n574). After
excluding patients with diabetes, the prev-
alence was 9.2 and 21.4%, respectively.
The agreement between the definitions
was assessed in 1,969 subjects; 189 cases
were eliminated due to the lack of a urine
sample.
The number of abnormal cases was
lower using the WHO criteria. Only 237
of the 545 subjects (43.4%) who fulfilled
the NCEP criteria were diagnosed as af-
fected using the WHO definition. Just 16
of 253 cases (6.3%) detected by the WHO
definition did not fulfill the NCEP defini-
tion. The agreement between the criteria
was moderate (␬⫽0.507). On the other
hand, the subjects diagnosed using the
WHO recommendations had a worse
profile than the cases detected by the
NCEP-III definition only—they had a
higher BMI and higher non-HDL choles-
terol, triglyceride, and glucose concentra-
tions. The demonstration of insulin
resistance among the nondiabetic popu-
lation caused the lack of agreement in 202
of the 242 cases that fulfilled the NCEP
definition but failed the WHO criteria.
Other reasons for disparity were the
higher thresholds used by the WHO cri-
teria; these differences explained the lack
of agreement in 66 of the 152 cases with
diabetes.
In conclusion, the prevalence of the
metabolic syndrome is influenced by the
selection of the diagnostic criteria. The
WHO criteria identified a lower number
of cases than the NCEP-III definition.
These differences were explained mainly
by the inclusion of abnormally high insu-
lin concentrations as a diagnostic crite-
rion. However, the presence of insulin
resistance may help to identify patients
more severely affected (4).
CARLOS A. AGUILAR-SALINAS,
MD
1
ROSALBA ROJAS,
PHD
2
FRANCISCO J. GO
´MEZ-PE
´REZ,
MD
1
VICTORIA VALLES,
MD
1
JUAN MANUEL ´OS-TORRES,
MD
1
AURORA FRANCO,
PHD
2
GUSTAVO OLAIZ,
PHD
2
JUAN A. RULL,
MD
1
JAIME SEPULVEDA,
PHD
2
From the
1
Departamento de Endocrinologı´a y Me-
tabolismo del Instituto Nacional de Ciencias Me´di-
cas y Nutricio´n “Salvador Zubira´n,” Mexico City,
Me´xico; and the
2
Instituto Nacional de Salud Pu´-
blica, Cuernavaca, Morelos, Me´xico.
Address correspondence to Carlos Alberto Agui-
lar-Salinas, MD, Vasco de Quiroga 15, Mexico City
14000, Me´ xico. E-mail: caguilarsalinas@yahoo.com.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Expert Panel on Detection, Evaluation,
and Treatment of High Blood Cholesterol
in Adults: Executive Summary of The
Third Report of The National Cholesterol
Education Program (NCEP) Expert Panel
on Detection, Evaluation, and Treatment
of High Blood Cholesterol In Adults
(Adult Treatment Panel III). JAMA 285:
2486–2497, 2001
2. Alberti FGMM, Zimmet PZ: Definition,
diagnosis and classification of diabetes
mellitus and its complications. Part 1: di-
agnosis and classification of diabetes mel-
litus provisional report of a WHO con-
sultation. Diabet Med 15:539–553, 1998
3. Aguilar-Salinas CA, Rojas R, Go´mez-Pe´ rez
FJ, Garcı´a E, Valles V, ´os-Torres JM,
Franco A, Olaiz G, Sepu´lveda J, Rull JA:
Prevalence and characteristics of early-
onset type 2 diabetes in Mexico. Am J Med
113:569–574, 2002
4. Hanson R, Imperatore G, Bennett PH,
Knowler W: Components of the “meta-
bolic syndrome” and incidence of type 2
diabetes. Diabetes 51:3120–3127, 2002
Complete Blockade
of the Renin-
Angiotensin System
in Patients With
Advanced Diabetic
Nephropathy
W
e read with interest the article by
Rossing et al. (1) in the January
2002 issue of Diabetes Care.We
had previously assessed the effect of com-
plete blockade of the renin-angiotensin
system (RAS) in patients with advanced
diabetic nephropathy (type 2 diabetes)
and severe proteinuria (2). To do so, we
studied 10 patients on prior treatment
with ACE inhibitors at recommended
doses, to which 50 mg/day of Losartan
was added.
After 3 months of the combined ther-
apy, urinary protein excretion decreased
from a mean of 6.9 (95% CI 4.3–9.6) to
5.8 g/24 h (3–8.6) (P0.025) and, with
the exception of two patients, was re-
duced in all cases. The proteinuria/
urinary creatinine ratio also decreased
from 7.6 (4.2–11.05) to 6.0 g/g (3.2–8.7)
(P0.02). In one patient in which pro-
teinuria did not vary, the proteinuria/
creatinine ratio also decreased, reflecting
a possible error in the quantification of
the patient’s proteinuria instead of a lack
of response to the treatment. The glomer-
ular filtration rate (GFR), calculated using
the mean of creatinine and urea clearance,
a measurement that avoids the overesti-
mation that creatinine clearance produces
in the GFR (3), decreased slightly from
21.5 8.5 to 19.9 7.8 ml/min (P
0.158). An interesting decrease in total
cholesterol from 6.21 1.03 to 5.33
1.27 mmol/l (P0.036) was observed.
Total proteins, serum albumin, protein
intake, and mean arterial pressure (MAP)
were not significantly modified. Only one
patient had hyperkaliemia (from 5.1 to
7.2 mmol/l). No secondary effects were
seen in the other patients. At 12 months of
treatment, proteinuria decreased to 3.4
LETTERS
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1635
g/24 h and the proteinuria/urinary creat-
inine ratio to 3.6. GFR decreased by 0.83
ml min
1
month
1
in the rst 6
months and only fell by 0.18 ml min
1
month
1
during the last 6 months. Two
patients received the combined treatment
for 42 months. During the rst 18
months, proteinuria decreased by 74 and
67% in each case; GFR fell by 0.5 and 1 ml
min
1
month
1
, respectively; and MAP
did not vary in the rst case and decreased
from 123 to 101 mmHg in the second.
Over the following 24 months, protein-
uria was 1 g/24 h in both cases and GFR
increased in one patient by 2 ml/min and
did not vary in the other patient. In this
period, MAP and protein intake remained
unvaried.
The data of Rossing et al. conrm that
in the short term, complete blockade of
the RAS system produces an antiprotein-
uric effect and affords a greater renopro-
tective effect since proteinuria is an
important risk factor for kidney disease
progression (4). Although our data can
only be considered as observational nd-
ings, they do allow us to intuit that this
renoprotective effect, due to the dual
blockade of the RAS, will be increased
when treatment is prolonged to medium
or long term. Moreover, the observed de-
crease in total cholesterol levels would
contribute to reducing both the progres-
sion of the nephropathy, since elevated
serum cholesterol levels act as an inde-
pendent promoter of progression in dia-
betic nephropathy (5), and the high
cardiovascular risk seen in this type of
patient.
JAVIER DEIRA,
MD, PHD
HUGO DIAZ,
MD
JESU
´SGRANDE,
MD, PHD
From the Division of Nephrology, Internal Medi-
cine, Hosptial Virgen de la Concha, Zamora, Spain.
Address correspondence to Dr. Javier Deira, Di-
vision of Nephrology, Internal Medicine, Hospital
Virgen de la Concha, Avenida de Requejo 35, 49022
Zamora, Spain. E-mail: jdeira@saludalia.com.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Rossing K, Christensen PK, Jensen BR,
Parving H-H: Dual blockade of the renin-
angiotensin system in diabetic nephropa-
thy. Diabetes Care 25:95100, 2002
2. Deira J, Dı´az H, Gonza´lez A, Grande J: Is
there a synergic antiproteinuric and reno-
protective efect between angiotensin-con-
verting-enzyme inhibitors and angiotensin
type1 receptor antagonists in patients
with advanced diabetic nephropathy (Ab-
stract). J Hypertens 19 (Suppl. 2):S137,
2001
3. Kasiske BL, Keane WF: Laboratory assess-
ment of renal disease: clearance, urinaly-
sis, and renal biopsy. In The Kidney. 5th
ed. Brener BM, Rector FC, Eds. Philadel-
phia, WB Saunders, 1996, p. 1140
4. Remuzzi G: Nephropathic nature of pro-
teinuria. Curr Opin Nephrol Hypertens
8:655663, 1999
5. Hovind P, Rossing P, Tarnow L, Smidt
UM, Parving HH: Progression of diabetic
nephropathy. Kidney Int 59:702709, 2001
Effect of 2-Week
Treatment With
Pirenzepine on
Fasting and
Postprandial Glucose
Concentrations in
Individuals With
Type 2 Diabetes
A
cute cholinergic muscarinic block-
ade with pirenzepine (PIR) induces
dose-related reductions in plasma
glucose (PG) and plasma insulin (PI) after
mixed meals in normal individuals and in
type 2 diabetic and polycystic ovary syn-
drome patients (12). Altogether, 24 pa-
tients with type 2 diabetes (12 male and
12 female) with a fasting plasma glucose
10 mmol/l were studied for the effects
of PIR (50 mg b.i.d.) given for 2 weeks on
fasting and postprandial plasma glucose
and insulin concentrations. Patients on
ACE inhibitors or thiazide diuretics were
excluded.
Oral hypoglycemics were stopped at
least 2 weeks before treating the patients
with either PIR or placebo in a random-
ized, cross-over, double-blind fashion
with a 2-week washout period between
the two treatment arms. At the beginning
(day 1) and end (day 14) of each treat-
ment period, blood was sampled over
12 h, during which time the patients re-
ceived three standard meals. The sched-
uling for blood sampling, meals, and PIR
administration is illustrated in Fig. 1. The
primary outcome measure for the study
was the difference between PIR- and pla-
cebo-treated fasting plasma glucose on
day 14 of the study. Statistical analysis
was performed using the paired Students
ttest.
There was a signicant reduction in
the fasting PG levels on day 14 (PIR) com-
pared with day 14 (placebo) (mean reduc-
tion SD, 1.2 1.6 mmol/l, 95% CI
0.51.9, P0.002 by paired Studentst
test) (Fig. 1). There were trends for reduc-
tion in the glucose area under the curve
and highest postprandial peaks for day 14
(PIR) compared with day 14 (placebo),
but the results did not reach statistical sig-
nicance. There was no signicant differ-
ence in fasting plasma insulin levels.
The mechanism through which PIR
produces its effect on PG is unknown.
Possible hypotheses are the obliteration of
an increased hypothalamic cholinergic
tone responsible for increased 24-h growth
hormone (GH) secretion, particularly
suppression of nocturnal GH secretion,
which we and others (3,4) have previ-
ously demonstrated. Another possibility
is the effect of PIR on gastrointestinal hor-
mones through its anticholinergic effect,
which may affect PG levels indirectly (5).
In conclusion, PIR caused a statisti-
cally signicant reduction in fasting PG
levels compared with placebo after 2
weeks of therapy, but there was no corre-
sponding increase in insulin levels. PIR
may be an effective drug in the treatment
of type 2 diabetes.
AcknowledgmentsThis study was sup-
ported by a grant from Boehringer Ingelheim.
We are indebted to Drs. Mary Lewis and
Mark Lewis for their assistance with the insu-
lin assay and Liz Gardner for her assistance
with statistical analysis.
BASIL G. ISSA,
MD
1
NICHOLA DAVIES,
RGN
1
KERENZA HOOD,
PHD
2
LAKDASA D.K.E. PREMAWARDHANA,
MD
3
JOHN R. PETERS,
MD
1
MAURICE F. SCANLON,
MD
1
From the
1
Department of Endocrinology, University
Hospital of Wales, Heath Park, Cardiff, U.K.; the
2
Department of Statistics, University Hospital of
Wales, Heath Park, Cardiff, U.K.; and the
3
Depart-
ment of Medicine, Caerphilly Miners District Hos-
pital, Caerphilly, Mid Glamorgan, U.K.
Address correspondence to Dr. Basil G. Issa, MD,
FRCP, MRCP(I), Diabetes Centre, North Manches-
ter General Hospital, Delaunays Road, Crumpsall,
Manchester M8 5RB U.K. E-mail: basil-issa@mail.
nmanhc-tr.nwest.nhs.uk.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Bevan JS, Ara J, Page MD, Scanlon MF,
Peters JR: Cholinergic blockade with
pirenzepine induces dose-related reduc-
Letters
1636 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
tion in glucose and insulin responses to a
mixed meal in normal subjects and non-
insulin dependent diabetics. Clin Endocri-
nol 35:8591, 1991
2. Premawardhana LDKE, Ismail IS, Riad-
Fahmy D, Miell JP, Peters JR, Scanlon MF:
Acute cholinergic blockade with low dose
pirenzepine reduces the insulin and glucose
responses to a mixed meal in obese women
with the polycystic ovary syndrome. Clin
Endocrinol 40:617621, 1994
3. Page MD, Bevan JS, Dieguez C, Peters JR,
Scanlon MF: Cholinergic blockade with
pirenzepine improves carbohydrate toler-
ance and abolishes the GH response to
meals in normal subjects. Clin Endocrinol
30:519524, 1989
4. Pietschmann P, Schernthaner G: The ef-
fect of pirenzepine on growth hormone
and blood glucose levels in type I diabetes
mellitus: a controlled study of patients on
basal bolus insulin treatment. Acta Endo-
crinologica (Copenh) 117:315319, 1988
5. El-Sabbagh HN, Bloom SR, Adrian TE,
Prinz RA, Baron JH, Welbourne RB: The
effect of pirenzepine on meal-stimulated
gastrointestinal hormones. Scandinavian J
Gastroenterol 66:5761, 1980
Improved Insulin
Sensitivity and
Metabolic Control in
Type 2 Diabetes
Does Not Inuence
Plasma Homocysteine
T
ype 2 diabetes is characterized by
both insulin resistance and an in-
creased cardiovascular mortality
(1). Elevated plasma total homocysteine
levels are an independent risk factor for
cardiovascular disease (2). Also, in pa-
tients with type 2 diabetes, elevated
plasma total homocysteine levels are asso-
ciated with an increased risk of cardiovas-
cular disease and an increased mortality
(3,4).
It is well known that insulin resis-
tance is aggravated by chronic hypergly-
cemia. This hyperglycemia-induced
insulin resistance is reversed by treat-
ment, resulting in a prolonged period of
euglycemia (5).
Although in animal studies evidence
was found that hyperinsulinemia and/or
insulin resistance increased total homo-
cysteine levels (6), in humans the relation
between insulin resistance and total ho-
mocysteine levels is unclear, in both
healthy subjects (712) and patients with
type 2 diabetes (1316). In patients with
type 2 diabetes, a positive association be-
tween insulin resistance and total homo-
cysteine levels was reported (13), but this
nding was not reproduced (14). Fur-
thermore, in patients with type 2 diabe-
tes, a positive association between the
degree of metabolic control and total ho-
mocysteine levels was reported (15,16).
We hypothesized that if insulin resis-
tance is associated with elevated homo-
cysteine levels, thereby providing another
explanation for the increased cardiovas-
cular disease in type 2 diabetes, then ho-
mocysteine levels will decrease after
amelioration of hyperglycemia-induced
insulin resistance and associated meta-
bolic abnormalities.
Here, we report the results of an in-
tervention study (17). Eight obese type 2
diabetic patients (aged 53 13 years,
HbA
1c
12.0 1.7%, BMI 38 5.8 kg/
m
2
) with severe insulin resistance (subcu-
taneous insulin dose, four-dose insulin
regimen 1.92 0.66 units kg
1
day
1
)
were studied before and after a period of
28 5 days of intravenous insulin treat-
ment. Intravenous insulin treatment re-
sulted in euglycemia. Before and after
intervention, insulin sensitivity was as-
sessed by a hyperinsulinemic-euglycemic
clamp. Fasting plasma total homocys-
teine, HbA
1c
, and lipid concentrations
were measured on the days of the clamps.
Also, 24-h urine creatinine excretion was
investigated before and after intervention.
Plasma total homocysteine levels
were measured with a high-performance
liquid chromatography method accord-
ing to Fiskerstrand (18,19) with some
modications. Because all variables were
normally distributed, statistical analyses
of differences were performed by paired
Studentsttest. P0.05 was considered
Figure 1—Day curve of plasma glucose (A) and insulin (B) concentrations after 14 days of PIR
(50 mg b.i.d.) (F) and placebo (E). The vertical continuous lines represent the time when piren-
zepine was administered and the interrupted lines represent the times at which breakfast, lunch,
and dinner were given.
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1637
signicant. Results are given as the
mean SD.
After a 4-week period of intravenous
insulin treatment resulting in an euglyce-
mic period of 17 4 days, insulin sensi-
tivity improved since body glucose
uptake, measured by clamp, increased
(12.7 5.7 before vs. 22.4 8.8 mol
kg
1
min
1
after euglycemia, P
0.0005) and the intravenous insulin dose
required for achieving and maintaining
euglycemia decreased (1.7 0.9 before
vs. 1.1 0.6 units kg
1
day
1
after
euglycemia, P0.005). Metabolic con-
trol improved; HbA
1c
decreased substan-
tially (12.0 1.7 before vs. 8.9 1.2%
after euglycemia, P0.0001); total cho-
lesterol decreased (5.40 1.09 before vs.
4.50 1.10 mmol/l after euglycemia, P
0.05); and triglycerides tended to de-
crease (4.34 3.32 before vs. 1.91
0.69 mmol/l after euglycemia, P0.06).
These results are described in detail else-
where (17). Plasma total homocysteine
levels were similar before and after im-
proved insulin sensitivity (total homocys-
teine levels 8.5 2.4 before vs. 9.2 3.5
mol/l after euglycemia, P0.5). Urine
creatinine excretion was similar before
and after euglycemia (15.0 4.8 before
vs. 12 4.3 mol/24 h after euglycemia).
This intervention study shows that
despite amelioration of hyperglycemia-
induced insulin resistance and improved
metabolic control, plasma total homocys-
teine concentrations did not decrease.
Homocysteine levels, measured in these
patients with type 2 diabetes, were actu-
ally within the normal range of healthy
subjects (2).
Our study is the rst intervention
study to investigate the relation between
insulin resistance and plasma total homo-
cysteine levels. To date only cross-
sectional studies have been performed
(1315), which are more susceptible for
confounding by other metabolic factors.
Also in contrast to previous reports
(8,9,11,12,14,15), we studied insulin
sensitivity by clamp, the gold standard.
We conclude that amelioration of hy-
perglycemia-induced insulin resistance
does not change plasma total homocys-
teine levels. These data refute the hypoth-
esis that homocysteine levels are
inuenced by insulin resistance and by
the degree of metabolic control, at least in
the patients we studied. Therefore, these
results do not support elevated homocys-
teine levels as an explanation for the link
between insulin resistance and the in-
creased risk of cardiovascular disease in
type 2 diabetes.
AcknowledgmentsThis study was sup-
ported by a grant from the Dutch Diabetes
Foundation. H.J.B. is an established investiga-
tor of the Netherlands Heart Foundation
(D97.021).
MARIE-JOSE J. POUWELS,
MD
1,2
MARTIN DEN HEIJER,
MD, PHD
1
HENK J. BLOM,
PHD
3
CEES J. TACK,
MD, PHD
1,2
ADR. HERMUS,
MD, PHD
1
From the
1
Department of Endocrinology, University
Medical Center Nijmegen, the Netherlands; the
2
De-
partment of General Internal Medicine, University
Medical Center Nijmegen, the Netherlands; and the
3
Laboratory of Pediatrics and Neurology, University
Medical Center Nijmegen, the Netherlands.
Address correspondence to Marie-Jose Pouwels,
MD, Division of General Internal Medicine, Medisch
Spectrum Twente, P.O. Box 50000, 7500 KA En-
schede, The Netherlands. E-mail: m.j.pouwels@
club.tip.nl.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Haffner SM, Lehto S, Ro¨nnemaa T,
Pyo¨ra¨la¨K, Laakso M: Mortality from cor-
onary heart disease in subjects with type 2
diabetes and in nondiabetic subjects with
and without prior myocardial infarction.
N Engl J Med 339:229234, 1998
2. Welch GN, Loscalzo J: Homocysteine and
atherothrombosis.NEngl JMed338:1042
1050, 1998
3. Hoogeveen EK, Kostense PJ, Beks PJ,
Mackaay AJC, Jakobs C, Bouter LM,
Heine RJ, Stehouwer CD: Hyperhomocys-
teinemia is associated with an increased
risk of cardiovascular disease, especially
in non-insulin-dependent diabetes melli-
tus: a population-based study. Arterioscler
Thromb Vasc Biol 18:133138, 1998
4. Hoogeveen EK, Kostense PJ, Jakobs C,
Dekker JM, Nijpels G, Heine RJ, Bouter
LM, Stehouwer CD: Hyperhomocysteine-
mia increases risk of death, especially in
type 2 diabetes: 5-year follow-up of the
hoorn study. Circulation 101:15061511,
2000
5. Rossetti L: Glucose toxicity. Clin Invest
Med 18:255260, 1995
6. Fonseca V, Dicker-Brown A, Ranganathan
S, Song W, Barnard RJ, Fink L, Kern PA:
Effects of a high-fat-sucrose diet on en-
zymes in homocysteine metabolism in the
rat. Metabolism 49:736741, 2000
7. Giltay EJ, Hoogeveen EK, Elber JM,
Gooren LJ, Asscheman H, Stehouwer CE:
Insulin resistance is associated with ele-
vated plasma total homocysteine levels in
healthy, non-obese subjects. Atherosclero-
sis 139:197198, 1998
8. Meigs JB, Jacques PF, Selhub J, Singer DE,
Nathan DM, Rifai N, DAgostino RB Sr,
Wilson PW: Fasting plasma homocys-
teine levels in the insulin resistance syn-
drome: the Framingham offspring study.
Diabetes Care 24:14031410, 2001
9. De Pergola G, Pannacciulli n, Zamboni M,
Minenna A, Brocco G, SciarafaM,
Bosello, Giorgino R: Homocysteine
plasma levels are independently associ-
ated with insulin resistance in normal
weight, overweight and obese pre-meno-
pausal women. Diabetes Nutr Metab 14:
253258, 2001
10. Abbasi F, Facchini F, Humphreys MH,
Reaven GM: Plasma homocysteine con-
centrations in healthy volunteers are not
related to differences in insulin-mediated
glucose disposal. Atherosclerosis 146:175
178, 1999
11. Bar-on H, Kidron M, Friedlander Y, Ben-
Yehuda A, Selhub J, Rosenberg IH, Fried-
man G: Plasma total homocysteine levels
in subjects with hyperinsulinemia. J Intern
Med 247:287294, 2000
12. Godsland IF, Rosankiewicz JR, Proudler
AJ, Johnston DG: Plasma total homocys-
teine concentrations are unrelated to in-
sulin sensitivity and components of the
metabolic syndrome in healthy men.
J Clin Endocrinol Metab 86:719723, 2001
13. Emoto M, Kanda H, Shoji T, Kawagishi T,
Komatsu M, Mori K, Tahara H, Ishimura
E, Inaba M, Okuno Y, Nishizawa Y: Im-
pact of insulin resistance and nephropa-
thy on homocysteine in type 2 diabetes.
Diabetes Care 24:533538, 2001
14. Buysschaert M, Dramais AS, Wallemacq
PE, Hermans MP: Hyperhomocysteine-
mia in type 2 diabetes: relationship to
macroangiopathy, nephropathy, and in-
sulin resistance. Diabetes Care 23:1816
1822, 2000
15. Drzewoski J, Czupryniak L, Chwatko G,
Bald E: Total plasma homocysteine and
insulin levels in type 2 diabetic patients
with secondary failure to oral agents. Di-
abetes Care 22:20972098, 1999
16. Chan NN: Homocysteine and insulin lev-
els in type 2 diabetic patients (Letter). Di-
abetes Care 23:1041, 2000
17. Pouwels MJ, Tack CJ, Hermus AR, Lutter-
man JA: Treatment with intravenous
insulin followed by continuous subcuta-
neous insulin infusion improves glycemic
control in severely resistant type 2 dia-
betic patients. Diabet Med 20:7679,
2003
18. Fiskerstrand T, Refsum H, Kvalheim G,
Ueland PM: Homocysteine and other thi-
ols in plasma and urine: automated deter-
mination and sample stability. Clin Chem
39:263271, 1993
Letters
1638 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
19. te Poele-Pothoff MT, van der Berg M,
Franken DG, Boers GH, Jacobs C, de
Kroon IF, Eskes TK, Trijbels JM, Blom HJ:
Three different methods for the determi-
nation of total homocysteine in plasma.
Ann Clin Biochem 32:218220, 1995
The True Overall
Mortality of
Unselected
Nephropathic Type 2
Diabetic Patients
With Mild to
Moderate Renal
Insufciency
D
espite signicant improvements in
therapy over the last decade, the
prognosis of nephropathic type 2
diabetic patients continues to be poor (1).
In recent antihypertensive intervention
studies performed in type 2 diabetic pa-
tients with overt nephropathy, the mean
5-year mortality rate was 20%. In the
irbesartan study published by Lewis et al.
(2), the mean death rate after 54 months
ranged from 15 (irbesartan group) to
16.3% (placebo group). Similar data were
obtained in the losartan study published
by Brenner et al. (3). These results might
lead to false conclusions concerning the
overall mortality of all unselected type 2
diabetic patients with overt nephropathy.
In the irbesartan study, major excretion
criteria were congestive heart failure
(New York Heart Association [NYHA]
class III or worse) and cardiovascular
events within the last 3 months.
In accordance with the irbesartan
study, in 1996 we recruited all nephro-
pathic type 2 diabetic patients who were
treated at that time in our hospital and/or
in the outpatient care unit and who ful-
lled the inclusion criteria of the study
(serum creatinine 1.03.0 mg/dl in fe-
male and 1.3- 3.0 mg/dl in male patients,
as well as protein excretion 1.0 g/24-h
urine), without considering the usual ex-
clusion criteria. We recruited 46 nephro-
pathic type 2 diabetic patients (aged 61
8 years, 26 women and 20 men, diabetes
duration 17 4 years), and we prospec-
tively studied their progression of diabetic
nephropathy and the 5-year mortality rate.
Cardiovascular disease was present in
43%, while 28% had heart failure (NYHA
class III and higher). Of patients, 36% had
exclusion criteria according to the irbe-
sartan study. All patients underwent eval-
uations in our outpatient unit (n24) or
at the ofces of their general physicians in
at least 6-month intervals over an obser-
vation period of 5 years. The following
parameters were routinely measured: cre-
atinine in serum and 24-h urine, creati-
nine clearance (calculated), protein in
24-h urine, HbA
1c
, and blood pressure.
The three end points of the study were
doubling of serum creatinine levels, end-
stage renal disease, or death. Patients were
also divided into those with and without
heart failure, and 5-year mortality rates
were assessed in both groups.
Serum creatinine had doubled in 37%
of patients over 60 months, compared
with 22% within 54 months in the irbe-
sartan study. The percentage of patients
who reached end-stage renal disease was
26% in our patients and 16.7% in the
irbesartan study. During the period of ob-
servation, mean HbA
1c
level in our pa-
tients was 7.6 1.0%, while their mean
blood pressure was 139 11/80 8
mmHg. In the irbesartan study, the mean
blood pressure at visits after baseline was
similar, with 140/77 mmHg in the irbe-
sartan group and 144/80 mmHg in the
placebo group.
The overall 5-year mortality of all of
our diabetic patients was 33%. Five-year
mortality rates were 57% in patients with
heart failure (NYHA III or IV) and 22%
(P0.05) in those without heart failure.
In comparison with the data of the inter-
vention studies, the mortality of our ne-
phropathic type 2 diabetic patients
without heart failure was similar to that of
the selected patients in the irbesartan
study. In contrast, survival was signi-
cantly lower in patients with heart failure,
and as a consequence, the overall mortal-
ity of all unselected type 2 diabetic pa-
tients with overt nephropathy was also
higher than that registered in the inter-
vention studies. There were no signicant
differences at baseline visit in creatinine
clearance (139 31 vs. 134 32 ml
min
1
1.73 m
2
) and urinary protein
(2.4 1.3 vs. 2.3 1.2 g/24 h) in the
groups with and without chronic heart
failure, respectively. The mean blood
pressure during the observation period
was approximately the same in both
groups (140 12 vs. 138 11 mmHg).
Twenty-eight percent of the patients with
and 25% of those without heart failure
reached end-stage renal disease. Our re-
sults are in agreement with those of Short
(4), who found, at a mean follow-up of
5.3 years, a mortality rate of 37% in ne-
phropathic type 2 diabetic patients com-
pared with just 8% in a nonnephropathic
group. Multivariate Cox regression anal-
yses conrmed a vefold excess risk for
cardiovascular mortality in type 2 diabetic
patients with overt nephropathy. Angio-
tensin II blockers have shown effective
cardiovascular protection in type 2 dia-
betic patients (5).
In conclusion, the real mortality rate
of all unselected type 2 diabetic patients
with overt nephropathy and mild-to-
moderate renal insufciency is much
higher than the mortality of nephropathic
type 2 diabetic patients selected for anti-
hypertensive studies. The difference be-
tween outcome in studies versus real life
is mainly due to the fact that patients with
heart failure are excluded from studies,
but obviously present in a nonstudy
situation.
GEORG BIESENBACH,
MD
OTMAR JANKO,
MD
From the 2nd Department of Medicine, General
Hospital, Linz, Austria.
Address correspondence to Georg Biesenbach,
MD, 2nd Department of Medicine, General Hospi-
tal, Krankenhausstrasse 9, 4020 Linz, Austria. E-
mail: georg.biesenbach@akh.linz.at.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Ismail N, Becker B, Strzelczyk P, Ritz E:
Renal disease and hypertension in non-
insulin-dependent diabetes mellitus. Kid-
ney Int 55:128, 1999
2. Lewis EJ, Hunsicker LG, Bain RP, Clarke
WR, Berl T, Pohl MA, Lewis JB, Ritz E,
Atkins RC, Rhode R: Renoprotective effect
of the angiotensin-receptor antagonist
irbesartan in patients with nephropathy
due to type 2 diabetes. N Engl J Med 345:
851860, 2001
3. Brenner BM, Cooper ME, Dick de Zeeuw,
Keane WF, Mitch WE, Parving HH, Re-
muzzi G, Snapinn SM, Zhang Z, Shahnaz
S, for the RENAAL Study Investigators:
Effects of losartan on renal and cardiovas-
cular outcomes in patients with type 2 di-
abetes and nephropathy. N Engl J Med
345:861869, 2001
4. Short R: Natural history and prognostic
factors: nephropathy in type 2 diabetes. Q
J Med 95:371377, 2002
5. Deferrari G, Ravers M, Deferrari L, Vettor-
etti S, Ratto E, Parodi D: Renal and cardio-
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1639
vascular protection in type 2 diabetes
mellitus: angiotensin II receptor blockers.
J Am Soc Nephrol 13 (Suppl. 3):224229,
2002
HLA-DR-DQ
Haplotype in Rapid-
Onset Type 1
Diabetes in
Japanese
T
ype 1 diabetes is characterized by
the presence of insulitis and auto-
antibodies, at least in the earlier
course of the disease. It is well known that
HLA locus confers strong genetic suscep-
tibility to the disease. Imagawa et al. (1)
described a unique subtype of type 1 di-
abetes that shows a rapid and fulminant
onset of the disease but lacks evidence of
insulitis and autoantibodies. Although
the etiology of this unique subtype of type
1 diabetes is unclear at present, Tanaka et
al. (2) very recently reported that specic
HLA-DQ genotypes may be associated
with this subtype of type 1 diabetes. Here,
we report clinical characteristics of sub-
jects with type 1 diabetes of a rapid and
fulminant onset who were collected from
the registration of type 1 diabetes in our
institution.
We identied 19 type 1 diabetic sub-
jects showing an acute and abrupt onset
of diabetes (duration of hyperglycemic
symptoms 6 3 days [mean SD],
range 110) accompanied by low HbA
1c
levels (6.8 0.9%, 5.48.2). The sub-
jects consisted of 11 men and 8 women
aged 38 17 years (range 1666) with
BMI of 22.2 3.2 kg/m
2
(17.127.7).
Family history of diabetes (possibly type 2
diabetes) was recorded in three subjects
(16%). Among eight women, four mani-
fested diabetes during gestation (n2) or
just after delivery (n2). History of pre-
ceding upper respiratory infection was
noted in nine subjects (47%). Despite
short duration of subjective symptoms
and low HbA
1c
levels, their initial concen-
trations of plasma glucose was as high as
34.1 13.2 mmol/l (range 13.672.4),
indicating a fulminant onset of diabetes.
In fact, 14 subjects showed diabetic keto-
acidosis at their initial presentation (arte-
rial pH 7.14 0.17, 6.807.34), and the
remaining ve had marked ketosis and
hyperglycemia. GAD autoantibodies,
measured by commercial RIA kits (Cos-
mic Corporation, Tokyo) (3), were nega-
tive in 18 of the 19 subjects (1.5 units/
ml), but a single subject showed a weakly
positive level (5.6 units/ml). Insulinoma-
associated protein 2 (IA2) autoantibody
and anti-thyroperoxidase and -thyroglob-
ulin antibodies were negative in all sub-
jects. Fasting serum level and urinary
excretion of C-peptide were decreased to
0.10 0.07 nmol/l (00.26) and 2.2
2.1 mol/day (0.26.9), respectively,
and they did not show any signs of remis-
sion of insulin dependency during the ob-
servation period.
As for HLA status, HLA-DRB1-DQB1
genotypes were determined and com-
pared with those in autoantibody (GAD
and/or IA2 autoantibodies)-positive sub-
jects with signicant type 1A diabetes
(n87) derived from our registration.
Frequency of HLA-DRB1*0405-
DQB1*0401 haplotype in a homozygous
manner was signicantly higher in the
rapid-onset group (5 of 19, 26%) than the
type 1A group (4 of 87, 5%, P0.009 by
Fishers exact probability). The frequency
was also far higher than that of nondia-
betic Japanese subjects reported in the lit-
erature (1 of 84, P0.001 [2] and 6 of
157, P0.003 [4]). Frequency of other
susceptible haplotypes (DRB1*0405-
DQB1*0401 in a heterozygous manner
and DRB1*0901-DQB1*0303 in a ho-
mozygous manner) (4) did not differ be-
tween the rapid-onset group and type 1A
group (data not shown).
In our experience, homozygous HLA-
DRB1*0405-DQB1*0401 haplotype con-
fers genetic susceptibility to rapid-onset
type 1 diabetes in Japanese, consistent
with the recent report by Tanaka et al. (2).
Frequency of GAD autoantibodies was ac-
tually low in this subgroup of type 1 dia-
betes (1,2), but a single subject showed a
positive result at the onset of diabetes.
There was also a case report in which
GAD autoantibodies turned positive 1
year after the fulminant onset of type 1
diabetes (5). Tanaka et al. (6) also dem-
onstrated presence of insulitis in a case of
rapid-onset type 1 diabetes. Collectively,
it is suggested that rapid-onset type 1 di-
abetes could share autoimmune etiology
with type 1A diabetes (7). Of interest is
that half of the female patients developed
diabetes during or just after pregnancy.
There have been many studies reporting
altered immune response during preg-
nancy (8,9), and pregnancy is considered
to be a precipitating factor for type 1 dia-
betes (10). We hypothesize that altered
immune response to unknown stimulus
may be causative of rapid and aggressive
-cell loss, together with the genetic sus-
ceptibility, at least in part, determined by
HLA-DR-DQ haplotypes.
TOMOATSU NAKAMURA,
MD
SHOICHIRO NAGASAKA,
MD
IKUYO KUSAKA,
MD
TOSHIMITSU YATAGAI,
MD
JIANMEI YANG,
MD
SHUN ISHIBASHI,
MD
From the Division of Endocrinology and Metabo-
lism, Jichi Medical School, Tochigi, Japan.
Address correspondence to Shoichiro Nagasaka,
MD, Division of Endocrinology and Metabolism, Ji-
chi Medical School, Yakushiji 3311-1, Minami-
kawachi, Tochigi 329-0498, Japan. E-mail:
sngsk@jichi.ac.jp.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Imagawa A, Hanafusa T, Miyagawa J, Mat-
suzawa Y: A novel subtype of type 1 dia-
betes mellitus characterized by a rapid
onset and an absence of diabetes-related
antibodies. N Engl J Med 342:301307,
2000
2. Tanaka S, Kobayashi T, Nakanishi K,
Koyama R, Okubo M, Murase T, Odawara
M, Inoko H: Association of HLA-DQ ge-
notype in autoantibody-negative and rap-
id-onset type 1 diabetes. Diabetes Care 25:
23022307, 2002
3. Akamine H, Komiya I, Shimabukuro T,
Asawa T, Tanaka H, Yagi N, Taira T, Na-
gata K, Arakaki K, Wakugami T, Takasu
N, Powell MJ, Furmaniak J, Smith BR:
High prevalence of GAD65 (and IA-2) an-
tibodies in Japanese IDDM patients by a
new immunoprecipitation assay based on
recombinant human GAD65. Diabet Med
14:778784, 1997
4. Kawabata Y, Ikegami H, Kawaguchi Y, Fu-
jisawa T, Shintani M, Ono M, Nishino M,
Uchigata Y, Lee I, Ogihara T: Asian-spe-
cic HLA haplotypes reveal heterogeneity
of the contribution of HLA-DR and -DQ
haplotypes to susceptibility to type 1 dia-
betes. Diabetes 51:545551, 2002
5. Shimada A, Oikawa Y, Shigihara T, Senda
T, Kodama K: A case of fulminant type 1
diabetes with strong evidence of autoim-
munity (Letter). Diabetes Care 25:1482
1483, 2002
6. Tanaka S, Kobayashi T, Momotsu T: A
novel subtype of type 1 diabetes mellitus.
N Engl J Med 342:18351837, 2000
7. Lernmark A: Rapid-onset type 1 diabetes
with pancreatic excorine dysfunction.
N Engl J Med 342:344345, 2000
Letters
1640 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
8. Whitacre CC, Reingold SC, OLooney PA:
A gender gap in autoimmunity. Science
283: 12771278
9. Elenkov IJ, Wilder RL, Bakalov VK, Link
AA, Dimitrov MA, Fisher S, Crane M,
Kanik KS, Chrousos GP: IL-12, TNF-,
and hormonal changes during late preg-
nancy and early postpartum: implications
for autoimmune disease activity during
these times. J Clin Endocrinol Metab 86:
49334938, 2001
10. Buschard K, Buch I, Molsted-Pedersen L,
Hougaad P, Kuhl C: Increased incidence
of true type 1 diabetes acquired during
pregnancy. BMJ 294:275279, 1987
Sex, Diabetes, and
Stroke After Carotid
Endarterectomy
W
hile diabetes is associated with
increased risk of perioperative
stroke among subjects who un-
dergo carotid endarterectomy (13),
women and minorities have been gener-
ally underrepresented in such trials (4).
The North American Symptomatic Ca-
rotid Endarterectomy Trial (NASCET)
was a randomized study of 2,885 eligible
patients with carotid stenosis 70% who
received either medical (n1,449) or
surgical therapy (n1,436 patients) (3).
The median study age was 66 years, and
93% of study subjects were white. Ap-
proximately 30% of NASCET subjects
were female, and 22% had a history of
diabetes, which was almost always type 2
diabetes (3). The study showed that end-
arterectomy safely and efcaciously re-
duced the risk of stroke in symptomatic
patients with higher-grade carotid arterial
stenosis when compared with medical
therapy (3). Multivariate analysis indi-
cated an increased risk of perioperative
stroke in subjects with a history of diabe-
tes (relative risk [RR] 2.0, 95% CI 1.2
3.1) (3). Because NASCET had a relatively
high proportion of both female and dia-
betic subjects, we addressed the hypoth-
esis that there would be differences
between diabetic men and women with
respect to the risk of peri- and postoper-
ative complications. Subjects were subdi-
vided by surgical or medical therapy and
then subdivided again by sex and diabetes
history. End point data regarding strokes
and deaths were collected.
Diabetic men had a signicantly in-
creased risk of stroke or death 30 days
postendarterectomy compared with non-
diabetic men (RR 2.3, 95% CI 1.43.7),
with no difference between diabetic and
nondiabetic women (1.1, 0.52.4). Fur-
thermore, diabetic men had a signi-
cantly increased risk of stroke 3 years
postendarterectomy compared with non-
diabetic men (RR 1.9, 95% CI 1.32.8),
again with no difference between diabetic
and nondiabetic women (1.0, 95% CI
0.61.9). However, at 3 years postran-
domization, diabetic men who received
medical treatment had no increased risk
of stroke compared with nondiabetic men
(RR 0.7, 0.51.1). In contrast, at 3 years
postrandomization, diabetic women who
received medical treatment had a signi-
cantly increased risk of stroke compared
with nondiabetic women (1.7, 1.12.8).
The results support the emerging im-
pression of sex-related differences among
diabetic subjects with respect to vascular
disease (5,6). Perioperative neurological
morbidity of carotid endarterectomy is
mainly due to embolization at the time of
clamping or clamp release, ischemia dur-
ing clamping of extracranial arteries, and
intracerebral hemorrhage (7). There has
been a steady decrease in postoperative
morbidity among patients who undergo
carotid endarterectomy as a result of both
improved surgical technique and patient
selection (7). Baseline clinical attributes
are also helpful predictors of postopera-
tive mortality and morbidity from stroke
(3). Our analysis suggests that among di-
abetic subjects who undergo carotid end-
arterectomy, there may be sex-related
differences in postoperative neurological
morbidity. As with any post hoc subgroup
analysis, the results should be interpreted
cautiously and must be replicated in other
studies. However, if these ndings are
conrmed, it might be appropriate to
consider sex and diabetes status in strate-
gies to optimize the management of pa-
tients with carotid disease (8).
ROBERT A. HEGELE,
MD, FRCPC
1
MICHAEL ELIASZIW,
PHD
2
MASSIMILIANO DEANGELIS,
MD
3
From the
1
Robarts Research Institute, London, On-
tario, Canada; the
2
Department of Community
Health Sciences, Faculty of Medicine, University of
Calgary, Calgary, Alberta, Canada; and the
3
Depart-
ment of Internal Medicine, Endocrinology and Me-
tabolism Section, University of Perugia, Perugia, Italy.
Address correspondence to Dr. Robert Hegele,
Robarts Research Institute, London, Ontario N6A
5K8, Canada. E-mail: hegele@robarts.ca.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Salenius JP, Harju E, Riekkinen H: Early
cerebral complications in carotid endar-
terectomy: risk factors. J Cardiovasc Surg
31:162167, 1990
2. Akbari CM, Pomposelli FBJ, Gibbons
GW, Campbell DR, Freeman DV, LoGerfo
FW: Diabetes mellitus: a risk factor for
carotid endarterectomy? J Vasc Surg 25:
10701075, 1997
3. Barnett HJ, Taylor DW, Eliasziw M, Fox
AJ, Ferguson GG, Haynes RB, Rankin RN,
Clagett GP, Hachinski VC, Sackett DL:
Benet of carotid endarterectomy in pa-
tients with symptomatic moderate or severe
stenosis: North American Symptomatic
Carotid Endarterectomy Trial Collabora-
tors. N Engl J Med 339:14151425, 1998
4. Rigdon EE: Racial and sex differences in
outcome after carotid endarterectomy.
Am Surg 64:527530, 1998
5. Anderson KM, Odell PM, Wilson PW,
Kannel WB: Cardiovascular disease risk
proles. Am Heart J 121:293298, 1991
6. Wilson PW: Diabetes mellitus and coro-
nary heart disease. Am J Kidney Dis 32:
S89S100, 1998
7. Riles TS, Imparato AM, Jacobowitz GR,
Lamparello PJ, Giangola G, Adelman MA,
Landis R: The cause of perioperative
stroke after carotid endarterectomy. J Vasc
Surg 19:206214, 1994
8. Barnett HJ, Meldrum HE, Eliasziw M,
North American Symptomatic Carotid
Endarterectomy Trial (NASCET) collabo-
rators: The appropriate use of carotid
endarterectomy.CMAJ 166:11691179,
2002
The Diabetes
Empowerment Scale
Short Form (DES-SF)
I
n 2000 we developed the Diabetes Em-
powerment Scale (DES) to measure the
psychosocial self-efcacy of people
with diabetes. The original questionnaire
contained 37 items representing eight
conceptual dimensions (i.e., assessing the
need for change, developing a plan, over-
coming barriers, asking for support, sup-
porting oneself, coping with emotion,
motivating oneself, and making diabetes
care choices appropriate for ones priori-
ties and circumstances). Using factor
analyses the questionnaire was reduced to
the current 28-item DES (␣⫽0.96) con-
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1641
taining three subscales (1). The three sub-
scales are: 1) managing the psychosocial
aspects of diabetes with 9 items (␣⫽
0.93), 2) assessing dissatisfaction and
readiness to change with 9 items (␣⫽
0.81), and 3) setting and achieving goals
with 10 items (␣⫽0.91). In addition to
providing an overall assessment of dia-
betes-related psychosocial self-efcacy,
the three subscales of the DES allow for an
examination of its underlying components.
To allow for a brief overall assessment
of diabetes-related psychosocial self-
efcacy, we developed an eight-item short
form of the DES (the DES-SF). The
DES-SF was created by choosing the item
from the remaining 28 items with highest
item to subscale correlation from each of
the original eight conceptual domains.
The reliability of the DES-SF using the
original dataset was ␣⫽0.85. We have
subsequently administered the DES-SF to
229 subjects in a new study. The reliabil-
ity of the DES-SF using the data from the
new sample was ␣⫽0.84. The content
validity of the DES-SF was supported in
the new study by the fact that both
DES-SF scores and HbA
1c
levels changed
in a positive direction after the 229 sub-
jects completed a 6-week problem-based
patient education program (2). The
change in DES-SF scores and HbA
1c
levels
were not correlated, suggesting that these
two measures vary independently.
These data provide preliminary evi-
dence that the DES-SF is a valid and reli-
able measure of overall diabetes-related
psychosocial self-efcacy. The DES and
the DES-SF, scoring information, and
permission to use them can be down-
loaded from the Michigan Diabetes Re-
search and Training Center web site at:
www.med.umich.edu/mdrtc.
ROBERT M. ANDERSON,
EDD
1
JAMES T. FITZGERALD,
PHD
1
LARRY D. GRUPPEN,
PHD
1
MARTHA M. FUNNELL,
MS, RN, CDE
2
MARY S. OH,
BS
1
From the
1
University of Michigan, DRTC, Depart-
ment of Medical Education, Ann Arbor, Michigan;
and the
2
University of Michigan, Diabetes Research
and Training Center, Ann Arbor, Michigan.
Address correspondence to Robert M. Anderson,
Department of Medical Education, G1100 Towsley
Center, 1500 East Medical Center Dr., Ann Arbor,
MI 48109-0201. E-mail: boba@umich.edu.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Anderson RM, Funnell MM, Fitzgerald JT,
Marrero DG: The diabetes empowerment
scale: a measure of psychosocial self-ef-
cacy. Diabetes Care 23:739743, 2000
2. Anderson RM, Funnell MM, Nwankwo R,
Gillard ML, Fitzgerald JT, Oh M: Eval-
uation of a problem-based, culturally
specic, patient education program for
African Americans with diabetes (Ab-
stract). Diabetes 50 (Suppl. 2):A195, 2001
Increased Oxidative
Stress Is Associated
With Elevated
Plasma Levels of
Adrenomedullin in
Hypertensive
Patients With Type 2
Diabetes
P
revious studies have demonstrated
that oxidative stress is associated not
only with hyperglycemia and hyper-
tension but also with the metabolic syn-
drome and cadiovascular disease (13).
Adrenomedullin (AM) is a novel va-
sorelaxant peptide isolated from human
pheochromocytoma (4,5). Oxidative
stress enhances AM production from en-
dothelium and vascular smooth muscle
cells in vitro (6,7). However, whether ox-
idative stress is associated with circulating
levels of AM in vivo is unknown. In the
present study, we evaluated the relation-
ship between the plasma levels of 8-
epi-prostaglandinF2(8-epi-PGF2;
currently regarded as the most reliable
marker for the assessment of oxidative
stress in humans) (8,9) and AM in normal
subjects and hypertensive patients with
type 2 diabetes.
This study comprised 17 hyperten-
sive patients with type 2 diabetes (15 men
and 2 women, age 47.3 3.0 years
[mean SE], BMI 23.2 0.8 kg/m
2
, fast-
ing plasma glucose 8.2 0.5 mmol/l,
HbA
1c
9.9 0.5%, fasting serum insulin
30.6 3.0 pmol/l, systolic blood pres-
sure 150.2 3.4 mmHg, and diastolic
blood pressure 76.5 2.2 mmHg) and
18 normal subjects (17 men and 1
women, age 43.0 1.9 years, BMI 23.8
0.4 kg/m
2
, fasting plasma glucose 5.1
0.2 mmol/l, fasting serum insulin 31.2
3.6 pmol/l, systolic blood pressure
126.8 2.2 mmHg, and diastolic blood
pressure 79.8 1.6 mmHg). All subjects
were nonsmokers. Hypertensive patients
with type 2 diabetes were diagnosed at a
local clinic 3.0 0.2 years before the be-
ginning of this study. All patients were
treated with diet (1,4401,720 kcal/day
and sodium restriction 304 mmol/day)
and exercise (walking 10,000 steps/day);
none of them were receiving any kind of
drug. Their blood glucose and blood pres-
sure were in good control (HbA
1c
6.5%,
systolic blood pressure 140 mmHg, and
diastolic blood pressure 90 mmHg)
during the initial several months. How-
ever, thereafter the blood glucose and
blood pressure of the patients gradually
increased (HbA
1c
8%, systolic blood
pressure 140 mmHg, and diastolic
blood pressure 90 mmHg) because of
overeating and inactivity. They were re-
ferred to our clinical department for con-
trol of hyperglycemia and hypertension.
None of them had evidence of micro- or
macroangiopathy. Informed consent was
obtained from all subjects before the be-
ginning of the study. Plasma levels of free
8-epi-PGF2were measured using a
commercially available enzyme immuno-
assay kit (Cayman Chemical, Ann Arbor,
MI). The detection limit of this assay was
1.5 pg/ml, and the intra- and interassay
coefcients of variation were 7.5 and
9.2%, respectively. AM in plasma samples
was measured using a commercially avail-
able immunoradiometric assay kit
(Shionogi Pharmaceuticals, Osaka, Ja-
pan). The detection limit of this assay was
2 fmol/ml, and the intra- and interassay
coefcients of variation were 7.0 and
6.9%. Serum insulin was measured using
an immunoradiometric assay kit (Insulin
Riabead II kit; Dainabot, Tokyo). The in-
tra- and interassay coefcients of varia-
tion of the assay were 1.9 and 2.0%. In
addition, we measured blood pressure in
supine position after a 5-min rest.
Both plasma levels of 8-epi-PGF2
and AM were signicantly increased in
hypertensive patients with type 2 diabetes
compared with normal subjects (8-epi-
PGF248.6 8.6 vs. 11.9 1.3 pg/ml,
P0.05; AM 14.8 0.7 vs. 12.4 0.2
fmol/ml, P0.02). The plasma levels of
8-epi-PGF2were proportionally corre-
lated with AM (r0.696, P0.01) in
only hypertensive patients with type 2 di-
abetes. Signicant positive correlations
were observed between plasma levels of
Letters
1642 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
8-epi-PGF2(r0.540, P0.05) or
AM (r0.875, P0.001) and systolic
blood pressure in patients with type 2 di-
abetes.
This is the rst report that demon-
strated a relationship between oxidative
stress and AM in vivo. The mechanism by
which plasma levels of 8-epi-PGF2cor-
relate with AM and the cellular source of
AM remain unknown. Previous studies
have shown that oxidative stress stimu-
lates secretion of AM from endothelium
and vascular smooth muscle cells and that
AM mRNA expression is increased by ac-
tivation of the nuclear factor-B pathway
(6,7). Increased AM secretion from endo-
thelium and vascular smooth muscle may
inuence the plasma levels of AM. On the
other hand, Shimosawa et al. (10) re-
ported that endogenous AM may protect
from organ damage by inhibiting oxida-
tive stress production. Increased AM lev-
els may compensate for oxidative stress
induced vasoconstriction and thus may
play a protective role against organ injury.
Adrenal medulla and pancreatic islets
may also be a source of AM (4,11). Fur-
ther study is needed to clarify the source
of plasma AM and the mechanism of cor-
relation between oxidative stress and AM.
In conclusion, there was a signicant
positive correlation between increased
oxidative stress and elevated plasma lev-
els of AM in hypertensive patients with
type 2 diabetes. Enhanced oxidative stress
may regulate the plasma levels of AM in
hypertensive patients with type 2 diabetes.
AKIRA KATSUKI,
MD
1
YASUHIRO SUMIDA,
MD
1
HIDEKI URAKAWA,
MD
1
ESTEBAN C. GABAZZA,
MD
1
NORIKO MARUYAMA,
MD
1
KOHEI MORIOKA,
MD
1
NAGAKO KITAGAWA,
MD
1
YASUKO HORI,
MD
1
KANAME NAKATANI,
MD
2
YUTAKA YANO,
MD
1
YUKIHIKO ADACHI,
MD
1
From the
1
Third Department of Internal Medicine,
Mie University School of Medicine, Mie, Japan; and
the
2
Department of Laboratory Medicine, Mie Uni-
versity School of Medicine, Mie, Japan.
Address correspondence to A. Katsuki, MD,
Third Department of Internal Medicine, Mie Univer-
sity School of Medicine, 2-174 Edobashi, Tsu, Mie
514-8507, Japan. E-mail: katuki-a@clin.medic.mie-
u.ac.jp.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Ceriello A, Bortolotti N, Motz E, Crescen-
tini A, Lizzio S, Russo A, Tonutti L,
Taboga C: Meal-generated oxidative stress
in type 2 diabetic patients. Diabetes Care
21:15291533, 1998
2. Yasunari K, Maeda K, Nakamura M, Yo-
shikawa J: Oxidative stress is a possible
link between blood pressure, blood glu-
cose, and C-reacting protein. Hypertension
39:777780, 2002
3. Evans JL, Goldne ID, Maddux BA, Grod-
sky GM: Oxidative stress and stress-acti-
vated signaling pathways: a unifying
hypothesis of type 2 diabetes. Endocr Rev
23:599622, 2002
4. Kitamura K, Kangawa K, Kawamoto M,
Ichiki Y, Nakamura S, Matsuo H, Eto T:
Adrenomedullin: a novel hypotensive
peptide isolated from human pheochro-
mocytoma. Biochem Biophys Res Commun
192:553560, 1993
5. Kinoshita H, Kato K, Kuroki M, Naka-
mura S, Kitamura K, Hisanaga S, Fuji-
moto S, Eto T: Plasma adrenomedullin
levels in patients with diabetes (Letter).
Diabetes Care 23:253254, 2000
6. Chun T-H, Itoh H, Saito T, Yamahara K,
Doi K, Mori Y, Ogawa Y, Yamashita J,
Tanaka T, Inoue M, Masatsugu K, Sawada
N, Fukunaga Y, Nakao K: Oxidative stress
augments secretion of endothelium-de-
rived relaxing peptides, C-type natriuretic
peptide and adrenomedullin. J Hypertens
18:575580, 2000
7. Ando K, Ito Y, Kumada M, Fujita T: Oxi-
dative stress increases adrenomedullin
mRNA levels in cultured rat vascular
smooth muscle cells. Hypertens Res 21:
187191, 1998
8. Sampson MJ, Gopaul N, Davies IR,
Hughes DA, Carrier MJ: Plasma F2 iso-
prostanes, direct evidence of increased
free radical damage during acute hyper-
glycemia in type 2 diabetes. Diabetes Care
25:537541, 2002
9. Gopaul NK, Manraj MD, Hebe A, Kwai
Yan SL, Johnston A, Carrier MJ, Anggard
EE: Oxidative stress could precede endo-
thelial dysfunction and insulin resistance
in Indian Mauritians with impaired glucose
metabolism. Diabetologia 44:706712, 2001
10. Shimosawa T, Shibagaki Y, Ishibashi K,
Kitamura K, Kangawa K, Kato S, Ando K,
Fujita T: Adrenomedullin, an endogenous
peptide, counteracts cardiovascular dam-
age. Circulation 105:106111, 2002
11. Katsuki A, Sumida Y, Gabazza EC, Mu-
rashima S, Urakawa H, Morioka K, Kita-
gawa N, Tanaka T, Araki-Sasaki R, Hori Y,
Nakatani K, Yano Y, Adachi Y: Acute hyper-
insulinemia is associated with increased
circulating levels of adrenomedullin in
patients with type 2 diabetes mellitus. Eur
J Endocrinol 147:7175, 2002
Renin-Angiotensin
System Gene
Polymorphisms and
Retinopathy in
Chinese Patients
With Type 2
Diabetes
D
iabetic eye disease is the leading
cause of new cases of blindness in
many developed countries, for
which macular edema and proliferative
retinopathy are major causes of loss of vi-
sion (1). Genetic studies have previously
suggested that diabetic retinopathy is
partly determined by genetic factors (2).
Angiotensin II, produced locally within
the retina, has potent hemodynamic and
growth-promoting effects and stimulates
ocular neovascularization (3,4). The pres-
ence of retinopathy is associated with an
activated renin-angiotensin system (RAS)
(5), and inhibition of the RAS signi-
cantly reduced the progression of reti-
nopathy in nonhypertensive patients with
type 1 diabetes (6). The ACE gene inser-
tion/deletion (I/D) polymorphism has
been reported to be associated with reti-
nopathy in type 1 diabetic subjects, but
the ndings are heterogeneous (7,8).
Only limited data are available describing
the angiotensinogen (AGT) M235T and
the angiotensin II type 1 receptor (AT
1
R)
A1166C polymorphisms and retinopathy
(9,10).
In this study, we examined the asso-
ciations between these three polymor-
phisms and diabetic retinopathy in 827
Chinese type 2 diabetic patients (based on
1985 World Health Organization crite-
ria). All subjects gave written informed
consent and were of Han Chinese origin.
They were examined by the physicians or
ophthalmologists through dilated pupils.
Retinopathy was considered to be present
if there was one or more areas of hemor-
rhages, microaneurysms, cotton wool
spots, and/or laser coagulation scars re-
lated to diabetic retinopathy or history of
vitrectomy (11). The RAS genotype and
allele frequencies were identied using
PCR/restriction fragmentlength poly-
morphism protocols (12) in 326 patients
with retinopathy and 501 diabetic pa-
tients without retinopathy who were
matched for age (59.8 11.4 vs. 60.4
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1643
9.3 years), sex (44.2 female vs. 39.3%
male), disease duration (6.3 [range 5.6
7.0] vs. 6.0 years [5.66.3]), and age of
onset of diabetes (53.2 9.7 vs. 51.9
12.4 years). Of those with retinopathy,
66.6% had nonproliferative, 8.3% pre-
proliferative, and 8.9% proliferative dis-
ease and 16.2% had advanced eye disease.
Of the patients, 30.1% with retinopathy
had received laser therapy in at least one
eye, with 18.7, 29.2, 53.8, and 69.4% be-
ing treated in the groups with increasing
severity of retinopathy.
The allele frequencies were 32.7 and
32.1% for the ACE D allele, 16.1 and
13.8% for the AGT M allele, and 4.6 and
3.4% for the AT
1
R C allele in those with-
out and with retinopathy, respectively.
The genotype distributions for these gene
polymorphisms are described in Table 1.
No differences were identied in the ge-
notype or allele frequencies between the
groups either for the dichotomous classi-
cation of retinopathy or when the sever-
ity of the retinopathy was graded.
The ACE D allele has been associated
with increasing ACE levels (12). Subjects
with the angiotensinogen M235T poly-
morphism TT genotype have increased
serum AGT levels, while the AT
1
R 1166C
allele is associated with enhanced respon-
siveness to angiotensin II (13). The ACE
DD genotype was found to be associated
with the presence of proliferative retinop-
athy in a relatively small case-control
study of type 1 diabetic subjects (7). In
the current study, the analysis compar-
ing age-, sex-, and diabetes duration
matched subjects found no evidence of
associations between the distribution of
genotypes or alleles of these RAS gene
polymorphisms and retinopathy. Our
data support the majority of the literature
regarding the ACE I/D gene polymor-
phism, which has reported no signicant
association with retinopathy in diabetic
patients (7,8).
G. NEIL THOMAS,
PHD
1
JULIAN A.J.H. CRITCHLEY,
FRCP
1
BRIAN TOMLINSON,
FRCP
1
VINCENT T.F. YEUNG,
MD
1
DENNIS LAM,
MD
2
CLIVE S. COCKRAM,
FRCP
1
JULIANA C.N. CHAN,
FRCP
1
From the
1
Department of Medicine and Therapeu-
tics, The Chinese University of Hong Kong, The
Prince of Wales Hospital, Shatin, Hong Kong; and
the
2
Department of Ophthalmology and Visual Sci-
ence, The Chinese University of Hong Kong, The
Prince of Wales Hospital, Shatin, Hong Kong.
Address correspondence to G. Neil Thomas, De-
partment of Medicine and Therapeutics, The Prince
of Wales Hospital, Shatin, Hong Kong. E-mail:
gneilthomas@yahoo.co.uk.
J.A.J.H.C. is deceased.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Klein R, Klein BEK, Moss SE: Visual im-
pairment in diabetes. Ophthalmology 91:
19, 1984
2. Leslie RD, Pyke DA: Diabetic retinopathy
in identical twins. Diabetes 31:1921, 1982
3. Fernandez LA, Twickler J, Mead A: Neo-
vascularisation produced by angiotensin
II. J Lab Clin Med 105:141145, 1985
4. Danser AH, Derkx FH, Admiraal PJ, Dei-
num J, de Jong PT, Schalekamp MA:
Angiotensin levels in the eye. Invest Oph-
thalmol Vis Sci 35:10081018, 1994
5. Migdalis IN, Iliopoulou V, Kalogeropou-
lou K, Koutoulidis K, Samartzis M:
Elevated serum levels of angiotensin-con-
verting enzyme in patients with diabetic
retinopathy. South Med J 83:425427, 1990
6. Chaturvedi N, Sjolie A-K, Stephenson JM,
Abrahamian H, Keipes M, Castellarin A,
Rogulja-Pepeonik Z, Fuller J, the EUCLID
Study Group: Effect of lisinopril on pro-
gression of retinopathy in normotensive
people with type 1 diabetes. Lancet 351:
2831, 1998
7. Rabensteiner D, Abrahamian H, Irsigler
K, Hermann KM, Kiener HP, Mayer G,
Kaider A, Prager R: ACE gene polymor-
phism and proliferative retinopathy in
type 1 diabetes: results of a case-control
study. Diabetes Care 22:15301535, 1999
8. Fujisawa T, Ikegami H, Kawaguchi Y,
Hamada Y, Ueda H, Shintani M, Fukuda
M, Ogihara T: Meta-analysis of associa-
tion of insertion/deletion polymorphism
of angiotensin I-converting enzyme gene
with diabetic nephropathy and retinopa-
thy. Diabetologia 41:4753, 1998
9. Tarnow L, Cambien F, Rossing P, Nielsen
FS, Hansen BV, Ricard S, Poirer O, Parv-
ing HH: Angiotensin II type 1 receptor
gene polymorphism and diabetic micro-
angiopathy. Nephrol Dial Transplant 11:
10191023, 1996
10. Staessen JA, Kuznetsova T, Wang JG,
Emelianov D, Vlietinck R, Fagard R:
M235T angiotensinogen gene polymor-
phism and cardiovascular renal risk. J Hy-
pertens 17:917, 1999
11. Ko GTC, Chan JCN, Lau MSW, Cockram
CS: Diabetic microangiopathic complica-
tions in young Chinese diabetic patients: a
clinic-based cross-sectional study. J Dia-
betes Complications 13:300306, 1999
12. Thomas GN, Critchley JAJH, Tomlinson
B, Lee ZSK, Young RP, Chan JCN: Al-
buminuria and the renin-angiotensin
system gene polymorphisms in type 2
diabetic and in normoglycaemic hyper-
tensive Chinese. Clin Nephrol 55:715,
2001
13. Peters J: Molecular basis of human hyper-
tension: the role of angiotensin. Baillieres
Clin Endocrinol Metab 9:657678, 1995
Prevalence of Type 1
DiabetesRelated
Autoantibodies in
Adults With Celiac
Disease
S
tudies have shown a 4% prevalence
of celiac disease in type 1 diabetic
patients. Few data, however, are
available on the prevalence of type 1 dia-
betesrelated antibodies in patients with
celiac disease, with studies being limited
by the recruitment of low numbers of
children only (1,2). We have assessed
prevalence of type 1 diabetesrelated au-
toantibodies (islet cell antibody [ICA],
GAD antibody [GADA], and antibodies to
the protein tyrosine phosphataserelated
IA-2 [anti-IA2]) in a cohort of 378 nondi-
abetic adults with celiac disease (89 un-
treated and 289 treated with a gluten-free
diet), aged 33.9 14.7 years (range
15.781.3), who were cared for at our
outpatient clinic. Known duration of glu-
Table 1Genotype distributions for the ACE I/D, AGT M235T, and angiotensin type 1
receptor A1166C gene polymorphisms in type 2 diabetic patients without and with retinopathy
Genotypes
No
retinopathy
Retinopathy
(total) Nonproliferative
Advanced retinopathy
(pre, proliferative,
advanced)
n501 326 225 101
ACE DD/ID/II 11.6/42.3/46.1 12.3/39.6/48.2 11.4/37.8/50.7 14.9/42.6/42.6
AGT TT/MT/MM 69.7/28.5/1.7 74.2/24.0/1.8 75.0/23.8/1.2 77.0/20.7/2.3
AT
1
R AC/AA 9.2/90.8 6.6/93.4 8.1/91.9 6.5/93.5
No signicant differences were identied between the groups.
Letters
1644 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
ten withdrawal was 5 years in 146
(48.2%), 59 years in 54 (17.8%), and
9 years in 103 (34.0%) patients. GADAs
were measured by a radioligand assay us-
ing human recombinant GAD 65 as anti-
gen (Medipan Diagnostica, Selchow,
Germany), ICAs by indirect immunou-
orescence on frozen sections of human
blood group 0 pancreas with uorescein
isothiocyanate-conjugated rabbit anti-
bodies, and anti-IA2 by a radioligand as-
say using highly puried human
recombinant IA2 labeled with
125
I (Medi-
pan Diagnostica).
Of 289 treated nondiabetic patients,
26 had type 1 diabetesrelated autoanti-
bodies, giving a prevalence of 9.0% (95%
CI 6.712.3). All of them, however,
showed no more than one marker positiv-
ity (2.8% ICA, 3.1% GADA, 3.1% anti-
IA2). Untreated celiac patients had a
similar prevalence (10.0%, P0.75). In
linear regression analysis, levels of anti-
IA2 were linearly associated with dura-
tion of celiac disease (␤⫽0.002, P
0.05). In logistic regression analysis, du-
ration of gluten withdrawal was indepen-
dently associated with a prevalence of
type 1 diabetesrelated autoantibodies.
With respect to duration 5 years, a four-
fold increased risk in patients with dura-
tion 9 years was found (95% CI 1.51
10.6), even after adjustment for age, sex,
presence of other autoimmune diseases,
and compliance to diet. In a mean fol-
low-up time of 3.0 1.3 years (range
1.36.5), however, no incident case of
type 1 diabetes was diagnosed in either
patients with (79.0 person-years) or with-
out (782.7 person-years) autoimmune
markers.
In conclusion, this study shows that
prevalence of type 1 diabetesrelated au-
toantibodies in adults with celiac disease
is high (9.0%), even after dietary gluten
withdrawal, and that it increases over
time but is associated with low risk of pro-
gression to diabetes. These ndings are
consistent with a role of common genetic
susceptibility of both diseases, such as
factors involved in intestinal permeabil-
ity. Case-control studies show association
between various dietary factors and risk of
type 1 diabetes (3). Dietary gluten could
act as modier rather than determinant of
type 1 diabetes, facilitating the progres-
sion of other dietary factors to the lamina
propria, where they activate the autoim-
mune response against -cells.
GRAZIELLA BRUNO,
MD
SILVIA PINACH,
MD
SILVIA MARTINI,
MD
MAURIZIO CASSADER,
MD
GIANFRANCO PAGANO,
MD
CARLA SATEGNA GUIDETTI,
MD
From the Department of Internal Medicine, Univer-
sity of Torino, Torino, Italy.
Address correspondence to Dr. Graziella Bruno,
Department of Internal Medicine, Turin University,
corso Dogliotti 14, I-10126 Torino, Italy. E-mail:
graziella.bruno@katamail.com.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Di Mario U, Anastasi E, Mariani P, Ballati
G, Perfetti R, Triglione P, Morellini M,
Bonamico M: Diabetes-related autoanti-
bodies do appear in children with coeliac
disease. Acta Paediatr 81:593597, 1992
2. Galli-Tsinopoulou A, Nousia-Arvanitakis
S, Dracoulacos D, Xefteri M, Karamouzis
M: Autoantibodies predicting diabetes
mellitus type 1 in celiac disease. Horm Res
52:119124, 1999
3. Dahlquist GG, Blom LG, Persson LA, Sad-
strom AI, Wall SG: Dietary factors and the
risk of developing insulin dependent dia-
betes in childhood. BMJ 300:13021306,
1990
Glycemic Relapse
After an Intensive
Outpatient
Intervention for Type
2 Diabetes
T
he effectiveness of interventions that
reduce hyperglycemia in patients
with diabetes (1,2) is limited by the
tendency for glycemic relapse after the in-
tervention ends. We sought to character-
ize the occurrence of glycemic relapse
after initial improvement and to describe
predictors of relapse.
Glycemic relapse was evaluated in
265 consecutive patients with type 2 dia-
betes who participated in a 3-month in-
tensive outpatient intervention and were
followed at least 1 year after completion of
the intervention. Details of the interven-
tion have been previously described (3).
All had HbA
1c
8% before the interven-
tion and had achieved marginal glycemic
control (nadir HbA
1c
8%) after the in-
tervention. Glycemic relapse was dened
as a subsequent HbA
1c
8% and an ab-
solute increase of at least 1% above the
postintervention nadir HbA
1c
. All data
were analyzed using SAS 8.12 (SAS Insti-
tute, Cary, NC).
The mean SD age was 56 13.4
years; 54% were women; 71% were Cau-
casian; and 27% were African American.
The mean duration of diabetes was 6.4
8.4 years. Mean BMI was 32.2 8.6 kg/
m
2
. The mean HbA
1c
before interven-
tion was 10.1 1.7%. The mean HbA
1c
nadir after intervention was 6.8 0.7%.
Twenty-ve percent were receiving insu-
lin therapy before the intervention, and
an additional 25% initiated insulin during
the program.
The cumulative incidence of relapse
at 1 year was 25%. The initiation of insu-
lin therapy during the intervention was
the only identiable independent predic-
tor of relapse (hazard ratio 1.96, 95% CI
1.023.74). Female sex, African-
American race, duration of diabetes, lack
of weight loss during the intervention pe-
riod, and the levels of HbA
1c
before the
intervention and at nadir after the inter-
vention increased the risk of relapse in
univariate analysis, but these associations
were not statistically signicant in multi-
variate modeling. For those patients who
relapsed, the median time to relapse was 9
months. Kaplan-Meier plots estimated
that 50% of the population would relapse
by 30.3 months.
Twenty-ve percent of type 2 patients
who attained satisfactory glycemic con-
trol after an intensive outpatient interven-
tion relapsed within a year. Patients who
initiated insulin therapy during the inter-
vention had an almost double risk of re-
lapse. Future studies should characterize
social and behavioral variables as well as
the frequency and type of insulin in order
to better understand the relapse process.
In the interim, this high-risk subgroup
should receive priority for continuation of
intensive care or other relapse preventive
measures.
AcknowledgmentsThis study was sup-
ported in part by NIDDK P60DK20593, the
American Diabetes Association, and the Rob-
ert Wood Johnson Foundation.
TOM A. ELASY,
MD, MPH
1
ALAN L. GRABER,
MD
1
KATHLEEN WOLFF,
RN, MSN
2
ANNE BROWN,
RN, MSN
2
AYUMI SHINTANI,
PHD
1
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1645
From the
1
Department of Medicine, Vanderbilt Uni-
versity School of Medicine, Nashville, Tennessee;
and
2
Vanderbilt University School of Nursing,
Nashville, Tennessee.
Address correspondence to Tom A. Elasy, S1121
MCN, Department of Medicine, Vanderbilt Univer-
sity School of Medicine, Nashville, TN 37232-2587.
E-mail: tom.elasy@mcmail.vanderbilt.edu.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. The Diabetes Control and Complications
Trial Research Group: The effect of inten-
sive treatment of diabetes on the develop-
ment and progression of long-term
complications in insulin-dependent dia-
betes mellitus. N Engl J Med 329:977986,
1993
2. UK Prospective Diabetes Study (UKPDS)
Group: Intensive blood-glucose control
with sulphonylureas or insulin compared
with conventional treatment and risk of
complications in patients with type 2 di-
abetes (UKPDS 33). Lancet 352:837853,
1998
3. Graber AL, Elasy TA, Quinn D, Wolff K,
Brown A: Improving glycemic control in
adults with diabetes mellitus: shared re-
sponsibility in primary care practices.
South Med J 95:684690, 2002
Gestational Glucose
Intolerance in
Multiple Pregnancy
T
win pregnancies have been associ-
ated with adverse obstetric and neo-
natal outcomes. The incidence of
respiratory distress syndrome in the new-
borns of multiple pregnancy patients is
probably related to the higher incidence
of preterm delivery. However, the com-
plications of twin pregnancies are not
limited to infants; miscarriages, pre-
eclampsia, anemia, placenta previa, poly-
hydramnios, and premature rupture of
membranes are more frequent than in sin-
gleton pregnancy. There have been a few
reports that test the hypothesis that mul-
tiple gestation could also be a risk factor
for gestational diabetes (16). These
studies utilized various criteria for diag-
nosing gestational diabetes (World
Health Organization [1], National Diabe-
tes Data Group [24], and Carpenter and
Coustan [7]) and did not report the per-
centage of the positive glucose challenge
test for each group.
To determine the prevalence of gesta-
tional diabetes (using the Carpenter and
Coustan criteria) in singleton and twin
pregnancies in a cohort population of
women who had had screening for ges-
tational diabetes from January 1990 to
January 2000, we started a retrospec-
tive study on 2,554 pregnant women
screened for gestational diabetes in the
Department of Obstetrics and Gynecol-
ogy of our University Hospital. Of these,
70 were multiple pregnancies. A 50-g oral
glucose challenge test was performed be-
tween 24 and 28 weeksgestation, and a
1-h value 135 mg/dl was considered
positive and followed by a 100-g oral glu-
cose tolerance test. The results were inter-
preted according to the Carpenter and
Coustan threshold values (95, 180, 155,
and 140 mg/dl) at fasting and after 1, 2,
and 3 h, respectively. If two thresholds
were met or exceeded, then the diagnosis
of gestational diabetes was made. Age, fa-
milial or personal anamnestic factors, par-
ity, and BMI at rst visit, all considered
risk factors for GDM, were recorded in
our database. A positive glucose challenge
test occurred in 520 of 2,554 pregnant
women (20.3%). The test was positive in
22 of 70 (31.4%) multiple pregnancies
and 498 of 2,484 (20.0%) singleton preg-
nancies (P0.029). The rates of GDM
were 80 of 2,484 (3.6%) in singleton and
4 of 70 (5.7%) in twin pregnancies (P
0.416). No statistically signicant differ-
ences exist for the maternal risk factors for
GDM between the two groups.
The decreased insulin sensitivity in
pregnancy may be modied by several
factors, such as diet, BMI, maternal age,
and the placental mass, all of which may
play a role affecting -cell function and
sensitivity to insulin. It has been sug-
gested that in multiple pregnancies with
two placentas or one that is larger, the
incidence of gestational diabetes may be
increased (3,4,6). Our data show that in
our Sicilian population, twin pregnancy
cannot be considered a proven risk factor
for gestational diabetes. In fact, the differ-
ences (3.6 vs. 5.7%) are not statistically
different. Our data showed that one-third
of twin pregnancies (22 of 70, 31.4%)
manifest a positive glucose challenge test,
as compared with 20% of singleton preg-
nancies (498 of 2,484). It seems possible
that with a larger sample size, the differ-
ence in incidence of gestational diabetes
might have been statistically signicant,
although of questionable clinical signi-
cance. Another hypothesis could be that
the greater relative elevation of anti-
insulin placental hormone levels in a twin
gestation may precipitate a mild degree of
glucose intolerance in a woman who
would have been normal with a singleton
pregnancy. If this were the case, the im-
pact of such a mild degree of glucose in-
tolerance remains undetermined.
AcknowledgmentsThe authors thank
Prof. D.R. Coustan (Brown University, Provi-
dence, RI) for critical reading of the manu-
script.
FRANCESCO CORRADO,
MD
FRANCESCO CAPUTO,
MD
GRAZIELLA FACCIOLA` ,
MD
ALFREDO MANCUSO,
MD
From the Department of Obstetrics and Gynecol-
ogy, University of Messina, Messina, Italy.
Address correspondence to F. Corrado, MD, De-
partment of Obstetrics and Gynecology, Policlinico
Universitario G. Martino98100, Messina, Italy. E-
mail: francesco.corrado@unime.it.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Wein P, Warwick MM, Beisher NA: Ges-
tational diabetes in twin pregnancy: prev-
alence and long-term implications. Aust
NZ J Obstet Gynecol 32:325327, 1992
2. Henderson CE, Scarpelli S, La Rosa D, Di-
von MY: Assessing the risk of gestational
diabetes in twin gestation. J Natl Med As-
soc 87:757758, 1995
3. Roach VJ, Lau TK, Wilson D, Rogers MS:
The incidence of gestational diabetes in
multiple pregnancy. Aust NZ J Obstet Gy-
necol 38:5657, 1998
4. Schwartz DB, Daoud Y, Zazula P, Goyert
G, Bronsteen R, Wright D, Copes J: Ges-
tational diabetes mellitus: metabolic and
blood glucose parameters in singleton
versus twin pregnancies. Am J Obstet Gy-
necol 181:912914, 1999
5. Egeland GM, Irgens LM: Is multiple birth
pregnancy a risk factor for gestational di-
abetes? (Letter). Am J Obstet Gynecol 185:
12751276, 2001
6. Sivan E, Maman E, Homko CJ, Lipitz S,
Cohen S, Schiff E: Impact of fetal reduc-
tion on the incidence of gestational diabe-
tes. Obstet Gynecol 99:9194, 2002
7. Carpenter MW, Coustan DR: Criteria for
screening test for gestational diabetes.
Am J Obstet Gynecol 144:768773, 1982
Letters
1646 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
Cytokine Milieu
Tends Toward
Inammation in
Type 2 Diabetes
I
t has been postulated that type 2 dia-
betes is a manifestation of the inam-
matory host response (1). Increased
inammatory activity is believed to play
a critical role in development of athero-
genesis and to predispose established
atherosclerotic plaques to rupture (2). In-
terleukin (IL)-8 is a potent chemoattractant
(chemokine) and induces recruitment of
neutrophils and T-cells into the subendo-
thelial space, as well as adhesion of mono-
cytes to endothelium (3). IL-18 is a potent
proinammatory cytokine reported to
play a role in plaque destabilization and to
predict cardiovascular death in patients
with coronary artery disease (4). Adi-
ponectin is an adipocyte-derived plasma
protein (adipokine) that accumulates in
the injured artery and has potential anti-
atherogenic properties; male patients
with hypoadiponectinemia present a two-
fold increase in coronary artery disease
prevalence (5).
We studied 30 patients (age 42 4
years, BMI 26.9 1.2 kg/m
2
, and HbA
1c
7.2 0.6% [mean SD]) with newly
diagnosed type 2 diabetes (within 6
months of diagnosis) without hyperten-
sion or evidence of vascular complica-
tions treated only by diet. Thirty healthy
subjects, matched for age, sex, and body
weight, served as the control group. None
of the subjects (diabetic and nondiabetic)
were taking any drugs. Serum samples for
IL-8, IL-18, and adiponectin were stored
at 80°C and measured in duplicate us-
ing enzyme-linked immunosorbent kits.
All samples for a given patient were ana-
lyzed in the same series.
Circulating levels of IL-8 and -18 con-
centrations were higher in diabetic pa-
tients (13.7 2.8 and 205 39 pg/ml,
respectively) than in control subjects
(8.7 1.9 and 120 25 pg/ml, P
0.050.001), while circulating adiponec-
tin levels were lower (4.9 1.2 vs. 7.1
1.9 g/ml, P0.01). There was a signif-
icant correlation between fasting glucose
and IL-8 levels in diabetic patients (r
0.31, P0.05); adiponectin concentra-
tions were negatively correlated with fast-
ing insulin levels (r⫽⫺0.43, P0.01).
Our study shows that circulating IL-8
concentrations are signicantly higher
and adiponectin levels are signicantly
lower in type 2 diabetic patients than
matched control subjects. Previous stud-
ies reported similar ndings (6,7). To our
knowledge, this is the rst demonstration
that circulating IL-18 concentrations are
increased in type 2 diabetic patients, as
compared with age-, sex-, and body
weightmatched nondiabetic subjects. In
a prospective study of 1,229 subjects, in-
cluding one-sixth of diabetic patients,
with documented coronary artery dis-
ease, serum IL-18 concentration was
identied as a strong independent predic-
tor of future cardiovascular events (5).
The correlation we found between IL-18
and glucose levels in diabetic patients and
the evidence that acute hyperglycemia
may increase circulating IL-18 levels in
healthy subjects and IGT patients (8) sug-
gest a role for this cytokine in plaque de-
stabilization associated with stress
hyperglycemia (9).
KATHERINE ESPOSITO,
MD
FRANCESCO NAPPO,
MD, PHD
FRANCESCO GIUGLIANO
CARMEN DIPALO
MYRIAM CIOTOLA
MICHELANGELA BARBIERI,
MD
GIUSEPPE PAOLISSO,
MD
DARIO GIUGLIANO,
MD, PHD
From the Department of Geriatrics and Metabolic
Diseases, Second University of Naples, Naples, Italy.
Address correspondence to Katherine Esposito,
MD, Dipartimento di Geriatria e Malattie del Me-
tabolismo, Policlinico Universitario, Piazza L. Mira-
glia,80138Napoli,Italy.E-mail: katherine.esposito@
unina2.it.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Pickup JC, Mattock MB, Chusney GD,
Burt D: NIDDM as a disease of the in-
nate immune system: association of
acute-phase reactants and interleukin-6
with metabolic syndrome. Diabetologia 40:
12861292, 1997
2. Libby P, Ridker PM, Maseri A: Inamma-
tion and atherosclerosis. Circulation 105:
11351143, 2002
3. Gerzten RE, Garcia-Zepeda EA, Limm YC,
Yoshida M, Ding HA, Gimbrone Jr MA,
Luster AD, Luscinskas FW, Rosenzweig
A: MCP-1 and IL-8 trigger rm adhesion
of monocytes to vascular endothelium
under ow conditions. Nature 398:718
723, 1999
4. Blankenberg S, Tiret L, Bickel C, Peetz D,
Cambien F, Meyer J, Rupprecth HJ: Inter-
leukin-18 is a strong predictor of cardio-
vascular death in stable and unstable
angina. Circulation 106:2430, 2002
5. Kumada M, Kihara S, Sumitsuji S,
Kawamoto T, Matsumoto S, Ouchi N,
Arita Y, Okamoto Y, Shimomura I,
Hiraoka H, Nakamura T, Funahashi T,
Matsuzawa Y, the Osaka CAD Study
Group: Association of hypoadiponectine-
mia with coronary artery disease in man.
Arterioscler Thromb Vasc Biol 23:8589, 2003
6. Zozulin˜ska D, Majchrzak A, Sobieska M,
Wiktorowicz K, Wierusz-Wysocka B:
Serum interleukin-8 levels is increased
in diabetic patients. Diabetologia 42:117
123, 1999
7. Weyer C, Funahashi T, Tanaka S, Hotta K,
Matsuzawa Y, Pratley RE, Tataranni PA:
Hypoadiponectinemia in obesity and type
2 diabetes: close association with insulin
resistance and hyperinsulinemia. J Clin
Endocrinol Metab 86:19301935, 2001
8. Esposito K, Nappo F, Marfella R, Giugli-
ano G, Giugliano F, Ciotola M, Quagliaro
L, Ceriello A, Giugliano D: Inammatory
cytokine concentrations are acutely in-
creased by hyperglycemia in humans: role
of oxidative stress. Circulation 106:2067
2072, 2002
9. Capes SE, Hunt D, Malberg K, Gerstein
HC: Stress hyperglycemia and increased
risk of death after myocardial infarction in
patients with and without diabetes: a sys-
temic overview. Lancet 355:773778, 2000
Lipoprotein
Abnormalities in
Human Genetic
CD36 Deciency
Associated With
Insulin Resistance
and Abnormal Fatty
Acid Metabolism
C
D36 is an 88-kDa membranous pro-
tein with multiple relevant function
that is widely expressed in human
tissues (1). Because we and others (2)
found human genetic deciency of this
molecule, we have elucidated the molec-
ular bases and pathophysiology. We have
reported that the human deciency may
be associated with insulin resistance (3)
and abnormal dynamics of long-chain
fatty acid (LCFA) (4). The aim of the
present study was to further characterize
lipid and lipoprotein metabolism in the
human genetic CD36 deciency, espe-
cially focusing on postprandial responses.
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1647
Seven Japanese patients with type I
CD36 deciency (cases 17) were sub-
jected. They were divided into the follow-
ing two groups: group A, two younger
patients we have recently identied (cases
1 and 2); and group B, patients 50 years
of age (cases 37, reported in our previ-
ous paper [3]). For the analysis of each
group, aged-matched healthy and nono-
bese volunteers were subjected as control
subjects (Table 1). First, we conrmed
that insulin resistance was also present in
the two younger patients (group A).
At the fasting state, midbands,
which imply the accumulation of rem-
nant lipoproteins, were observed in group
B but not in group A. Preparative ultra-
centrifugation conrmed an increase in
intermediate-density lipoprotein choles-
terol in the all of the CD36-decient sub-
jects in group B compared with the
control subjects. The oral fat loading test
demonstrated that the patients in group B
presented postprandial hyperlipidemia
with the accumulation of small intestine
derived lipids. Whereas the patients in
group A did not show apparent abnor-
malities, though they did have insulin
resistance.
The present study indicated that this
monogenic disorder with the abnormal
metabolism in LCFA and insulin resis-
tance may cause the phenotypic expres-
sion of hyperlipidemia at both fasting and
postprandial states. These data also sug-
gest that the clinical manifestation is af-
fected by the acquired factors, such as
aging and dietary contents. We believe
that human genetic CD36 deciency may
be a monogenic form of metabolic syn-
drome.Further studies to elucidate mo-
lecular mechanism for the phenotypic
expression in CD36-decient states are
under way in our laboratories.
AcknowledgmentsThis work was sup-
ported by grants-in-aid to K.H. (no.
13671191) and S.Y. (nos. 11557055 and
10671070) from the Ministry of Education,
Science, Sports, and Culture of Japan.
We gratefully thank Natsu Iwamoto, Sonoe
Shima, and Atsuko Ohya for the skillful tech-
nical assistance.
TAKAHIRO KUWASAKO,
MD
1
KEN-ICHI HIRANO,
MD, PHD
1
NAOHIKO SAKAI,
MD, PHD
1
MASATO ISHIGAMI,
MD, PHD
1
HISATOYO HIRAOKA,
MD, PHD
1
MOHAMED JANABI YAKUB,
MD, PHD
1
KEIKO YAMAUCHI-TAKIHARA,
MD, PHD
2
SHIZUYA YAMASHITA,
MD, PHD
1
YUJI MATSUZAWA,
MD, PHD
1
From the
1
Department of Internal Medicine and
Molecular Science, Graduate School of Medicine,
Osaka University, Osaka, Japan; and the
2
Depart-
ment of Molecular Medicine, Graduate School of
Medicine, Osaka University, Osaka, Japan.
Address correspondence to Ken-ichi Hirano,
MD, PhD, Department of Internal Medicine and Mo-
lecular Science, Graduate School of Medicine, B5,
Osaka University, 2-2 Yamadaoka, Suita, Osaka
565-0871, Japan. E-mail: khirano@kb3.so-net.ne.jp.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Febbraio M, Hajjar DP, Silverstein RL:
CD36: a class B scavenger receptor in-
volved in angiogenesis, atherosclerosis,
inammation, and lipid metabolism.
J Clin Invest 108:785791, 2001
2. Yamamoto N, Ikeda H, Tandon NN, Her-
man J, Tomiyama Y, Mitani T, Sekiguchi
S, Lipsky R, Kralisz U, Jamieson GA: A
platelet membrane glycoprotein (GP) de-
ciency in healthy blood donors: Nak
a
,
platelet lack detectable GP IV(CD36).
Blood 76:16981703, 1990
3. Miyaoka K, Kuwasako T, Hirano K,
Nozaki S, Yamashita S, Matsuzawa Y:
CD36 deciency associated with insulin
resistance. Lancet 357:686687, 2001
4. Yoshizumi T, Nozaki S, Fukuchi K, Ya-
masaki K, Fukuchi T, Maruyama T, To-
miyama Y, Yamashita S, Nishimura T,
Matsuzawa Y: Pharmacokinetics and
metabolism of iodine-123-BMIPP fatty
acid analogue in normal and CD36-de-
cient subjects. J Nucl Med 41:11341138,
2000
The Relation
Between Physical
Activity and
Metabolic Control in
Type 2 Diabetes
With <20 Years of
Evolution
O
besity and reduced levels of physi-
cal activity are directly associated
with the insulin resistance that
characterizes type 2 diabetes (1,2). Phys-
ical activity is one of the cornerstones of
type 2 diabetes management (3). Few
Table 1Lipid and lipoprotein abnormalities at fasting and postprandial states in human genetic CD36 deficiency
Group A Group B
Case 1 Case 2 Control (n4) Case 3 Case 4 Case 5 Case 6 Case 7 Control (n8)
Age (years)/sex (M/F) 34/M 32/M 33 2 (4M) 58/M 65/F 54/F 54/F 54/M 55 10 (3M/5F)
BMI (kg/m
2
) 22.9 32.0 22.4 1.0 26.2 22.2 32.8 17.3 24.9 22.7 2.3
WBGU (mg kg
1
min
1
)* 7.8 2.8 9.1 0.5 2.677.095.494.855.298.6 0.5
Fasting state
Total cholesterol (mmol/l) 4.20 5.01 4.78 0.38 5.81 6.18 5.97 5.45 5.84 5.10 0.69
Triglyceride (mmol/l) 0.78 1.23 1.00 0.17 2.73 1.43 1.77 1.28 3.41 1.20 0.38
Midband in PAG disc
electrophoresis
⫹⫹⫹
IDL cholelsterol (mmol/l)§ND ND 0.48 0.38 0.29 0.34 0.22 0.11 0.03
Postprandial state
AUC-TG (mmol/l 6 h) 5.3 8.8 8.8 0.7 20.6 13.4 14.0 14.0 26.8 10.0 2.9
AUC-Apo B48 (AU/ml 6 h)** 223 268 329 107 866 671 941 551 541 360 120
Data are means SD unless otherwise indicated. apo, apolipoprotein; AUC, area under the curve; PAG, polyacrylamide gel; WBGU, mean whole-body glucose
uptake. *Insulin resistance was determined by euglycemic clamp technique (ref. 3); part of data from cases 37 were described previously (Ref. 3); lipoproteins
were analyzed by PAG electrophotesis. §IDL was separated by preparative ultracentrifugation (1.019 d1.063 g/ml); postprandial hyperlipidemia was analyzed
by the oral fat loading test with fat cream containing 30 g fat/m
2
body surface area; TG and apo B48 levels were measured at 0, 2, 4, and 6 h after loading, and AUCs
were calculated; **serum apo B48 levels were measured by the enzyme-linked immunosorbent assay we have developed.
Letters
1648 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
large studies have assessed the impact of
intensity and quality of exercise on meta-
bolic control in these patients, and the
inuence of age on the metabolic re-
sponse to physical activity is still contro-
versial (4). We conducted the Intervention
on Key Cardiovascular Risk Factors in
Type 2 Diabetes National Study (KAREN),
a country-based case-control study nested
in a survey of 1,485 consecutive type 2
diabetic patients treated by 208 endocri-
nologists across Spain to assess the impact
of physical activity on metabolic control.
Patient risk factors, history characteris-
tics, medication and diet compliance, gly-
cemia, fasting HbA
1c
, and lipids were
determined.
Patient characteristics are presented
in Table 1 by quartile of physical activity.
After adjustment for age, sex, triglycer-
ides, BMI, and diet compliance, patients
in the lowest quartile of physical activity
had an OR of 1.52 and a 95% CI of 1.11
2.08 for having an HbA
1c
7.5%. The
ORs for patients 65 years of age and in
the lowest quartile of physical activity
were 1.55 (95% CI 1.04 2.31) compared
with the fourth, 1.64 (1.092.47) with
the third, and 1.84 (1.222.75) with the
second quartile. Patients 65 years of age
and in the lowest quartile of physical ac-
tivity had a similar adjusted risk com-
pared with the rest of quartiles (OR 1.14,
95% CI 0.701.85; 1.38, 0.842.26, and
0.87, 0.531.43, respectively). A signi-
cant reduction of 0.2% (95% CI 0.36 to
0.04) in HbA
1c
was observed for pa-
tients expending 50 kcal/day versus the
rst quartile; the reduction for patients
65 years of age was 0.27% (0.49 to
0.06), but for patients 65 there was no
signicant difference.
These results suggest that physical ac-
tivity produces better glycemic control
and improved cardiovascular risk prole
in diabetic patients, particularly in those
65 years of age.
PEDRO DE PABLOS-VELASCO,
MD, PHD
1
WILFREDO RICART,
MD, PHD
2
SUSANA MONEREO,
MD
3
BASILIO MORENO,
MD, PHD
4
JAUME MARRUGAT,
MD, PHD, FESC
5
ON BEHALF OF THE KAREN INVESTIGATORS
From the
1
Department of Endocrinology, Hospital
Doctor Negrı´n, Las Palmas de Gran Canaria, Spain;
the
2
Department of Endocrinology, Hospital Trueta,
Gerona, Spain; the
3
Department of Endocrinology,
Hospital Getafe, Madrid, Spain; the
4
Department of
Endocrinology, Hospital Gregorio Maran˜on, Ma-
drid, Spain; and the
5
Unit of Lipid and Cardiovas-
cular Epidemiology, Institut Municipal dInvestigacio´
Me`dica (IMIM) and Universitat Auto´noma School
of Medicine, Barcelona, Spain.
Address correspondence to Dr. Jaume Marrugat,
Lipid and Cardiovascular Epidemiology Unit, Insti-
tut Municipal dInvestigacio´Me`dica (IMIM), Carrer
Dr. Aiguader, 80, E-08003 Barcelona, Spain. E-mail:
jmarrugat@imim.es.
© 2003 by the American Diabetes Association.
Appendix
The KAREN Investigators comprise 208
Spanish endocrinologists who work in
primary outpatient diabetes clinics of the
National Health Service.
The KAREN investigators are as fol-
lows: Abella´n T, Acha FJ, Acosta D, Agui-
lar M, Aguillo E, Albero R, Alcaraz M,
Alvarez JA, Alvarez P, Alvarez J, Andia
VM, Aranda S, Arranz A, Arribas LD, Ar-
teaga R, Asensio C, Ayala C, Ballester E,
Barril R, Becerra A, Blanco B, Blay V, Botas
P, Brito MA, Bruscas M, Bueno J, Burgo R,
Cabezas J, Caldero´n D, Calero M, Calvo F,
Ca´ncer E, Carrasco R, Casal F, Castan˜oG,
Castro JC, Cepero D, Cervello G, Chena
JA, Conget I, Corrales JJ, Costilla E, Coves
MJ, Darı´as R, de la Cuesta C, de Pablos P,
Dı´az C, Diez A, Domı´nguez JR, Duran S,
Duran M, Elviro R, Enrı´quez L, Escalada
FJ, Escobar L, Esmatges E, Espiga J, Es-
topin˜an V, Ezquerra R, Famades A, Fer-
na´ndez JI, Ferna´ndez M, Ferna´ndez JM,
Ferna´ndez F, Ferna´ndez T, Ferna´ndez JA,
Ferna´ndez J, Fla´ndez B, Flaquert P, Fraile
AL, Fraile J, Freijanes J, Galvez A, Garcı´a
Table 1Demographic, comorbidity, lifestyle, biochemical, and somatometric characteristics by physical activity levels (quartiles) of type 2
diabetic patients who participated in the KAREN study
51 kcal/day 51148 kcal/day 149264 kcal/day 264 kcal/day P
n368 373 375 369
Age (years) 62 10 61 10 61 96110 NS
Women (%) 49.6 45.3 54.4 69.6 0.001
Time with diabetes (years) 10.3 6.4 9.9 6.3 10.1 6.4 10.3 6.2 NS
Hypertension (%) 51.8 58.3 50.7 46.3 0.017
Current smokers (%)* 19.4 18.9 19.0 16.0 NS
Systolic BP (mmHg) 140 17 139 17 139 16 139 19 NS
Diastolic BP (mmHg) 80 10 79 97910 78 11 NS
BMI (kg/m
2
) 28.8 3.4 28.5 3.5 28.1 3.3 28.3 3.3 0.035
Coronary heart disease (%)17.9 14.4 18.0 14.0 NS
Cerebrovascular disease (%)8.5 6.4 6.6 5.1 NS
Glycemia (mg/dl) 9.5 3.2 9.1 2.9 9.1 3.0 8.8 2.7 0.001
HbA
1c
(%)* 7.9 1.6 7.6 1.4 7.6 1.4 7.4 1.0 0.001
Total cholesterol (mmol/l) 5.3 1.0 5.4 1.0 5.2 1.0 5.1 1.0 0.001
HDL cholesterol (mmol/l) 1.2 0.3 1.3 0.3 1.2 0.3 1.3 0.4 0.014
LDL cholesterol (mmol/l) 3.4 0.9 3.4 0.9 3.2 34.7 3.3 0.9 NS
Triglycerides (mmol/l) 1.9 1.8 1.7 0.9 1.7 1.1 1.5 0.9 0.001
Creatinine (mol/l) 91.9 31.8 85.7 24.7 86.6 26.5 88.4 19.4 0.035
Good diet compliance (%) 37.1 35.1 49.2 52.2 0.001
Daily alcohol consumption (g) 12.9 12.8 12.5 12.3 12.8 13.3 12.5 12.3 NS
Data are means SD and %. *Smokers of at least one cigarette per day in average; myocardial infarction or angina; transient ischemic attack or stroke.
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1649
JA, Garcı´a A, Garcı´a C, Garcı´a G, Garcı´aI,
Garcı´a J, Garcı´a M, Garcia-Hierro V, Gar-
gallo MA, Gentil AC, Gil A, Gilsanz A,
Gime´nez G, Gippini A, Girbe´s AJ, Goday
A, Godoy DV, Goena M, Goicolea I, Go´-
mez F, Gonza´lez A, Gonza´lez JM, Gonza´-
lez O, Gon˜i F, Guerrero E, Herna´ndez C,
Herna´ndez A, Herna´ndez JA, Herna´ndez
C, Herna´ndez A, Herna´ndez C, Iglesias P,
Ikobalceta P, Illan F, In˜igo P, Iriarte A,
Irigoyen L, Jaunsolo MA, Lain T, Levy I,
Llorente I, Lo´pez AJ, Lo´pez de la Torre M,
Lo´pez-Guzman A, Losada F, Luna R, Ma-
canas G, Macia C, Maldonado JA, Mangas
MA, Manzanares JM, Maravall FJ, Marco
A, Marco AL, Marin M, Martı´n T, Martı´n
A, Martı´nez MP, Martı´nez FJ, Martı´nez
MA, Matia P, Mato JA, Megia A, Mene´n-
dez E, Meoro A, Merino JF, Mesa J,
Mezquita P, Micalo T, Miralles JM, Molina
MJ, Monereo S, Morales MJ, Moreno A,
Moreno C, Mories T, Mun˜oz L, Mun˜oz M,
Mun˜oz A, Muros T, Nubiola A, Oleaga A,
Oliva´n B, Palacios N, Palomares R, Pazos
F, Pena E, Pen˜ael FJ, Pereyra F, Pe´rez A,
Pesquera C, Picon MJ, Piedrola G, Pinedo
R, Pinie´s J, Pizarro E, Prieto J, Puig M,
Puigdevall V, Pumar A, Recas I, Recio JM,
Reverter J, Ricart W, Rius F, Rivas M,
Rosell R, Rubio JA, Ruiz JM, Sagarra E,
Sales J, Salinas I, Sa´nchez C, Sa´nchez JC,
Santiago P, Santiago L, Sastre J, Segura P,
Simo R, Simo´n I, Solache I, Soriano J, Soto
A, Suarez-Lledo E, Surruca L, Tebar FJ,
Tenes T, Todolı´ J, Tofe S, Tolosa M, Tor-
ras R, Torres A, Vadillo V, Velasco JL,
Vences LF, Vendrell JJ, Vicente A, Vidal JI,
Villa M, Villar A, Zurro J.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Everhart JE, Pettitt DJ, Bennett PH,
Knowler WC: Duration of obesity in-
creases the incidence of NIDDM. Diabetes
41:235240, 1992
2. American Diabetes Association: Diabetes
mellitus and exercise (Position State-
ment). Diabetes Care 25 (Suppl. 1):S64
S68, 2002
3. Harding AH, Williams DE, Hennings SH,
Mitchell J, Wareham NJ: Is the association
between dietary fat intake and insulin re-
sistance modied by physical activity?
Metabolism 50:11861192, 2001
4. Boule NG, Haddad E, Kenny GP, Wells
GA, Sigal RJ: Effects of exercise on glycemic
control and body mass in type 2 diabetes
mellitus: a meta-analysis of controlled
clinical trials. JAMA 286: 12181227,
2001
Education and
Mortality in Type 2
Diabetes
A
dults with diabetes tend to have
higher age-adjusted mortality rates
(1,2). Evidence from Britain (3)
suggests that mortality in diabetic adults
is inversely related to socioeconomic sta-
tus. The importance of self-care and
blood glucose control in adults with dia-
betes suggests that education may be an
important factor in long-term health out-
comes, a hypothesis that is also consistent
with evidence in the health economics lit-
erature that education increases the ef-
ciency of investment in health (4,5). The
specic hypothesis tested here is that
mortality in adults with diabetes is in-
versely related to education.
The data are taken from the National
Health Interview Survey (NHIS) of 1989,
including its diabetes supplement, with
mortality status taken from the NHIS
Multiple Cause of Death Public Use Data
File, including deaths from 1989 to 1995.
The statistical analysis is applied to those
adults who self-reported that they had
been diagnosed with diabetes, excluding
those with apparent type 1 diabetes (age
of onset 30 years, with insulin depen-
dence, and a BMI 27.2 kg/m
2
for men
and 26.9 for women). The resulting
sample includes 2,387 adults with type 2
diabetes having an average age of 61.8
years.
Logistic regression analysis is used to
explore the relation between mortality
and educational attainment, family in-
come (greater than or less than $20,000
per year in 1989), age, sex, marital status,
race, and duration of diabetes. The statis-
tical analysis is carried out with the svy-
logit procedure in STATA software,
version 7 (STATA, College Station, TX),
using the NHIS sampling weights.
The primary nding is that mortality
is inversely related to education for dia-
betic adults (OR [95% CI] 0.61 [0.40
0.93] for college graduation or higher and
0.78 [0.610.99] for high school gradu-
ation or higher). Mortality is not signi-
cantly related to family income, marital
status, or race, but age, male sex, and du-
ration of diabetes have the expected pos-
itive and statistically signicant associations
with mortality. The nding that educa-
tion reduces the mortality risk of diabetic
adults is consistent with the predictions
from health investment theory that edu-
cation increases the efciency of health
investment. Education may also be a fac-
tor in the relatively poor health status and
outcomes of adults with diabetes (6).
AcknowledgmentsThe author thanks
Philip Jacobs for comments, Glenn Harrison
for generous help with the data, and Paula
Veiga for expert research assistance. Any re-
maining errors are mine.
RONALD P. WILDER,
PHD
From the Department of Economics, Moore School
of Business, University of South Carolina, Colum-
bia, South Carolina.
Address correspondence to Ronald P. Wilder,
PhD, Department of Economics, Moore School of
Business, University of South Carolina, Columbia,
SC 29208. E-mail: ronwildr@darla.badm.sc.edu.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Geiss LS, Herman WH, Smith PJ: Mortal-
ity in non-insulin dependent diabetes. In
Diabetes in America. 2nd ed. National
Diabetes Data Group, National Institutes
of Health, National Institute of Diabetes
and Digestive and Kidney Diseases, 1995
(NIH Publ. no. 95-1468)
2. Gu K, Cowie CC, Harris MI: Mortality in
adults with and without diabetes in a na-
tional cohort of the U.S. population,
19711993. Diabetes Care 21:1138
1145, 1998
3. Roper NA, Bilous RW, Kelly WF, Unwin
NC, Connolly VM: Excess mortality in a
population with diabetes and the impact
of material deprivation: longitudinal,
population based study. BMJ 322:1389
1393, 2001
4. Grossman M, Kaestner R: Effects of edu-
cation on health. In The Social Benefits of
Education. Behrman J, Stacey N, Eds. Ann
Arbor, MI, University of Michigan Press,
1997
5. Kahn ME: Educations role in explaining
diabetic health investment differentials.
Econ Educ Rev 17:257266, 1998
6. Harris MI: Health care and health status
and outcomes for patients with type 2 di-
abetes. Diabetes Care 23:854758, 2000
Stress
Hyperglycemia,
Inammation, and
Cardiovascular Events
O
ne-third of all individuals with hy-
perglycemia admitted to an urban
general hospital do not have a pre-
vious diagnosis of diabetes; in these pa-
Letters
1650 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
tients, hyperglycemia is a risk factor for
adverse outcomes during acute illness (1).
For example, in patients presenting with
acute myocardial infarction, glucose val-
ues in excess of 6.18.0 mmol/l were as-
sociated with a threefold increase in
mortality and a higher risk of heart failure
(2). Control of hyperglycemia with inten-
sive insulin therapy during acute illness
results in marked improvements of clini-
cal outcomes (3).
Dandona et al. (4) put forward a
working hypothesis linking in a feedback
loop of glucose (and possibly other nutri-
ents), insulin, and inammation. Accord-
ing to this paradigm, hyperglycemia has a
proinammatory action that is normally
restrained by the anti-inammatory effect
of insulin secreted in response to that
stimulus. A likely corollary of this para-
digm is that the proinammatory effects
of acute (stress) hyperglycemia may be
implicated in the poor prognosis of hos-
pitalized patients with or without diabe-
tes (particularly death, disability after
acute cardiovascular events, and infec-
tions). We have demonstrated that circu-
lating levels of some proinammatory
cytokines are regulated by glucose levels
in humans (5). Acute hyperglycemia
(15 mmol/l) induced by glucose clamp-
ing in normal subjects and lasting 5 h
determines a signicant increase of inter-
leukin (IL)-6, IL-18, and tumor necrosis
factor-(TNF-) circulating levels. These
effects were more sustained in patients
with impaired glucose tolerance, as well
as following consecutive pulses of intra-
venous glucose, and were annulled by
glutathione, implicating an oxidative
mechanism. Interestingly enough, these
cytokines have been implicated in insulin
resistance (TNF-and IL-6), atheroscle-
rotic plaque destabilization (IL-18), and
future cardiovascular events (IL-6, IL-18,
and TNF-). So, an increased oxidative
stress may be a likely mechanism linking
stress hyperglycemia to cardiovascular
events via an increased cytokine production.
Free fatty acids (FFAs) also induce
proinammatory changes. During illness,
stress increases the concentration of
counterregulatory hormones (mainly glu-
cagon, an epinephrine). Given this back-
ground, it is plausible that in the presence
of high concentrations of both glucose
and FFA, inammation is more promi-
nent. We tested this hypothesis by giving
type 2 diabetic patients two different
meals: a high-carbohydrate meal (high
glucose levels) and a high-fat meal (high
glucose plus high FFA levels). In the latter
condition, the serum concentrations of
cytokines (TNF-and IL-6) and adhesion
molecules (intracellular adhesion mole-
cule-1 and vascular cell adhesion mole-
cule-1) were at the highest level (6).
In conclusion, high circulating con-
centrations of glucose and FFAs may ex-
plain, at least in part, the oxidative and
inammatory derangements during acute
illness; insulin may exert its anti-inam-
matory action by ameliorating glucose
and lipid parameters. However, any addi-
tional anti-inammatory effect insulin
may have of its own is welcome. The im-
portance is to recognize that treatment of
even trivial hyperglycemia during acute
illness is fundamental to improve sur-
vival. Because insulin is the best choice to
normalize glucose control during stress,
accepting the premise will inevitably
bring increased insulin use in intensive
care units.
KATHERINE ESPOSITO,
MD
RAFFAELE MARFELLA,
MD, PHD
DARIO GIUGLIANO,
MD, PHD
From the Department of Geriatrics and Metabolic
Diseases, Cardiovascular Research Center, Second
University of Naples, Naples, Italy.
Address correspondence to Dario Giugliano, MD,
PhD, Department of Geriatrics and Metabolic Dis-
eases, Policlinico della Seconda Universita`di
Napoli, Piazza L. Miraglia, 80138 Naples, Italy.
Email: dario.giugliano@unina2.it.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Umpierrez GE, Isaacs SD, Bazargan N,
You X, Thaler LM, Kitabchi AE: Hy-
perglycemia: an independent marker of
in-hospital mortality in patients with
undiagnosed diabetes. J Clin Endocrinol
Metab 87:978982, 2002
2. Capes SE, Hunt D, Malmberg K, Pathak P,
Gerstein HC: Stress hyperglycemia and
increased risk of death after myocardial
infarction in patients with and without di-
abetes: a systematic overview. Lancet 355:
773778, 2000
3. Van den berghe G, Wouters P, Weekers F,
Verwaest C, Bruyninckx F, Schetz M,
Vlasselaers D, Ferdinande P, Lauwers P,
Bouillon R: Intensive insulin therapy in
critically ill patients. N Engl J Med 345:
13591367, 2001
4. Dandona P, Aljada A, Bandyopadhyay A:
The potential therapeutic role of insulin in
acute myocardial infarction in patients
admitted to intensive care and in those
with unspecied hyperglycemia. Diabetes
Care 26:516519, 2003
5. Esposito K, Nappo F, Marfella R, Giugli-
ano G, Giugliano F, Ciotola M, Quagliaro
L, Ceriello A, Giugliano D: Inammatory
cytokine concentrations are acutely in-
creased by hyperglycemia in humans: role
of oxidative stress. Circulation 106:2067
2072, 2002
6. Nappo F, Esposito K, CiofM, Giugliano
G, Molinari AM, Paolisso G, Marfella R,
Giugliano D: Postprandial endothelial ac-
tivation in healthy subjects and in type 2
diabetic patients: role of fat and carbohy-
drate meals. J Am Coll Cardiol 39:1145
1150, 2002
Laughter Lowered
the Increase in
Postprandial Blood
Glucose
N
egative emotions such as anxiety,
fear, and sorrow are known to be
factors that elevate the blood glu-
cose level (1). Conversely, positive emo-
tions such as laughter have been reported
to modify the levels of neuroendocrine
factors involved in negative emotions
(2,3) and to modulate immune function
(3,4). However, there have been no stud-
ies on the effects of laughter on blood glu-
cose level. The purpose of this study was
to clarify changes in the blood glucose
level after laughing episodes in patients
with diabetes.
A 2-day experiment was performed in
19 patients with type 2 diabetes not re-
ceiving insulin therapy (16 men and 3
women, age 63.4 1.3 years, BMI
23.5 0.7 kg/m
2
, HbA
1c
, 7.2 0.1%
[means SE]) and 5 healthy subjects
(2 men and 3 women, age 53.6 3.5
years, BMI 24.3 1.6 kg/m
2
, HbA
1c
4.8 0.1% [means SE]). On both ex-
perimental days, they consumed the same
500-kcal meal (79.9 g carbohydrate,
21.0 g protein, 7.8 g fat, and 1.0 g ber).
On the rst day, they attended a monot-
onous lecture (40 min) without humor-
ous content. On the second day, as part
of an audience of 1,000 people attend-
ing MANZAI (a Japanese cross-talk com-
edy) (40 min) in a civic hall, the subjects
laughed. Blood glucose was measured
from the ngertip by enzyme colorimet-
ric assay using a blood glucose self-
measurement apparatus. The subjects
estimated their laughter level on a scale of
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1651
05, and most of them considered that
they laughed well (level 4 or 5). Self-
monitoring of blood glucose was per-
formed before food intake (fasting blood
glucose [FBG]) and 2 h after the meal was
started (2-h postprandial blood glucose
[PPBG]).
The results are presented as means
SE. In the patients, the mean 2-h PPBG
was 6.8 0.7 mmol/l higher than the
FBG after the lecture and 4.3 0.8
mmol/l higher after the comedy show.
The difference in the mean increase be-
tween the lecture and comedy show was
2.5 0.7 mmol/l (P0.005). In the
healthy subjects, the mean increases were
2.0 0.7 and 1.2 0.4 mmol/l after the
lecture and comedy show, respectively,
and the difference was 0.8 0.5 mmol/l
(P0.138).
These results suggest a signicant
suppression of the increase in 2-h PPBG
by comedy show in patients with diabe-
tes, suggesting that laughter ameliorates
the postprandial glucose excursion in the
presence of insufcient insulin action.
This favorable effect of laughter may in-
clude the acceleration of glucose utiliza-
tion by the muscle motion during the
comedy show. However, it is possible that
positive emotions such as laughter acted
on the neuroendocrine system and sup-
pressed the elevation of blood glucose
level.
In conclusion, the present study elu-
cidates the inhibitory effect of laughter on
the increase in PPBG and suggests the im-
portance of daily opportunities for laugh-
ter in patients with diabetes.
KEIKO HAYASHI,
RN, PHD
1
TAKASHI HAYASHI,
BAG
2
SHIZUKO IWANAGA,
RN
3
KOICHI KAWAI,
MD, PHD
3
HITOSHI ISHII,
MD, PHD
4
SHINICHI SHOJI,
MD, PHD
5
KAZUO MURAKAMI,
PHD
2
From the
1
Institute of Community Medicine, Uni-
versity of Tsukuba, Tsukuba, Ibaraki, Japan; the
2
Foundation for Advancement of International Sci-
ence, Bio-Laboratory, Tsukuba, Ibaraki, Japan; the
3
Tsukuba Diabetes Center, Kawai Clinic, Tsukuba,
Ibaraki, Japan; the
4
Department of Endocrinology,
Tenri Hospital, Tenri, Nara, Japan; and the
5
Institute
of Clinical Medicine, University of Tsukuba,
Tsukuba, Ibaraki, Japan.
Address correspondence to Keiko Hayashi, RN,
PhD, 1-1-1, Tennoudai, Tsukuba, Ibaraki, 305-
8577, Japan. E-mail: khys@sakura.cc.tsukuba.ac.
jp.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Surwit RS, Schneider MS: Role of stress in
the etiology and treatment of diabetes
mellitus. Psychosom Med 55:380393,
1993
2. Berk LS, Tan SA, Fry WF, Napier BJ,
Lee JW, Hubbard RW, Lewis JE, Eby
WC:Neuroendocrine and stress hormone
changes during mirthful laughter. Am J
Med Sci 298:390396, 1989
3. Berk LS, Tan SA, Napier BJ, Eby WC: Eu-
stress of mirthful laughter modies natu-
ral killer cell activity (Abstract). Clin Res
37:115, 1989
4. Takahashi K, Iwase M, Yamashita K, Tat-
sumoto Y, Ue H, Kuratsune H, Shimizu A,
Takeda M: The elevation of natural killer
cell activity induced by laughter in a cross-
over designed study. Int J Mol Med 8:645
650, 2001
Differential Levels
of -Glutamyl
Transferase Activity
and Apolipoprotein
CIII in Men on Either
Statin or Fibrate
Therapy
H
igh triglycerides and low HDL cho-
lesterol are frequently associated
with insulin resistance. Statins are
potent hypocholesterolemic drugs, while
brates are better at reducing triglycer-
ides and enhancing HDL. However, little
is known of their effects on other param-
eters potentially associated with insulin
resistance, such as -glutamyl transferase
(GGT) (1) and apolipoprotein CIII (2).
In this cross-sectional study, we have
analyzed the biochemical proles of 500
men from Southwestern France as a func-
tion of their hypolipidemic therapy.
Comparison between untreated (n
361) and brate (n50)- and statin (n
89)-treated subjects revealed no differ-
ence in their levels of total, LDL, and HDL
cholesterol, apo B, apo AI, and apo E, after
adjustment for age, BMI, alcohol intake,
smoking, and physical activity. This sug-
gests that both therapies are equally effec-
tive at normalizing classic lipid variables.
By contrast, apo CIII was higher in the
statin group (34.3 13.9 mg/l) than in
both the brate (26.7 10.3, P0.01)
and nontreated (28.7 10.9 mg/l, P
0.001) groups after adjustment for con-
founders, including triglycerides. Similar
differences were found for Lp B:CIII, a
marker measuring apo CIII associated
with apo B. These observations might in-
dicate an additional benecial effect of -
brates, considering that Lp B:CIII has
been described as an independent predic-
tor of coronary events, better than triglyc-
erides (3).
Most interesting, GGT was two times
lower for brates (36.7 24.4 units/l)
than for the two other groups (67.1
71.3 units/l for statins, P0.01 and
58.1 51.7 units/l in nontreated sub-
jects, P0.01), taking into consideration
potential confounders, including alcohol
intake and triglycerides. GGT is often el-
evated in obese subjects and has been as-
sociated with steatosis, which could be
due to increased effects of insulin in the
liver and could contribute to the develop-
ment of systemic insulin resistance and
hyperinsulinemia in obesity. GGT was re-
ported as one of the independent predic-
tors of the metabolic syndrome and type 2
diabetes (1,4). Moreover, GGT was de-
tected in atheromatous plaques and has
been shown to promote LDL oxidation by
hydrolyzing glutathione into more potent
iron reductants (5). Thus, brates may
prove effective in the treatment of dyslip-
idemic diabetic or obese patients, since
they should improve the hepatic suffering
induced by the triglyceride load and also
limit the potential proatherogenic effects
of GGT.
AcknowledgmentsThis study was sup-
ported by Laboratoires Fournier.
LAURENCE MABILE,
PHD
1,2
JEAN-BERNARD RUIDAVETS,
MD
1
JOSETTE FAUVEL,
PHD
3
BERTRAND PERRET,
MD
2
JEAN FERRIE` RES,
MD, FESC
1
From the
1
De´partement dEpide´miologie, Inserm
U558, Faculte´de Me´decine, Toulouse, France;
2
In-
serm U563, CHU Toulouse, France; and the
3
Labo-
ratoire de Biochimie III, CHU Toulouse, France.
Address correspondence to Prof. Jean Ferrie`res,
Inserm U558, Faculte´de Me´decine, 37 alle´es Jules
Guesde, 31 073, Toulouse Cedex 03, France. E-
mail: ferriere@cict.fr.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Rantala AO, Lilja M, Kauma H, Savolainen
MJ, Reunanen A, Kesaniemi YA: Gamma-
glutamyl transpeptidase and the meta-
Letters
1652 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
bolic syndrome. J Intern Med 248:230
238, 2000
2. Dallongeville J, Meirhaeghe A, Cottel D,
Fruchart JC, Amouyel P, Helbecque N:
Polymorphisms in the insulin response el-
ement of APOC-III gene promoter inu-
ence the correlation between insulin and
triglyceride-rich lipoproteins in humans.
Int J Obes Relat Metab Disord 25:1012
1017, 2001
3. Sacks FM, Alaupovic P, Moye LA, Cole
TG, Sussex B, Stampfer MJ, Pfeffer MA,
Braunwald E: VLDL, apolipoproteins B,
CIII, and E, and risk of recurrent coro-
nary events in the Cholesterol and Recur-
rent Events (CARE) trial. Circulation 102:
18861892, 2000
4. Perry IJ, Wannamethee SG, Shaper AG:
Prospective study of serum gamma-glu-
tamyltransferase and risk of NIDDM. Di-
abetes Care 21:732737, 1998
5. Paolicchi A, Minotti G, Tonarelli P, Tongi-
ani R, De Cesare D, Mezzetti A, Dominici
S, Comporti M, Pompella A: Gamma-glu-
tamyl transpeptidase-dependent iron re-
duction and LDL oxidation: a potential
mechanism in atherosclerosis. J Investig
Med 47:151160, 1999
COMMENTS AND
RESPONSES
IGF-1 and
Macrovascular
Complications of
Diabetes
Alternative interpretations of
recently published data
T
he hypothesis of a detrimental role
for IGF-1 and other growth factors
in the development of vascular dis-
ease (1) has not been consistently sup-
ported by recent studies (2,3). New
evidence suggests that cellular senescence
(4) and impaired vascular endothelial
proliferation, adhesion, and incorpora-
tion play a pivotal role in the development
of macrovascular disease (5). Indeed, re-
cent data on large numbers suggest that
higher IGF-1 bioavailability may protect
against the onset of ischemic heart disease
(6,7) and glucose intolerance (8) and, in
type 2 diabetic patients, may offer im-
proved metabolic control and prevent
vascular complications (9,10). Other po-
tential benecial actions of IGF-1 in car-
diovascular physiology include increased
nitric oxide synthesis and K
channel
opening (11,12), and this may explain the
impaired small-vessel function associated
with low IGF-1 levels in patients with car-
diovascular syndrome X (12). By counter-
acting oxidized LDL-induced cytotoxicity
and vascular smooth muscle cell apopto-
sis, IGF-1 may protect against plaque in-
stability and rupture (2). In patients with
acute myocardial infarction, markedly re-
duced IGF-1 values are associated with a
worse outcome (13,14), and recent data
suggest that intramyocardial (15,16) or
vascular (17) gene delivery of growth fac-
tors can improve symptoms and exercise
capacity in patients with coronary or pe-
ripheral vascular disease.
We were therefore surprised by two
recent studies proposing IGF-1 as a me-
diator of harmful vascular effects (18,19).
The authors found an inverse association
between IGF binding protein (IGFBP)-1
and carotid intimal-medial thickness (r
0.135, P0.041) (18), macrovascular
disease (19), and hypertension (19) in pa-
tients with type 2 diabetes. IGFBP-1 is a
minor component of the system of
IGFBPs 16, rst discovered in the
1990s, and regulated by a complex group
of proteases and phosphatases (20). As
IGFBP-1 is generally assumed to inhibit
IGF-1 bioavailability (21), the authors
conclude that IGF-1 exerted harmful vas-
cular effect (18,19).
We propose two different interpreta-
tions of the above data, not necessarily
exclusive. First, the reduced IGFBP-1
concentration observed in situations of
vascular disease (18,19) could result from
activation of a compensatory mechanism
leading to higher IGF-1 bioavailability.
This interpretation is conrmed by the
nding in hypertensive patients of higher
IGF-1 levels than in control subjects (22),
and of a positive association among
IGF-1, insulin sensitivity, and preserved
vasodilator capacity (23,24). Indeed, in
these patients, IGF-1 was the main inde-
pendent predictor of both coronary re-
serve and insulin sensitivity (23). Second,
alternatively or additionally, one should
consider that the interaction between
IGF-1 and its binding proteins is com-
plex. An adequate serum amount of total
IGFBP-1 is essential for IGF-1s biological
activity (8,25), independently of its phos-
phorilation status. Moreover, overexpres-
sion of IGFBP-1 attenuates IGF-1s
growth-promoting actions in vitro but en-
hances them in vivo (26). Thus, a thresh-
old concentration of IGFBP-1 might
enhance, rather than impair, IGF-1s bio-
activity through a partial agonism-like
action.
Because IGFBPs have been character-
ized only recently and their physiology in
humans is not completely understood, we
think further studies are needed to better
understand the true value of the IGF-1/
IGFBP axis in promoting or protecting
against vascular complications.
ELENA CONTI,
MD
1
DARIO PITOCCO,
MD
2
ETTORE CAPOLUONGO,
BSC, PHD
3
CECILIA ZUPPI,
MD
3
GIOVANNI GHIRLANDA,
MD
2
FILIPPO CREA,
MD, FESC, FACC
1
FELICITA ANDREOTTI,
MD, PHD, FESC
1
From the
1
Department of Cardiology, Catholic Uni-
versity, Rome, Italy; the
2
Department of Diabetolo-
gia, Catholic University, Rome, Italy; and the
3
Department of Hormonal Chemistry, Rome, Italy.
Address correspondence to Elena Conti, MD, In-
stitute of Cardiology, Catholic University, Via Todi
60, 00181 Rome. E-mail: e_conti02@hotmail.com.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Grant MB, Wargovich TJ, Ellis EA, Cabal-
lero S, Mansour M, Pepine CJ: Localiza-
tion of insulin-like growth factor 1 and
inhibition of coronary smooth muscle cell
growth by somatostatin analogues in hu-
man coronary smooth muscle cells: a po-
tential treatment for restenosis? Circulation
89:15111517, 1994
2. Frystyk J, Ledet T, Moller N, Flyvbjerg A,
Orskov H: Cardiovascular disease and in-
sulin-like growth factor 1. Circulation
106:893895, 2002
3. Janssen JA, Lamberts SWJ: The role of
IGF-1 in the development of cardiovascu-
lar disease in type 2 diabetes mellitus: is
prevention possible? Eur J Endocrinol 146:
467477, 2002
4. Erusalimsky JD, Fenton M: Further in
vivo evidence that cellular senescence is
implicated in vascular pathophysiology
(Letters). Circulation 106:e144, 2002
5. Tepper OM, Galiano RD, Capla JM, Kalka
C, Gagne PJ, Jacobowitz GR, Levine JP,
Gurtner GC: Human endothelial progen-
itor cells from type II diabetics exhibit
impaired proliferation, adhesion, and in-
corporation into vascular structures. Cir-
culation 106:27812786, 2002
6. Juul A, Scheike T, Davidsen M, Gyllen-
borg J, Jorgensen T: Low serum insulin-
like growth factor 1 is associated with
increased risk of ischemic heart disease: a
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1653
population-based case control study. Cir-
culation 106:939944, 2002
7. Conti E, Crea F, Andreotti F: IGF-1 and
risk of ischemic heart disease. Circulation.
In press
8. Sandhu MS, Heald AH, Gibson JM,
Cruickshank JK, Dunger DB, Wareham
NJ: Circulating concentrations of insulin-
like growth factor-1 and development of
glucose intolerance: a prospective obser-
vational study. Lancet 359:17401745,
2002
9. Garay-Sevilla ME, Nava LE, Malacara JM,
Wrobel K, Perez U: Advance glycosylation
end products (AGEs), insulin-like growth
factor-1 (IGF-1) and IGF binding pro-
tein-3 (IGFBP-3) in patients with type 2
diabetes mellitus. Diabetes Metab Res Rev
16:106113, 2000
10. Janssen JAMIL, Lamberts SWJ: Circulat-
ing IGF-1 and its protective role in the
pathogenesis of diabetic angiopathy. Clin-
ical Endocrinology 52:19, 2000
11. Muniyappa R, Walsh MF, Rangi JS, Zayas
RM, Standley PR, Ram JL: Insulin-like
growth factor 1 increases vascular smooth
muscle nitric oxide production. Life Sci-
ence 61:925931, 1997
12. Conti E, Andreotti F, Sestito A, Riccardi P,
Menini E, Crea F, Maseri A, Lanza GA:
Markedly reduced insulin-like growth
factor-1 associated with insulin resistance
in syndrome X patients. Am J Cardiol 89:
973975, 2002
13. Conti E, Andreotti F, Sciahbasi A, Ric-
cardi P, Marra G, Menini E, Ghirlanda G,
Maseri A: Markedly reduced insulin-like
growth factor-1 in the acute phase of
myocardial infarction. J Am Coll Cardiol
38:2632, 2001
14. Friberg L, Werner S, Eggertsen G, Ahnve
S: Growth hormone and insulin-like
growth factor-1 in acute myocardial in-
farction. Eur Heart J 21:15471554, 2000
15. Stewart DJ, on behalf of the REVASC
Study Investigators: A phase 2, random-
ized, multicenter, 26-week study to assess
the efcacy and safety of BIOBYPASS
(AdGVVEGF121,10) delivered through
minimally invasive surgery versus maxi-
mum medical treatment in patients with
severe angina, advanced coronary artery
disease, and no options for revasculariza-
tion (Abstract). Circulation 106:2986, 2002
16. Losordo DW, Kawamoto A: Biological re-
vascularization and the interventional
molecular cardiologist: bypass for the
next generation. Circulation 106:3002
3005, 2002
17. Vale PR, Isner JM, Roseneld K: Thera-
peutic angiogenesis in critical limb and
myocardial ischemia. J Interv Cardiol 14:
511528, 2001
18. Leinonen ES, Salonen JT, Salonen RM,
Koistinen RA, Leinonen PJ, Sarna SS,
Taskinen MR: Reduced IGFBP-1 is asso-
ciated with thickening of the carotid wall
in type 2 diabetes. Diabetes Care 25:1807
1812, 2002
19. Heald AH, Siddals KW, Fraser W, Taylor
W, Kaushal K, Morris J, Young RJ, White
A, Gibson JM: Low circulating levels of
insulin-like growth factor binding pro-
tein-1 (IGFBP-1) are closely associated
with the presence of macrovascular dis-
ease and hypertension in type 2 diabetes.
Diabetes 51:26292636, 2002
20. Baxter RC: Insulin-like growth factor
(IGF)-binding proteins: interactions with
IGFs and intrinsic bioactivities. Am J Physiol
Endocrinol Metab 278:E967E976, 2000
21. Schneider MR, Lahm H, Wu M, Hoeich
A, Wolf E: Transgenic mouse models for
studying the functions of insulin-like
growth factors. FASEB J 14:629640, 2000
22. Galderisi M, Vitale G, Lupoli G, Barbieri
M, Varricchio G, Carella C, de Divitiis O,
Paolisso G: Inverse association between
free insulin-like growth factor-1 and iso-
volumic relaxation in arterial systemic hy-
pertension. Hypertension 38:240245, 2001
23. Galderisi M, Caso P, Cicala S, De Simone
L, Barbieri M, Vitale G, de Divitiis O, Pa-
olisso G: Positive association between cir-
culating free insulin-like growth factor-1
levels and coronary ow reserve in arterial
systemic hypertension. Am J Hypertens
15:766772, 2002
24. Laviades C, Gil MJ, Monreal I, Gonzalez
A, Diez J: Tissue availability of insulin-like
growth factor I is inversely related to in-
sulin resistance in essential hypertension:
effects of angiotensin converting enzyme
inhibition. J Hypertens 16:863870, 1998
25. Shinobe M, Sanaka T, Nihei H, Nobuhiro
S: IGF-1/IGFBP-1 as an index for discrim-
ination between responder and nonre-
sponder to recombinant human growth
hormone in malnourished uremic pa-
tients on hemodialysis. Nephron 77:29
36, 1997
26. Modric T, Rajkumar K, Murphy LJ: Thy-
roid gland function and growth in IGF
binding protein-1 transgenic mice. Eur J
Endocrinol 141:149159, 1999
IGF Binding
Protein-1 and
Carotid Intima-
Media Thickness in
Type 2 Diabetes
Response to Conti et al.
W
e thank Conti et al. (1) for their
interest in our recent article (2)
and the editor for the opportu-
nity to further discuss the role of the IGF
system in atherosclerosis.
We found an inverse relation between
IGF binding protein-1 (IGFBP-1) and ca-
rotid intima-media thickness (IMT) in
type 2 diabetic patients. In the linear re-
gression model for diabetic subjects with
cardiovascular disease (CVD), IGFBP-1
was one of the main determinants of IMT
(standardized coefcient ␤⫽⫺0.353,
P0.027) together with age, Apo B, and
pulse pressure; other determinants were
diabetes duration, smoking, BMI, sex,
and lipoprotein [Lp(a)]. This model ex-
plained 57.3% of maximum IMT varia-
tion. IGFBP-1 also remained in the linear
regression model for IMT in subjects
without CVD. Our cross-sectional study
setting does not allow the interpretation
to which extent these associations are
causal. In agreement with previous data
(3), IGFBP-1 concentration was inversely
related to insulin resistance examined by
homeostasis model assessment.
On the contrary, after adjusting for
confounders, we found no association be-
tween total IGF-I and either IMT or pres-
ence of CVD. This indicates the relation
between IGFBP-1 and IMT to be indepen-
dent of IGF-I. Our results underline
IGFBP-1 as a marker of insulin resistance
and the metabolic syndrome, but this
cannot be extrapolated to reect an IGF-I
effect on the vasculature. Similarly, Heald
et al. (4) demonstrated an inverse corre-
lation between IGFBP-1, but not IGF-I,
and clinical cardiovascular disease in type
2 diabetic subjects.
Another nding was the positive cor-
relation of total IGF-I with LDL choles-
terol and Lp(a) and IGFBP-3 with LDL
cholesterol, Lp(a), total cholesterol, and
Apo B after controlling for age, sex, BMI,
and diabetes duration. Thus, in hyper-
cholesterolemic diabetic patients the IGF
axis seems to be activated.
The relation of the IGF system to CVD
is complex and awaits a conclusive an-
swer after over 1,000 published articles
(5). Therefore, we agree with Conti et al.
that further research is needed to under-
stand the entire picture.
EEVA S. LEINONEN,
MD
1
PEKKA J. LEINONEN,
MD, PHD
2
MARJA-RIITTA TASKINEN,
MD, PHD
1
From the
1
Department of Internal Medicine, Hel-
sinki University Central Hospital, Helsinki, Finland;
and the
2
Department of Obstetrics and Gynecology,
Helsinki University Central Hospital, Helsinki, Fin-
land.
Address correspondence to Professor Marja-
Letters
1654 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
Riitta Taskinen, Helsinki University Hospital, Bio-
medicum, Haartmaninkatu 8, P.O. Box 700, 00029
Helsinki, Finland. E-mail: mataskin@helsinki..
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Conti E, Pitocco D, Capoluongo E, Zuppi
C, Ghirlanda G, Crea F, Andreotti F:
IGF-1 and macrovascular complications
of diabetes: alternative interpretations of
recently published data (Letter). Diabetes
Care 26:16531654, 2003
2. Leinonen ES, Salonen JT, Salonen RM,
Koistinen RA, Leinonen PJ, Sarna SS,
Taskinen MR: Reduced IGFBP-1 is asso-
ciated with thickening of the carotid wall
in type 2 diabetes. Diabetes Care 25:1807
1812, 2002
3. Mohamed-Ali V, Pinkney JH, Panahloo A,
Cwyfan-Hughes S, Holly JM, Yudkin JS:
Insulin-like growth factor binding pro-
tein-1 in NIDDM: relationship with the
insulin resistance syndrome. Clin Endocri-
nol (Oxf) 50:221228, 1999
4. Heald AH, Siddals KW, Fraser W, Taylor
W, Kaushal K, Morris J, Young RJ, White
A, Gibson JM: Low circulating levels of
IGF binding protein-1 are closely associ-
ated with the presence of macrovascular
disease and hypertension in type 2 diabe-
tes. Diabetes 51:26292636, 2002
5. Frystyk J, Ledet T, Møller N, Flyvbjerg A,
Ørskov H: Cardiovascular disease and in-
sulin-like growth factor I. Circulation 106:
893895, 2002
Metformin in Type 1
Diabetes
Is this a good or bad idea?
T
he article by Meyer et al. (1) revives
a debate regarding the appropriate-
ness of metformin use for people
with type 1 diabetes. Given the potential
for coexisting lactic acidosis and diabetic
ketoacidosis, how can one justify its use?
Indeed, there was little reason to expect a
benet in patients who were studied:
nonobese type 1 diabetic subjects with
HbA
1c
9.0% who were taking 0.7
units kg insulin
1
day
1
. A modest
average reduction of daily insulin require-
ments, 4.3 units, as compared with an in-
crease of 1.7 units for placebo, does not
seem to be worth the trade-off of in-
creased risk for severe hypoglycemia (19
events in metformin group vs. 8 events in
placebo group). There was no differential
effect in terms of HbA
1c
. Only 7 of 31
patients (23%) treated with metformin re-
sponded in terms of a signicant (20%)
reduction in insulin requirement. Fur-
thermore, it is likely that the incidence of
hypoglycemia would be much greater if
more aggressive metabolic targets of
HbA
1c
had been applied. Despite the fail-
ure to observe diabetic ketoacidosis, the
limited number and short period of ob-
servation does not permit the conclusion
that metformin is safe in ketosis-prone di-
abetic subjects.
We have seen a number of type 1
diabetic patients who have received met-
formin prescriptions by other practitio-
ners. It appears that these prescriptions
were given because of a failure to identify
latent autoimmune diabetes in adults or
because the physician believed that the
potential for insulin dose reduction and
lipid improvement justied a putative
small risk for diabetic ketoacidosis and
lactic acidosis. The temptation to pre-
scribe metformin is increased because of
the high prevalence of metabolic syn-
drome among U.S. adults (2). Indeed, the
diagnosis of metabolic syndrome can fre-
quently be made in the type 1 diabetic
population. For example, using BMI as a
marker for metabolic syndrome, we ob-
served an average BMI of 27 kg/m
2
in 343
consecutive subjects with type 1 diabetes;
this means that our average type 1 dia-
betic patient is overweight. A BMI 30
kg/m
2
, sufcient to diagnosis obesity, was
observed in 89 of 343 subjects (26%).
Seven of our type 1 diabetic patients had a
BMI 41 kg/m
2
. Of these severely obese
subjects, two were receiving metformin
therapy as well as insulin.
The results of the study by Meyer et
al. suggest that a small subset of type 1
diabetic patients benet in terms of insu-
lin dose reduction when metformin is
added to insulin. Questions about long-
term safety and efcacy in this patient
population remain unanswered. There-
fore, when is it reasonable and defensible
to prescribe metformin in type 1 diabetes?
We suggest that metformin should be
avoided unless the following criteria are
met: 1) insulin resistence is clearly inter-
fering with satisfactory glucose control
despite lifestyle interventions; 2) the risk
of diabetic ketoacidosis is minimized by
an intensive program of insulin, self-
monitoring of blood glucose, urine ke-
tone measurement if blood glucose
exceeds 300 mg/dl, and regular medical
supervision; 3) the patient receives coun-
seling so that he or she understands the
potential risk for lactic acidosis; and 4)
efcacy is frequently evaluated to justify
continued use of metformin.
J. DAVID FAICHNEY,
MD, FACP
PHILIP W. TATE,
MD
From the Center for Diabetes and Endocrinology,
Saint Marys Mercy Medical Center, Grand Rapids,
Michigan.
Address correspondence to J. David Faichney,
Center for Diabetes and Endocrinology, Saint Marys
Mercy Medical Center, 300 Lafayette SE, Suite 2045,
Grand Rapids, MI 49503. E-mail: faichned@trinity-
health.org.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Meyer L, Bohme P, Delbachian I, Lehert P,
Cugnardey N, Drouin P, Guerci B: The
benets of metfomin therapy during con-
tinuous subcutaneous insulin infusion
treatment of type 1 diabetic patients. Di-
abetes Care 25:21532158, 2002
2. Ford ES, Giles WH, Dietz WH: Prevalence
of the metabolic syndrome among US
adults: ndings of the third National
Health and Nutrition Examination Sur-
vey. JAMA 287:356359, 2002
Metformin and
Insulin in Type 1
Diabetes
The rst step
T
he insulin-sparing effect of met-
formin and its positive effect on glu-
cose metabolism is now well
documented in insulin-treated type 2 di-
abetic patients, although the mechanisms
of these effects are not overly clear (1). In
insulin-treated type 1 diabetic patients,
the use of metformin has only been as-
sessed in a few studies and, surprisingly,
the same insulin-sparing effect has been
found despite some methodological dif-
culties. However, the clinical interest of
metformin in the treatment of type 1 dia-
betes has remained questionable. For the
rst time, we have shown in a randomized
double-blind trial that metformin has an
insulin-sparing effect in type 1 diabetic
patients (even if the studied population
was not clinically insulin resistant) (2).
The selected patients were C-peptide
negative after intravenous glucagon injec-
tion and were not identied as having
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1655
metabolic syndrome. The patients were
moderately overweight (BMI close to 25
kg/m
2
), and their daily insulin needs were
not greater than that seen for other type 1
diabetic patients. The insulin-sparing ef-
fect of metformin was obtained for basal
insulin needs but not for prandial needs.
Although no denitive explanation could
be given, it is now clear that the use of
metformin may be appropriate for some
type 1 diabetic patients.
Our study was designed to assess the
insulin-sparing effect of metformin in in-
sulin-treated type 1 diabetic patients, and
this end was achieved. However, it was
not designed to give clinical or biological
criteria of indications associated with
metformin use and insulin. We found that
a subset of patients had a reduction of at
least 20% in insulin requirements with
stable and satisfactory glucose control,
but a backward logistic regression did not
allow prediction criteria of a good re-
sponse to be dened. Thus, an expanded
study using selection criteria identied in
our previous study coupled with a larger
range of BMI or bodyweight and a larger
range of daily insulin doses in selected
patients must be performed to determine
therapeutic indications of this association
We believe that metformin use could
be benecial in type 1 diabetic patients
who are overweight or obese, are receiv-
ing large doses of insulin, and have an
HbA
1c
8%. The use of metformin could
be very useful in these patients but must
be carried out under clinical and biologi-
cal supervision to avoid complications
such as lactic acidosis, which remains ex-
ceptional in our experience of type 1 and
in type 2 diabetic patients.
LAURENT MEYER,
MD
BRUNO GUERCI,
MD, PHD
From the Service de Diabe´tologie, Maladies
Me´taboliques & Maladies de la Nutrition, CIC-
INSERM, Hoˆpital Jeanne dArc, Centre Hospitalo-
Universitaire de Nancy, Toul Cedex, France.
Address correspondence to Laurent Meyer, Ser-
vice de Diabe´tologie, Maladies Me´taboliques & Mal-
adies de la Nutrition, CIC-INSERM, Hoˆpital Jeanne
dArc, Centre Hospitalo-Universitaire de Nancy,
B.P. 303, 54201 Toul Cedex, France. E-mail:
laurentmeyer@hotmail.com.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Wulffele´MG, Kooy A, Lehert P, Bets D,
Ogterop JC, Borger van der Burg B,
Donker AJ, Stehouwer CD: Combination
of insulin and metformin in the treatment
of type 2 diabetes. Diabetes Care 25:2133
2140, 2002
2. Meyer L, Bo¨hme P, Delbachian I, Lehert P,
Cugnardey N, Drouin P, Guerci B: The
benets of metformin therapy during
continuous subcutaneous insulin infu-
sion treatment of type 1 diabetic patients.
Diabetes Care 25:21532158, 2002
Prospective Relation
of C-Reactive Protein
With Type 2
Diabetes
Response to Han et al.
W
e read with great interest the ar-
ticle by Han et al., which was re-
cently published in Diabetes Care
(1). The authors studied the association
between baseline levels of C-reactive pro-
tein (CRP) and the 6-year incidence of the
metabolic syndrome and type 2 diabetes
in Mexican subjects. They found an odds
ratio (OR) of 4.1 (95% CI 2.18.0) for
developing the metabolic syndrome and
an OR of 5.4 (2.213.4) for incident type
2 diabetes among women in the highest
compared with the lowest tertile of CRP.
ORs were adjusted for age, smoking,
physical activity, and alcohol intake. In
men, no such association was found. Ad-
ditional adjustment for BMI slightly low-
ered the ORs; adjustment for waist-to-hip
ratio (WHR) did not.
We have studied baseline CRP levels
and their relation to incident type 2 dia-
betes in an age-, sex-, and glucose-
stratied sample of the Hoorn Study, a
population-based cohort study of glucose
tolerance among Caucasian people (2).
The study methods and follow-up dura-
tion are closely similar to the Mexico City
Diabetes Study (MCDS) described by Han
et al., except that our subjects were 15
years older at baseline. Glucose tolerance
status was assessed by a 75-g oral glucose
tolerance test at baseline and after 6.4
years of follow-up. CRP was measured in
plasma by high-sensitive enzyme-linked
immunosorbent assay, while information
was available on BMI, WHR, smoking,
and physical activity. Of the 140 men and
139 women who had follow-up measure-
ments and were free of diabetes at base-
line, 17.8% of the men and 20.9% of the
women developed diabetes. In contrast to
the ndings of Han et al., we did not ob-
serve an association between baseline
CRP levels and incident diabetes in
women, while in men we found an OR of
3.0 (95% CI 1.09.3) in the highest com-
pared with the lowest tertile of CRP, after
adjustment for age (Table 1). Further ad-
justment for BMI, smoking, or physical
activity did not materially change these
results, while adding WHR to the model
substantially lowered the OR in men.
Thus, in the Hoorn Study, CRP is not a
very strong determinant of the develop-
ment of type 2 diabetes, in contrast to
WHR and impaired glucose metabolism
(3). We realize that our study sample of
279 subjects is much smaller than the
study presented by Han et al. (n1,244).
The relationship in men, however, was
strong, whereas in the MCDS it was
strong only in women. In the previous
prospective study by Barzilay et al. (4) an
OR of 2.0 (1.42.9) was found when ex-
treme quartiles of CRP were compared.
They did not analyze men and women or
black and white subjects separately.
Pradhan et al. (5) found an OR of 4.2
(1.512.0) in women. Both studies did
not take WHR into account. Ethnicity
Table 1Relative risk associated with high C-reactive protein (second and third tertile com-
pared with the rst tertile) for developing type 2 diabetes after 6.4 years of follow-up in the
Hoorn Study
Model
Men Women
2nd tertile 3rd tertile 2nd tertile 3rd tertile
1 Age 1.4 (0.44.8) 3.0 (1.09.3) 0.7 (0.32.1) 1.1 (0.43.0)
2 Model 1 BMI 1.4 (0.44.8) 3.0 (1.09.3) 0.6 (0.21.8) 0.9 (0.32.5)
3 Model 1 WHR 1.4 (0.44.8) 1.9 (0.66.4) 0.6 (0.21.7) 0.9 (0.32.5)
4 Model 1 smoking 1.3 (0.44.7) 2.9 (0.99.2) 0.7 (0.32.1) 1.2 (0.43.2)
5 Model 1 physical activity 1.4 (0.44.7) 3.0 (1.09.3) 0.7 (0.32.1) 1.1 (0.43.0)
Data are OR (95% CI).
Letters
1656 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
may also play an important role in ex-
plaining the inconsistent results.
In conclusion, we suggest caution in
the interpretation of the results because of
inconsistent ndings, in particular be-
tween sexes. It is unclear why inamma-
tion would be important in the patho-
genesis of type 2 diabetes only in women
or only in men.
MARIEKE B. SNIJDER,
MSC
1
JACQUELINE M. DEKKER,
PHD
1
MARJOLEIN VISSER,
PHD
1
COEN D.A. STEHOUWER,
MD, PHD
1,2
JOHN S. YUDKIN,
MD
1,3
LEX M. BOUTER,
PHD
1
ROBERT J. HEINE,
MD, PHD
1,4
GIEL NIJPELS,
MD, PHD
1
JACOB C. SEIDELL,
PHD
1,5
From the
1
Institute for Research in Extramural Med-
icine, VU University Medical Center, Amsterdam,
the Netherlands; the
2
Department of Internal Med-
icine, VU University Medical Center, Amsterdam,
the Netherlands; the
3
Department of Medicine, Di-
abetes and Cardiovascular Disease Academic Unit,
University College London Medical School, Lon-
don, U.K.; the
4
Department of Endocrinology, VU
University Medical Center, Amsterdam, the Nether-
lands; and the
5
Department of Nutrition and Health,
Faculty of Earth and Life Sciences, Vrije Universiteit,
Amsterdam, the Netherlands.
Address correspondence to Marieke B. Snijder,
MSC, Institute for Research in Extramural Medicine,
VU University Medical Center, Van der Boechorst-
straat 7, 1081 BT Amsterdam, The Netherlands. E-
mail: mb.snijder.emgo@med.vu.nl.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Han TS, Sattar N, Williams K, Gonzalez-
Villalpando C, Lean ME, Haffner SM: Pro-
spective study of C-reactive protein in
relation to the development of diabetes
and metabolic syndrome in the Mexico
City Diabetes Study. Diabetes Care 25:
20162021, 2002
2. Mooy JM, Grootenhuis PA, de Vries H,
Valkenburg HA, Bouter LM, Kostense PJ,
Heine RJ: Prevalence and determinants of
glucose intolerance in a Dutch Caucasian
population: the Hoorn Study. Diabetes
Care 18:12701273, 1995
3. de Vegt F, Dekker JM, Jager A, Hienkens
E, Kostense PJ, Stehouwer CD, Nijpels G,
Bouter LM, Heine RJ: Relation of impaired
fasting and postload glucose with incident
type 2 diabetes in a Dutch population: the
Hoorn Study. JAMA 285:21092113, 2001
4. Barzilay JI, Abraham L, Heckbert SR,
Cushman M, Kuller LH, Resnick HE,
Tracy RP: The relation of markers of in-
ammation to the development of glucose
disorders in the elderly: the Cardiovas-
cular Health Study. Diabetes 50:2384
2389, 2001
5. Pradhan AD, Manson JE, Rifai N, Buring
JE, Ridker PM: C-reactive protein, inter-
leukin 6, and risk of developing type 2 dia-
betes mellitus. JAMA 286:327334, 2001
Prospective Relation
of C-Reactive Protein
With Type 2 Diabetes
Response to Snijder et al.
W
e thank Snijder et al. (1) for their
interest in our article (2) and for
sharing their data relating C-re-
active protein (CRP) to the risk of diabetes
development in the Hoorn Study. We
readily agree that it is unclear why inam-
mation might be more important to the
pathogenesis of type 2 diabetes in women
compared with men or indeed vice versa.
We have reanalyzed our CRP data using
the same covariates as Snijder et al. and
essentially our results are unchanged
the relation between CRP and diabetes re-
mains strong in women, even after
inclusion of waist-to-hip ratio (WHR),
but is absent in men (Table 1). Interest-
ingly, on reanalysis of similarly relevant
data from the Insulin Resistance Athero-
sclerosis Study (IRAS) (3), a study of sim-
ilar size to the Mexico City Diabetes Study
(MCDS), the relation between baseline
CRP and diabetes development was also
stronger in women compared with men
using the same covariate adjustments
(data not shown). Moreover, in both the
MCDS and IRAS, adjustment for BMI at-
tenuated the link between CRP and dia-
betes more so than WHR.
One possibility of the divergent re-
sults could be the younger baseline age
(46 years) in MCDS in comparison to
that of the Hoorn study (61 years). In
this respect it is important to consider the
additional potential confounding inu-
ence of hormonal replacement therapy
(HRT) in women. HRT use increases CRP
concentrations in women (4), but para-
doxically recent data from the Heart and
Estrogen/Progestin Replacement Study
suggest that HRT reduces risk of diabetes
development by 35% (5). In MCDS, only
4% of women were on HRT at baseline
and hormonal use did not confound the
link between CRP and diabetes. However,
because of higher age, a potentially much
higher proportion of women in the Hoorn
Study may have been on HRT at baseline,
which could thus disguise any potential
association between CRP and diabetes
risk.
We acknowledge that our inability to
nd an association between CRP and di-
abetes risk in men may simply be due to a
lack of power. A recent relevant study (6)
from the West of Scotland Coronary Pre-
vention Study (WOSCOPS) group that
included 127 new cases (predominantly
Caucasian) of diabetes reported a strong
association between baseline CRP and di-
abetes risk independent of BMI, blood
pressure, smoking, and fasting lipids and
glucose concentrations. Alternatively and
as Snijder et al. suggest, ethnicity may also
play a role in explaining inconsistent
results.
Clearly, further studies are needed to
examine the relation between inamma-
Table 1ORs (with 95% CIs) associated with high C-reactive protein (second and third tertile
compared to rst tertile) for developing type 2 diabetes after 6.5 years of follow-up in the
Mexico City Diabetes Study
Model
Men
(35 incident cases/n460)
Women
(54 incident cases/n672)
Second tertile
(0.911.64 ng/ml)
Third tertile
(1.64 ng/ml)
Second tertile
(1.242.16 ng/ml)
Third tertile
(2.16 ng/ml)
1 Age 0.7 (0.31.7) 1.0 (0.52.3) 3.1 (1.27.9)* 5.7 (2.313.9)
2 Model 1 BMI 0.6 (0.21.4) 0.8 (0.41.9) 2.2 (0.95.8)* 3.1 (1.28.1)*
3 Model 1 WHR 0.6 (0.21.5) 0.9 (0.42.1) 3.0 (1.27.7)* 5.4 (2.213.3)
4 Model 1 smoking 0.7 (0.31.7) 1.0 (0.52.3) 3.1 (1.28.0)* 5.7 (2.314.1)
5 Model 1 physical
activity
0.7 (0.31.6) 0.9 (0.42.0) 2.7 (1.07.1)* 5.3 (2.213.2)
The main effect of gender is signicant (P0.05) for each model. Gender-CRP tertile interaction: *P0.05,
P0.01.
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1657
tory markers and diabetes risk. It would
be useful if such studies were to report
associations separately in men and
women in order to enable identication of
any potential gender differences.
NAVEED SATTAR,
MD, PHD
1
KEN WILLIAMS,
MS
2
THANG S. HAN,
MD, PHD
3
CLICERIO GONZALEZ-VILLALPANDO,
MD
4
MICHAEL E.J. LEAN,
MD, FRCP
5
STEVEN M. HAFFNER,
MD, MPH
2
From the
1
University Department of Pathological
Biochemistry, Glasgow Royal Inrmary, Glasgow
U.K.; the
2
Department of Medicine #7873, Univer-
sity of Texas Health Science Center at San Antonio,
San Antonio, Texas;
3
Addenbrookes Hospital,
Cambridge University Medical School, Cambridge,
U.K.; the
4
Center de Estudios in Diabetes, Mexico
City, Mexico; and the
5
University Department of
Human Nutrition, Glasgow Royal Inrmary, Glas-
gow, U.K.
Address correspondence to Steven M. Haffner,
MD, Department of Medicine #7873, University of
Texas Health Science Center at San Antonio, San
Antonio, TX 78229-3900. E-mail: haffner@
uthscsa.edu.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Snijder MB, Dekker JM, Visser JM, Ste-
houwer CDA,Yudkin JS, Bouter LM,
Heine RJ, Nijpels G, Seidell JC: Prospec-
tive relation of C-reactive protein with
type 2 diabetes (Letter). Diabetes Care 26:
16561657, 2003
2. Han TS, Sattar N, Williams K, Gonzalez-
Villalpando C, Lean ME, Haffner SM: Pro-
spective study of C-reactive protein in
relation to the development of diabetes
and metabolic syndrome in the Mexico
City Diabetes Study. Diabetes Care 25:
20162021, 2002
3. Festa A, DAgostino R Jr, Tracy RP,
Haffner SM: Elevated levels of acute-
phase proteins and plasminogen activator
inhibitor-1 predict the development of
type 2 diabetes: the insulin resistance ath-
erosclerosis study. Diabetes 51:1131
1137, 2002
4. Manning PJ, Sutherland WH, Allum AR,
de Jong SA, Jones SD: Effect of hormone
replacement therapy on inammation-
sensitive proteins in post-menopausal
women with type 2 diabetes. Diabet Med
19:847852, 2002
5. Kanaya AM, Herrington D, Vittinghoff E,
Lin F, Grady D, Bittner V, Cauley JA, Bar-
rett-Connor E: Glycemic effects of post-
menopausal hormone therapy: the Heart
and Estrogen/progestin Replacement
Study: a randomized, double-blind, pla-
cebo-controlled trial. Ann Intern Med 138:
19, 2003
6. Freeman DJ, Norrie J, Caslake MJ, Gaw A,
Ford I, Lowe GD, OReilly DS, Packard CJ,
Sattar N: C-reactive protein is an indepen-
dent predictor of risk for the development
of diabetes in the West of Scotland Coro-
nary Prevention Study. Diabetes 51:
1596600, 2002
A Multivariate
Logistic Regression
Equation to Screen
for Diabetes
Response to Tabaei and Herman
W
e read the article by Tabaei and
Herman (1) in the November
2002 issue of Diabetes Care with
great interest and feel that it covers an
exciting and vital area of diabetes re-
search. We applaud the authorsattempts
to improve current standards of diabetes
screening and the journals willingness to
publish important new ndings in the
eld. However, when we reviewed the ar-
ticle, we found the results difcult to re-
produce, especially as there were no
numerical examples provided in the text.
Using the precise formula given in the ar-
ticle, our calculations produced negative
probability values. We would therefore
like to take this opportunity to point out a
source of possible error in the equation
and clarify the logistic regression model.
The logistic regression model and the
logitmodel can be written as:
lnP/1P)] ⫽␤
0⫹␤
iXie (2)
where X ⫽␤
0
⫹␤
i
X
i
are the coefcients
(parameters) for the linear predictor and
X
i
represents the values of the explanatory
variables (we have assumed that these co-
efcients have been correctly estimated in
the article); ln is the natural logarithm,
log
exp
, where exp 2.71828. . . ; Pis the
probability that the event previously un-
diagnosed diabetesoccurs; P/(1 P)is
the odds ratio; and ln[P/(1 P)] is the
log odds ratio, or logit.
In contrast to the simple linear prob-
ability model (Y abX e), the logit
distribution constrains the estimated
probabilities to lie between 0 and 1. For
instance, the estimated probability is:
P[exp(0⫹␤
iXi)]/[1 exp(0⫹␤
iXi)]
or
P1/[1 exp (⫺␤0⫺␤
iXi)]
For a 45-year-old man with a BMI of
29 kg/m
2
, a plasma glucose level of 125
mg/dl, and a postprandial time of 3 h, the
probability calculated with the published
formula would be P⫽⫺0.0555 (i.e. P
0). This result does not match the require-
ment for probabilities to lie between 0 and
1. For the same case, but with the cor-
rected formula, the probability of being
previously undiagnosed for diabetes is
P0.04996.
Details of the calculation with the
formula published in the article:
P1/[1 exp(X)]
X⫽⫺10.0382 0.0331*(age 45
years) 0.0308*(plasma glucose 125
mg/dl) 0.25*(postprandial time 3h)
0.562*(sex 0) 0.0346*(BMI 29
kg/m
2
). Xⴝⴚ2.9453 and P1/[1
exp(X)] 1/(1 exp(2.9453) 1/(1
19.01636644) 1/18.01636644. P
0.0555.
Details of the calculation with the
corrected formula:
P1/[1 exp(X)]
X⫽⫺2.9453; P1/[1 exp(X)]
1/(1 exp[2.9453]) 1/(1
19.01636644) 1/20.01636644; P
0.04996.
Therefore, we would suggest that the
following equations should be used:
X⫽⫺10.0382 0.0331*(age) 0.0308*
(plasma glucose) 0.25* (postprandial
time) 0.562 (if female) 0.0346* (BMI)
and P1/[1 exp(X)]
We hope these comments serve to
clarify this logistic regression equation to
screen for diabetes and, in conjunction
with the valuable work of our colleagues
Drs. Tabaei and Herman, help expand the
knowledge base in this key area of diabe-
tes research.
STE
´PHANE ROZE,
MSC
ANDREW J. PALMER,
BSC, MBBS
WILLIAM J. VALENTINE,
PHD
Letters
1658 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
From the Center for Outcomes Research (CORE),
Basel, Switzerland.
Address correspondence to Ste´phane Roze,
CORE: Center for Outcomes Research, St. Johanns-
Ring 139, 4056 Basel, Switzerland. E-mail: roze@
thecenter.ch.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Tabaei BP, Herman WH: A multivariate
logistic regression equation to screen for
diabetes. Diabetes Care 25:19992003,
2002
2. Hosmer D, Lemeshow S: Applied Logistic
Regression. New York, Wiley, 1989
A Multivariate
Logistic Regression
Equation to Screen
for Diabetes
Response to Roze, Palmer, and
Valentine
W
e would like to thank Roze,
Palmer, and Valentine (1) for
their careful reading of our arti-
cle (2) entitled A multivariate logistic
regression to screen for diabetes: develop-
ment and validation.The estimated
probability formula for the logistic regres-
sion model was indeed misprinted with a
minus sign. The correct estimated proba-
bility formula is:
P1/(1 e
x
), where x
10.0382 [0.0331 (age in years)
0.0308 (random plasma glucose in mg/
dl) 0.2500 (postprandial time assessed
as0to8hours) 0.5620 (if female)
0.0346 (BMI)].
BAHMAN P. TABAEI,
MPH
1
WILLIAM H. HERMAN,
MD, MPH
1,2
From the
1
Department of Internal Medicine, Uni-
versity of Michigan Health System, Ann Arbor,
Michigan; and the
2
Department of Epidemiology,
University of Michigan Health System, Ann Arbor,
Michigan.
Address correspondence to William H. Herman,
MD, MPH, University of Michigan Health System,
Division of Endocrinology and Metabolism, 1500
East Medical Center Dr., 3920 Taubman Center,
Ann Arbor, MI 48109-0354. E-mail: wherman@
umich.edu.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. Roze S, Palmer AJ, Valentine WJ: A mul-
tivariate logistic regression equation to
screen for diabetes (Letter). Diabetes Care
26:16581659, 2003
2. Tabael BP, Herman WH: A multivariate
logistic regression equation to screen for
diabetes. Diabetes Care 25:19992003,
2002
2-Adrenergic
Receptor
Polymorphism and
Obesity in Type 2
Diabetes
Response to van Tilburg et al.
W
e read with interest the observa-
tion letter by van Tilburg et al.
(1) reporting that the Glu27Gln
polymorphism of the 2-adrenergic re-
ceptor (B2ADR) has no important effect
on BMI, metabolic control, or plasma lip-
ids. However, the authors did not men-
tion the physical activity and diet regimen
in their study. B2ADR is a major lipolytic
receptor in human adipocytes, activated
by catecholamines especially during a
weight reduction regimen. Meirhaeghe et
al. (2) reported that B2ADR Glu27Gln
polymorphism is signicantly associated
with obesity in individuals who do not
have regular physical activity, while no
effect of such polymorphism is found in
those who do have physical activity, and
suggested that obese individuals with
B2ADR Glu27Gln genotype may benet
from physical activity to reduce their body
weight. We also demonstrated the same as-
sociation between another B2ADR poly-
morphism (Arg16Gly) and obesity in
individuals treated with a combined low-
energy diet and exercise regimen (3). There-
fore, the authors should have further
divided the subjects according to the degree
of their physical activity and analyzed the
data, because body weight and body fat or
metabolic control are inuenced by physi-
cal activity and dietary changes, especially
in individuals with such polymorphisms.
KEIJI YOSHIOKA,
MD
1
TOSHIHIDE YOSHIDA,
MD
2
TOSHIKAZU YOSHIKAWA,
MD
2
From the
1
Department of Diabetes and Endocrinol-
ogy, Matsushita Memorial Hospital, Moriguchi,
Osaka, Japan; and the
2
Department of Diabetes, En-
docrinology, and Metabolism, Kyoto Prefectural
University of Medicine, Koyto, Japan.
Address correspondence to Keiji Yoshioka. De-
partment of Diabetes and Endocrinology, Matsushi-
ta Memorial Hospital, 5-55, Sotojma-cho, Moriguchi,
Osaka, Japan. E-mail: yoshik@mue.biglobe.ne.jp.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. van Tilburg JH, Wijmenga C, van Bakel H,
Rozeman L, Pearson PL, van Haeften TW:
Relationship of 2-adrenergic receptor
polymorphism with obesity in type 2
diabetes (Letter). Diabetes Care 26:251
252, 2003
2. Meirhaeghe A, Helbecque N, Cottel D,
Amouyel P:
2
-adrenoceptor gene poly-
morphism, body weight, and physical ac-
tivity. Lancet 353:896, 1999
3. Sakane N, Yoshida T, Umekawa T, Ko-
gure A, Kondo M:
2
-adrenoceptor gene
polymorphism and obesity. Lancet 353:
1976, 1999
2-Adrenergic
Receptor
Polymorphism and
Obesity in Type 2
Diabetes
Response to Yoshioka, Yoshida,
and Yoshikawa
W
e recently reported that our
group (n502) of type 2 dia-
betic subjects did not reveal sta-
tistically signicant differences in BMI
with respect to the three groups of carriers
of 2-adrenergic receptor polymor-
phisms (1). Meirhaghe et al. (1) report
that in men not participating in physical
activity, Gln27Gln carriers had a higher
BMI than the Glu27 (combined heterozy-
gous and homozygous) carriers (27.2
0.4 vs. 25.2 0.3 kg/m
2
), while no effect
on BMI was found in the men not partic-
ipating in physical activity.
We have not collected data on the
physical activity of our subjects. How-
ever, taking into account that our subjects
had a mean age of 61 9 years (roughly
10 years older than the subjects reported
by Meirhaghe et al.), we suspect that the
vast majority of them do not participate in
much physical activity. In any case, their
physical activity is presumably less than
that of the subjects studied by Meirhaghe
et al. Much to our surprise, we actually
Letters
DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003 1659
nd the opposite, which is a lower (and
not a higher!) BMI (27.2 3.8) in the
Gln27Gln carriers compared with the
others (27.9 4.4 for Gln27Glu and
28.2 4.0 kg/m
2
for Glu27Glu), al-
though the differences did not attain sta-
tistical signicance. These ndings were
similar for men and women when taken
together or separately.
Obviously, there are two major differ-
ences between these two studies. The sub-
jects in our study (2) had type 2 diabetes
and were considerably older. Whether
these two differences explain the diver-
gent ndings of the study of Meirhaghe et
al. and our study is uncertain. However,
the suggestion by van Tilburg et al. that, if
anything, the BMI was lower (and not
higher) in the Gln27Gln27 group might
point to a chance nding in the studies by
Meirhaghe et al. As for the observations
regarding the Arg16Gly polymorphism,
we have not studied this polymorphisms
in our group of subjects.
It has become more and more clear
that association studies of a particular
polymorphism with a certain physiologi-
cal parameter are often hard to replicate;
this necessitates the need to perform rep-
lication studies before a nding can be
regarded as truly positive.
JONATHAN H. VAN TILBURG,
PHD
1
CISCA WIJMENGA,
PHD
1
TIMON W. VAN HAEFTEN,
MD
2
From the
1
Department of Biomedical Genetics, Uni-
versity Medical Center, Utrecht, the Netherlands;
and the
2
Department of Internal Medicine, Univer-
sity Medical Center, Utrecht, the Netherlands.
Address correspondence to Timon W. van
Haeften, MD, Department of Internal Medicine, G
02.228, University Medical Center, P.O. Box 85500,
3508 GA Utrecht, The Netherlands. E-mail: t.w.
vanhaeften@azu.nl.
© 2003 by the American Diabetes Association.
●●●●●●●●●●●●●●●●●●●●●●●
References
1. van Tilburg JH, Wijmenga C, van Bakel H,
Rozeman L, Pearson PL, van Haeften TW:
Relationship of 2-adrenergic receptor
polymorphism with obesity in type 2
diabetes (Letter). Diabetes Care 26:251
252, 2003
2. Meirhaghe A, Helbecque N, Cottel D,
Amouyel P: B
2
-adrenoceptor gene poly-
morphism, body weight, and physical ac-
tivity. Lancet 353:896, 1999
Letters
1660 DIABETES CARE,VOLUME 26, NUMBER 5, MAY 2003
... The relationship between CRP and T2DM is inde- pendent of insulin resistance and body-mass index (BMI) 11,13 . Many studies have elucidated the role of CRP in the development of T2DM, reporting that even after adjustment for BMI, the relationship between CRP level and incidence of T2DM remained statistically significant 11,[14][15][16][17] . Moreover, observed gender-specific differences have suggested that women have a higher risk of diabetes than men 11,12 . ...
Article
Full-text available
This study was undertaken to assess the associations of C-reactive protein (CRP) with incident type-2 diabetes mellitus (T2DM) and to determine the joint effect of obesity and hypertension on them in the large-scale population-based Korean cohort of the Korean Genome and Epidemiology study (KoGES). We included 22,946 men and women from 11 rural communities at baseline (2005–2011). Epidemiological data and blood samples were collected. Incident physician-diagnosed T2DM cases (130 men and 148 women) were self-reported or based on fasting glucose ≥126 mg/dL or HbA1c level ≥6.5% during a median follow-up of 3.0 years (58,916 person-years) between 2007 and 2014. After multivariate adjustment for T2DM risk factors, the hazard ratios for developing T2DM in the highest CRP tertile (T3), compared with the lowest (T1), was 2.80 (1.73–4.52; p for trend <0.0001) in women and 1.67 (1.00–2.45; p for trend 0.02) in men. The associations between CRP and incident T2DM were more prominent among the older group (≥50 years). And CRP and its combination with obesity and hypertension were associated with increased risk of T2DM. In conclusion, we found positive associations between CRP and incident T2DM in a large population-based Korean cohort.
... Various studies introduced this factor as the connection between H. pylori and Type 2 diabetes; as of 11 prospective studies, seven reported a significant positive association between hsCRP levels and diabetes risk [30,[42][43][44][45][46][47][48] while it found no association in the others. [49][50][51][52] [30,37,53] A study by Aydemir et al. showed higher homeostatic model assessment-estimated IR scores in H. pylori-positive individuals. [54] Furthermore, a Japanese study in 2009 conducted on a large population of 1107 asymptomatic individuals showed that H. pylori was significantly and independently related to IR. [55] However, Gillum supported that there are no consistent associations between H. pylori infection and prevalence of diabetes or syndrome-related variables of the IR in American men of 40-74 years. ...
Article
Full-text available
Helicobacter pylori is one of the most common human pathogens that can cause gastrointestinal (GI) disorders, including simple gastritis, gastric ulcer, and malignant gastritis. In some cases, such as immunodeficiency and underlying diseases, it can be problematic as opportunistic infections. Diabetes mellitus (type 2) (T2DM) is one of the H. pylori underlying diseases. Since GI problems are observed in diabetic patients, it is necessary to treat H. pylori infection. In this review, we aimed to evaluate the possible relationship between H. pylori and T2DM according to epidemiological surveys of 70 studies retrieved from databases, including Scopus, PubMed, and Google Scholar about the relationship between H. pylori and T2DM, and discuss the reported background mechanisms of this correlation. According to the results of our study, the different studies have shown that H. pylori is more prevalent in Type 2 diabetic patients than healthy individuals or nondiabetic patients. The reason is development of H. pylori infection-induced inflammation and production of inflammatory cytokines as well as different hormonal imbalance by this bacterium, which are associated with diabetes mellitus. On the other hand, by tracing anti-H. pylori antibodies in patients with diabetes mellitus and occurrence of symptoms such as digestive problems in >75% of these patients, it can be concluded that there is a relationship between this bacterium and T2DM. Considering the evidence, it is crucially important that the probability of infection with H. pylori is evaluated in patients with T2DM so that medical process of the patient is followed with higher cautious.
... C-reactive protein (CRP) is a nonspecific biomarker of acute inflammation and is produced primarily in the liver. Several prospective studies had shown that serum CRP accelerate or increase the development of diabetes [5,6] particularly in women [7,6] . In acute infections, its serum level would be 50-100 mg/L, but usually not more than 10 mg/L in case of chronic inflammatory conditions like atherosclerosis [8] . ...
Article
Full-text available
The aim of the study is to examine the relationship between Gamma-glutamyltransferase and the marker of inflammation, i.e., high sensitive C reactive protein, in type 2 diabetic patients in Sudan. Materials and Methods: A prospective, analytical, hospital based, case control study was carried out Fedail Hospital and Khartoum Teaching Hospital. 100 patients (50 males and 50 females) presenting with type 2 Diabetes Mellitus were enrolled in this study compared to and 50 healthy control subjects from May 2012 to May 2013 were included in the study. Fasting blood glucose, Serum gamma-glutamyltransferase and High sensitive C-reactive protein were measured. Results: A significant positive correlation between Gamma Glutamyltransferase and high sensitivity-C-reactive protein in patients with type 2 diabetes (r = 0.431, p <0.001). Conclusion: These results suggested that high sensitivity-C-reactive protein levels increase continuously across the fasting blood glucose spectrum starting from the lowest fasting blood glucose in both men and women.
... Snijder et al., in the Hoorn study also stated that CRP level progressively increased as the glucose metabolism deteriorates. 27 Ridker et al., also stated that CRP was an important predictor of T2DM. 28 In the Hong Kong cardiovascular risk factor prevalence study, subjects with IGT with the highest CRP concentration were at three-fold risk of either remaining in IGT or progressing to diabetes compared to subjects with lowest CRP concentration. ...
Article
Full-text available
Objective. The aim of the study was to evaluate the beta cell function, insulin sensitivity and low grade systemic inflammation in different categories of glucose tolerance in Myanmar. Methodology. A cross-sectional study was conducted on 202 Myanmar subjects of both sexes, aged between 45-65 years old. Fasting blood glucose, insulin, C-peptide and hs-CRP levels were measured. A 75g oral glucose tolerance test was performed. Insulin resistance and beta cell function were assessed by homeostasis-model-assessment (HOMA). Results. The subjects were categorized as normal glucose tolerance (NGT), impaired fasting glucose (IFG), impaired glucose tolerance (IGT) and diabetes mellitus (DM) according to WHO-2006 criteria. Fasting serum insulin, C-peptide and hs-CRP levels and insulin resistance index (HOMA-IR) progressively increased from NGT through prediabetes (IFG, IGT) to DM (p<0.01). Beta-cell function did not change significantly in any other group as compared to normal group. Conclusion. After multivariate analysis, increases in fasting C-peptide, hs-CRP and HOMA-IR index were significantly associated with diabetes. It was also found that insulin resistance was a predominant feature in deterioration of the glucose tolerance in Myanmar subjects.
... Inflammation plays an important role in the development of disturbances in both glucose metabolism and CVD [7]. It had been hypothesized that elevated CRP levels contributes to increased cardiovascular risk (8). Festa et al. reported that CRP was positively correlated with considerable components of MetS, such like body mass index (BMI), waist circumference, blood pressure (BP), triglycerides, cholesterol, LDL cholesterol, plasma glucose, and fasting insulin, and then increases in subjects with metabolic syndrome [9]. ...
Article
Full-text available
Abstract Objectives: The objective of this study is to evaluate the levels of gamma-glutamyltransferase and highly sensitive C-reactive protein in patients with metabolic disorders in Sudan. Material and method: A case control hospital based study was carried out at Atbara-River Nile State-Sudan. Total of 294 participants, 194 patients were with at least one of the metabolic disorders (Diabetic, Obese, or Hypertensive) were enrolled in this study together with 100 healthy control subjects during the period from October to November 2013. For all participants, body mass index (BMI), gamma-glutamyltransferase (GGT) and highly sensitive C-reactive protein (hs-CRP) were measured using standard methods. Result: A significant elevation of GGT levels was observed in all patients (diabetic, hypertensive, and obese). Levels of hs-CRP significantly increased in obese and diabetic. While in hypertensive patients insignificant elevation was observed. Conclusion: patients with metabolic disorders, included in this study, have significantly elevated GGT levels and hs-CRP except hypertensive.
... Han et al. reported that CRP was associated with incident T2D independent of BMI in women but not in men (64), and similarly, Thorand et al. reported that CRP was independently associated with incident T2D after adjustment for BMI and insulin resistance in women but not in men (65). Others have reported that adjusting for BMI, waist circumference, fasting insulin, a homeostasis model assessment of insulin resistance (HOMA-IR), or insulin sensitivity, had large attenuating effects on the initial relationship observed between CRP and incident T2D (66)(67)(68)(69)(70). In the Framingham Offspring Study, CRP was not independently associated with incident T2D even prior to adjustment for HOMA-IR or waist circumference (71). ...
... Human CRP is primarily synthesized by hepatocytes and regulated by inflammatory cytokines (mostly TNF-α and IL-6), and levels of high-sensitive CRP (hsCRP) have been the main focus of investigation for diabetes risk. Of 11 prospective studies, seven reported a significant positive association between hsCRP levels and diabetes risk [65,[79][80][81][82][83][84] , and four studies found no association [85][86][87][88] . However, it is not known whether hsCRP itself directly influences IR or diabetes. ...
Article
Full-text available
Helicobacter pylori (H. pylori) is one of the most common human bacterial pathogens, and infection causes a wide array of gastric disorders, including simple gastritis, peptic ulcers and gastric malignancies. Gastrointestinal inflammation caused by H. pylori can influence the absorption of glucose and lipids, which are also abnormal in diabetes mellitus. Type 2 diabetes mellitus (T2DM), formerly known as non-insulin-dependent diabetes mellitus or adult-onset diabetes, is a metabolic disorder that is characterized by high levels of blood glucose resulting from insulin resistance and relative insulin deficiency. It is an emerging pandemic and is rapidly becoming a serious threat to public health. Emerging data now indicate a strong relationship between H. pylori infection and the incidence of T2DM. The mechanisms underlying the pathogenesis of diabetes are complex, involving insulin resistance, chronic inflammation, insulin secretion deficiency as a result of pancreas β-cell dysfunction, glucotoxicity, and lipotoxicity. H. pylori infection is known to be involved in the pathogenesis of insulin resistance, and the growing awareness of its role in diabetes is important for the early detection of glucose dysregulation and prevention of T2DM in high-risk communities. This review probes the possible relationship between H. pylori and diabetes according to epidemiological surveys and discusses putative mechanisms underlying this correlation.
Article
Full-text available
Our aim was to identify whether low-grade inflammation, reflected by C-reactive protein (CRP), explains the higher risk for incident type 2 diabetes (T2D) among ethnic minorities. We included 837 Dutch, 712 South-Asian Surinamese, 797 African Surinamese, 804 Ghanaian, 817 Turkish and 778 Moroccan origin participants of the HELIUS study (Amsterdam, the Netherlands). We used multiple linear regression to assess ethnic differences in CRP levels. We determined the association of CRP with T2D and the modifying effect of ethnicity by cox regression, and compared hazard ratios for the association between ethnicity and T2D before and after adjustment for CRP. CRP levels were higher in ethnic minority groups than in Dutch origin participants. CRP was associated with a higher T2D incidence, similarly across ethnic groups (overall HR per SD 1.38 [95% CI 1.14; 1.68]). However, the association was attenuated and no longer statistically significant after adjustment for adiposity measures (HR 1.11 [95% CI 0.90; 1.37]). CRP accounted for a very small part of the ethnic differences in T2D, but only in models unadjusted for adiposity. Low-grade inflammation does not substantially contribute to the higher risk of T2D among ethnic minority populations compared to the Dutch.
Article
We recently reported that our group ( n = 502) of type 2 diabetic subjects did not reveal statistically significant differences in BMI with respect to the three groups of carriers of β2-adrenergic receptor polymorphisms (1). Meirhaghe et al. (1) report that in men not participating in physical activity, Gln27Gln carriers had a higher BMI than the Glu27 (combined heterozygous and homozygous) carriers (27.2 ± 0.4 vs. 25.2 ± 0.3 kg/m2), while no effect on BMI was found …
Article
Full-text available
To study different nutrients and food additives as risk factors for insulin dependent diabetes mellitus in childhood. Prospective case-control study. Parents of the children being studied were asked to fill in a questionnaire regarding the children's frequency of consumption of various foods. Parents of children with diabetes were asked about the period before onset of the disease. Population based study throughout Sweden. 339 Children aged 0-14 who had recently developed insulin dependent diabetes mellitus and 528 control children matched for age, sex, and county of residence who were traced through the official Swedish population register. Foods were classified according to their content of protein, fat, carbohydrates, monosaccharides or disaccharides, nitrosamines, nitrates or nitrites, vitamin C, and fibres. The frequency of intake was categorised as high, medium, and low and the relative risk for developing insulin dependent diabetes was estimated for the three frequencies of intake and calculated as odds ratios. Significant linear trends for dose response in odds ratios by frequency of intake were shown for solid foods containing high amounts of protein (odds ratio for low frequency of intake 1.0; medium 2.3; and high 5.5), and nitrosamines (1.0; 1.7; 2.6) and significant but non-linear trends were found for carbohydrates (1.0; 1.3; 4.4) and nitrates or nitrites (1.0; 0.8; 2.4). The significant trends were not affected when the results were standardised for possible confounders. No significant increases in odds ratios were found for protein, monosaccharides and disaccharides, vitamin C, and fibres. Nutrients and food additives such as protein, carbohydrate, and nitrosamine compounds may influence the risk of developing insulin dependent diabetes in childhood and significant trends in odds ratios indicate a causal relation.
Article
Full-text available
To study the prevalence and determinants of glucose intolerance in a general Caucasian population. A random sample of 50- to 74-year old Caucasians (n = 2,484) underwent oral glucose tolerance tests. Multiple regression analyses were performed to study the association of 2-h postload plasma glucose values with potential determinants. Prevalence of known and newly detected diabetes and impaired glucose tolerance was 3.6, 4.8, and 10.3%, respectively. In women, but not in men, the association of body mass index with 2-h glucose was fully accounted for by the waist-to-hip ratio. Maternal history of diabetes was twice as prevalent as paternal history, but paternal history only was associated with 2-h glucose. In addition, paternal history was a stronger determinant in men than in women. An independent positive association with 2-h plasma glucose was found for alcohol use of > 30 g/day in women and for intake of total protein, animal protein, and polyunsaturated fatty acids in men. An independent inverse association with 2-h plasma glucose was demonstrated for height (both sexes), alcohol use of < or = 30 g/day (both sexes), energy intake (in men), and, unexpectedly, current smoking (in men). The prevalence of diabetes in elderly Caucasians was 8.3%. In men, dietary habits may unfavorably influence glucose tolerance independent of obesity.
Article
Full-text available
gamma-Glutamyl transpeptidase (gamma-GT) is found in serum and in the plasma membranes of virtually all cell types. Its physiologic role is to initiate the hydrolysis of extracellular glutathione (GSH), a tripeptide in which cysteine lies between alpha-glycine and gamma-glutamate residues. Cysteine and other thiol compounds are known to promote LDL oxidation by reducing Fe(III) to redox active Fe(II); therefore, we sought to determine whether similar reactions can be sustained by GSH and influenced by gamma-GT. Fe(III) reduction and LDL oxidation were studied by monitoring the formation bathophenanthroline-chelatable Fe(II) and the accumulation of thiobarbituric acid-reactive substances, respectively. Human atheromatous tissues were examined by histochemical techniques for the presence of oxidized LDL and their colocalization with cells expressing gamma-GT activity. A series of experiments showed that the gamma-glutamate residue of GSH affected interactions of the juxtaposed cysteine thiol with iron, precluding Fe(III) reduction and hence LDL oxidation. Both processes increased remarkably after addition of purified gamma-GT, which acts by removing the gamma-glutamate residue. GSH-dependent LDL oxidation was similarly promoted by gamma-GT associated with the plasma membrane of human monoblastoid cells, and this process required iron traces that can be found in advanced or late stage atheromas. Collectively, these findings suggested a possible role for gamma-GT in the cellular processes of LDL oxidation and atherogenesis. Histochemical analyses confirmed that this may be the case, showing that gamma-GT activity is expressed by macrophage-derived foam cells within human atheromas, and that these cells colocalize with oxidized LDL. Biochemical and histochemical correlates indicate that gamma-GT can promote LDL oxidation by hydrolyzing GSH into more potent iron reductants. These findings may provide mechanistic clues to the epidemiologic evidence for a possible correlation between persistent elevation of gamma-GT and the risk of fatal reinfarction in patients with ischemic heart disease.
Article
Research in animal models of ischemia has shown that administration of angiogenic growth factors, either as a recombinant protein or by gene transfer, can augment nutrient perfusion through neovascularization to promote the development of supplemental collateral blood vessels that will constitute endogenous bypass conduits around occluded native arteries; a strategy termed “therapeutic angiogenesis.” In animal models and clinicul trials, the best studied cytokines with angiogenic activity are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). Clinical trials of therapeutic angiogenesis in patients with critical limb ischemia demonstrated resolution of rest pain and/or improved limb integrity, increased pain-free walking time and ankle-brachial index, newly visible collateral vessels by digital subtraction angiography, and qualitative evidence of improved distal flow by magnetic resonance imaging. Initial clinical trials in patients with end-stage coronary artery disease using direct myocardial injection via thoracotomy resulted in large increases in exercise time and marked reductions in anginal symptoms, as well as objective evidence of improved perfusion and leji ventricular function. Larger scale placebo-controlled trials have been limited to intracoronary and intravenous administration of recombinant protein, and have not shown significant improvement in exercise time or angina compared to placebo. Larger scale placebocontrolled studies of gene transfer using catheter-based endocardial delivery are in progress. Future clinical studies are required to determine the optimal dose, formulation, route of administration, and combinations of growth factors, as well as the requirement for endothelial progenitor cell or stem cell supplementation, to provide effective and safe therapeutic angiogenesis for patients with critical limb ischemia and chronic myocardial ischemia who are not candidates for conventional revascularization procedures.
Article
Humoral immune factors related to type 1 diabetes have been investigated in children with coeliac disease. Anti-insulin (IAAb), immunoglobulin (alpha IgAb), islet cell (ICA) and glucagon autoantibodies were examined in 15 children with coeliac disease at diagnosis (group 1), in 15 children with coeliac disease following a gluten-free diet (group 2) and in 30 control patients (groups 3 and 4). IAAb were present in 27% of group 1 and in 20% of group 2 patients and alpha IgAb were significantly increased in group 1 and 2 patients; two patients in group 2 were positive for ICA; none of the coeliac disease patients were positive for anti-glucagon antibodies. The levels of anti-gliadin antibodies in group 1 were positively correlated with those of alpha IgAb. Coeliac disease-related HLA antigens were not correlated with antibody presence. The presence of diabetes-related humoral immune factors in coeliac disease raises the question as to whether or not they are predictive of subclinical pancreatic damage or whether they are simply indicators of a more general autoimmune diathesis.
Article
Insulin like growth factor 1 (IGF-1) vasodilates, increases blood flow and lowers blood pressure; nitric oxide (NO) has been suggested to mediate some of these effects. We studied the role of IGF-1 in the regulation of NO production in vascular smooth muscle cells (VSMC). IGF-1 induced a concentration and time-dependent increase in NO release from endothelium-denuded aortic rings. Pre-incubation with cycloheximide or aminoguanidine blocked IGF-1-stimulated NO release. In addition, a six-fold increase in NO production by VSMC was seen upon incubation with IGF-1. These results suggest that IGF-1 induces NO release in VSMC through a process that involves new protein synthesis and the inducible isoform of nitric oxide synthase.
Article
In order to examine the role of insulin-like growth factors in the pathogenesis of accelerated macrovascular disease in noninsulin-dependent diabetes mellitus (NIDDM), we investigated the relationship between the insulin resistance syndrome and the IGF axis. Cross-sectional analysis of the relationship between insulin resistance syndrome variables and concentrations of IGF-1, IGF-2, IGFBP-1 and IGFBP-3 in 80 subjects with NIDDM. After correcting for age, sex and body mass index, concentrations of IGFBP-1, correlated with those of HDL-cholesterol (r = 0.40; P < 0.001), triglycerides (r = -0.24; P = 0.04), insulin (r = -0.39; P < 0.001), intact proinsulin (r = -0.32; P = 0.006), des 31,32 proinsulin (r = -0.40; P = 0.001), and with insulin sensitivity (r = 0.38; P = 0.001) and PAI-1 activity (r = -0.24; P = 0.05); IGF-1 levels only correlated with those of HDL-cholesterol (r = -0.33; P = 0.005), and this was not explained by IGFBP-1 or insulin sensitivity. With additional correction for insulin, concentrations of IGFBP-1 still correlated with HDL-cholesterol (r = 0.40; P < 0.001), but not those of triglycerides or PAI-1 activity. There were no significant relationships between levels of IGF-2 and any of the variables investigated, and IGFBP-3 levels only correlated with those of total cholesterol (r = 0.24, P = 0.04). In NIDDM, concentrations of IGFBP-1 are related to those of insulin, insulin sensitivity, serum lipoproteins and PAI-1 activity. The relationship between concentrations of IGFBP-1 and HDL-cholesterol is not explained by insulin. Concentrations of IGF-1 are linked to HDL-cholesterol, and this is not explained by levels of IGFBP-1. IGFBP-1 concentrations were related to PAI-1 activity, and this may be explained by insulin, which regulates the production of IGFBP-1 and PAI-1.
Article
Poor glycaemic control in type 1 diabetes is associated with elevated serum IGFBP-1 levels and reduced rather than elevated serum IGF-I levels. Increasing age is accompanied by a further decrease in serum IGF-I levels as well as an increase in IGFBP-l levels in adult diabetic type 1 and type 2 subjects. This is especially observed in diabetic type 1 subjects with manifest microvascular complications. IGFBP-I has been proposed as one of the IGF-I inhibitors in the serum of diabetics. Lowered IGF-I and increased IGFBP-1 levels in the blood may thus result in decreased IGF-I bioavailability at the tissue level. We hypothesize that the premature and progressive decline in serum IGF-I bioactivity during ageing in diabetics ultimately results in insufficient protective effects by IGF-I in the kidneys, eyes and neurones, and thus the progression of diabetic microvascular complications. If this hypothesis is proven to be right, treatment of diabetic patients with IGF-I (eventually complexed to IGFBPs) as an adjunct to insulin might prevent and not worsen the development of diabetic microvascular complications.