ArticlePDF Available

Early Diabetic Nephropathy A complication of reduced insulin sensitivity in type 1 diabetes

Authors:

Abstract

OBJECTIVE Diabetic nephropathy (DN) is a major cause of mortality in type 1 diabetes. Reduced insulin sensitivity is a well-documented component of type 1 diabetes. We hypothesized that baseline insulin sensitivity would predict development of DN over 6 years.RESEARCH DESIGN AND METHODS We assessed the relationship between insulin sensitivity at baseline and development of early phenotypes of DN-microalbuminuria (albumin-creatinine ratio [ACR] ≥30 mg/g) and rapid renal function decline (glomerular filtration rate [GFR] loss >3 mL/min/1.73 m(2) per year)-with three Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equations over 6 years. Subjects with diabetes (n = 449) and without diabetes (n = 565) in the Coronary Artery Calcification in Type 1 Diabetes study had an estimated insulin sensitivity index (ISI) at baseline and 6-year follow-up.RESULTSThe ISI was lower in subjects with diabetes than in those without diabetes (P < 0.0001). A higher ISI at baseline predicted a lower odds of developing an ACR ≥30 mg/g (odds ratio 0.65 [95% CI 0.49-0.85], P = 0.003) univariately and after adjusting for HbA1c (0.69 [0.51-0.93], P = 0.01). A higher ISI at baseline conferred protection from a rapid decline of GFR as assessed by CKD-EPI cystatin C (0.77 [0.64-0.92], P = 0.004) and remained significant after adjusting for HbA1c and age (0.80 [0.67-0.97], P = 0.02). We found no relation between ISI and rapid GFR decline estimated by CKD-EPI creatinine (P = 0.38) or CKD-EPI combined cystatin C and creatinine (P = 0.50).CONCLUSIONS Over 6 years, a higher ISI independently predicts a lower odds of developing microalbuminuria and rapid GFR decline as estimated with cystatin C, suggesting a relationship between insulin sensitivity and early phenotypes of DN.
Early Diabetic Nephropathy
A complication of reduced insulin sensitivity in type 1 diabetes
PETTER BJORNSTAD, MD
1
JANET K. SNELL-BERGEON, PHD, MPH
1,2
MARIAN REWERS, MD, PHD
1,2
DIANA JALAL, MD
3
MICHEL B. CHONCHOL, MD
3
RICHARD J. JOHNSON, MD
3
DAVID M. MAAHS, MD, PHD
1,2,3
OBJECTIVE dDiabetic nephropathy (DN) is a major cause of morta lity in type 1 diabetes.
Reduced insulin sensitivity is a well-documented component of type 1 diabete s. We hypothe-
sized that baseline insulin sensitivity would predict development of DN over 6 years.
RESEARCH DESIGN AND METHODSdWe as sessed the relationship between in-
sulin sensitiv ity at baseline and development of early phenotype s of DNdmicroalbuminuria
(albumin-creatinine ratio [ACR] $30 mg/g) and rapid renal fu nction decline (glomerular ltra-
tion rate [GFR] loss .3 mL/mi n/1.73 m
2
per year)dwith three Chronic Kidney Disease Epide-
miology Collaboration (CKD-EP I) equations over 6 years. Subjects with diabetes (n =449)and
without diabetes (n = 565) in the Coronary Artery Calcication in Type 1 Diabetes study had an
estimated insulin sensitivity index (ISI ) at baseline and 6-year follow-up.
RESULTSdThe ISI was lower in subjects with diabetes than in those withou t diabete s (P ,
0.0001). A higher ISI at baseline predicted a lower odds of developing an ACR $30 mg/g (odds
ratio 0.65 [95% CI 0.490. 85], P = 0.00 3) univariately and after adjus ting for HbA
1c
(0.69 [0.51
0.93], P = 0.01). A higher ISI at baseline conferred prote ction from a rapid decline of GFR as
assessed by CKD-EPI cystatin C (0.77 [0.64 0.92], P = 0.004) and remained signicant after
adjusting for HbA
1c
and age (0.80 [0.670.97], P = 0.02). We found no relation between ISI and
rapid GFR decline estimated by CKD-EPI creatinine (P = 0.38) or CKD-EPI combined cystatin
C and creatinine (P = 0.50).
CONCLUSIONSdOver 6 years, a higher I SI independently predicts a lower odds of devel-
oping microalbuminuria and rapid GFR de cline as estimated with cystatin C, suggesting a re-
lationship between insulin sensi tivity and early phenotypes of DN.
Diabetes Care 36:36783683, 2013
D
iabetic nephr opathy (DN) is a com-
mon and serious complication of
diabetes. Its incidence is rising rap-
idly (1), and it is the most common cause
of end-stage renal di sease in the U.S. and
Europe (2). The 2011 U.S. R enal Data
System showed that DN accounted for
44.5% of all cases of end-stage renal dis-
ease in 2009 (3). Despite improvements
in the outlook of this complication in past
decades, it continues to be one of the ma-
jor causes of morbidity and mortality in
type 1 diabetes (4,5). DN is an important
risk factor for coronary artery disease (68)
and overall mortality (6,9). These nd-
ings highlight the need for improved
methods of identifying persons at high
risk for DN (10).
The role of insulin sensitivity in the
development and progression of macro-
(7,11,12) and microvascular complications
(12,13) in type 1 diabetes is increasingly
recognized. Reduced insulin sensitivity
also is a plausible mechanism linking renal
disease with excess mortality in type 1 di-
abetes. Historically, when glyc emic con-
trol is poor, reduced insulin sensitivity
was believed to be directly related to
body weight and HbA
1c
(14,15), but
more recent data suggest that reduced in-
sulin sensitivity cannot simply be explained
by weight or poor glycemic control. In fact,
reduced insulin sensitivity has been docu-
mented in type 1 diabetic subjects with
normal BMI and HbA
1c
compared with
nondiabetic individuals (16). The Coro-
nary Artery Calcication in Type 1 Diabe-
tes (CACTI) longitudinal cohort study of
adults with type 1 diabetes investigated
the determinants of early and accelerated
atherosclerosis and found that insulin sen-
sitivity independently predicted coronary
artery calcication (17,18). Reduced insu-
lin sensitivity has also been shown to pre-
dict diabetic retinopathy, neuropathy,
and nephropathy in subjects with type 1
diabetes (13).
Despite advances in the estimation of
insulin sensitivity (insulin sensitivity in-
dex[ISI])(19)andglomerularltration
rate (GFR) (20), research in the associa-
tion of insulin sensitivity with DN has
been limited since the Pittsburgh Epide-
miology of Diabetes Complications
(EDC) cohort showed more than a decade
ago that the estimated glucose disposal
rat e (eGDR) predicts ov ert nephropathy
(13). To readdress this relationship with
contemporary data and estimat ing equa-
tions, we hy pothesized that higher insuli n
sensitivity measured by ISI at baseline
wouldbeassociatedwithdecreased
odds of developing two early phenotypes
of DNdmicroalbuminuria (albumin-
creatinine ratio [ACR] $30 mg/g) and
rapid renal function decline (GFR l oss
.3mL/min/1.73m
2
per year) (2123)d
calculated by the three Chronic Kidney Dis-
ease Epidemiology Collaboration (CKD-
EPI) equations (20) over 6 years in the
CACTI study.
RESEARCH DESIGN AND
METHOD S dThe CACTI study en-
rolled subjects 1956 years of age with
and without type 1 diabetes who were
asympto matic for cardiovascular disease
at the baseline visit in 20002002 and
then wer e reexamined 3 and 6 years later
as previously described (24). Subjects
with type 1 diabetes had to have a disease
duration of at least 10 years at enro llment,
with the exce ptio n of 109 su bjects w ho
had taken part in a pilot study in 1997
1998 and had 248 years of diabetes du-
ration. Subjects with serum creatinine
levels .2 mg/dL were excluded at baseline
cccccccccccccccccccccccccccccccccccccccccccccccc c
From the
1
Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; the
2
Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado; and the
3
Department
of Nephrology, University of Colorado Denver, Aurora, Colorado.
Corresponding author: Petter Bjornstad, petter.bjornstad@childrenscolorado.org.
Received 14 March 2013 and accepted 7 June 2013.
DOI: 10.2337/dc13-0631
© 2013 by the American Diabetes Association. Readers may use this article as long as the work is properly
cited, the use is educational and not for prot, and the work is not altered. See http://creativecommons.org/
licenses/by-nc-nd/3.0/ for details.
3678 DIABETES CARE, VOLUME 36, NOVEMBER 201 3 care.diabetesjournals.org
Pathophysiology/Complications
ORIGINAL ARTICLE
unless they were participants in the pilot
study. All subjects with the data needed to
calculate ISI and GFR at the baseline and
6-year visits were included in this analysis
(n = 449 with type 1 diabetes, n =565
wit hout diabetes). Among subjects with
type 1 diabetes, those with missing data
had slightly worse baseline renal function
(e.g., CKD-EPI cystatin C eGFR 108 6 18
vs. 100 6 30 mL/min/1.73 m
2
) and fasting
lipid prole than those with complete
data. In contrast, nondiabetic control sub-
jects with missing data were younger
(35 6 9 vs. 40 6 9 years) and had better
renal function (CKD-EPI cystatin C eGFR
111 6 13 vs. 108 6 12 mL/min/1.73 m
2
).
The study was approved by the Colorado
Multiple Institutional Review Board, and
all subjects provided informed consent.
We measured heig ht and we ight and
calculated B MI as kilograms pe r meter
squared. Resting systolic blood pressure
(SBP) and fth phase diastolic blood
pressure (DBP) were measured three
times while the subj ect was seate d, and
the second and third measurements were
averaged. Hypertension was dened as
current antihypertensive therapy or un-
treated hypertension (blood pressure
$140/90 mmHg) at the time of the study
visit. A ntihypertension medicati on use
was determined by a medication inven-
tory as previously described (24), and
use of an angio tensin-convert ing enzyme
inhi bitor (ACEi) or an angiotensin recep-
tor blocker (ARB) was combined for these
anal yses.
After an overnight fast, blood was
collected, centrifuged, a nd separated.
Plasma was stored at 48C until assayed.
Total plasma cholesterol and triglyceride
levels were measured by standard enzy-
matic meth ods, HDL cholesterol was sep-
arated with the use of dextran sulfate, and
LDL cholest erol was calculated by the
Friedewald equation. High-performance
liquid chromatography (Bio-Rad variant)
was used to measure HbA
1c
.
Ove rnight urine samples were col-
lected in duplicate, and urine creatinine
and albumin levels were measured by
radioimmunoassay (Dia gnostic Pro ducts)
and averaged. In sub jects (n = 35) who did
not have two timed overnight urine sam-
ples, only one timed overnight urine sam-
ple was collected. Two subjects did not
provide a timed overnight urine sample,
and instead, spot urine was collected.
Early DN was dened as microalbu-
minuria (ACR $30 mg/g) and rapid GFR
decline (.3 mL/min/1.73 m
2
per year)
(23,25) by all three CKD-EPI equations.
Twenty-ve type 1 diabetic subjects had
an ACR ,30 mg/g at the rst visit (V1) ,
and ACR $30 mg/g developed in them by
the third visit (V3). In contrast, 284 sub-
jects had ACR ,30 mg/g at both V1 and
V3. The analysis compa red t hese two
groups of subjects. Microalbuminuria de-
veloped in only ve control subjects, so
we did not have sufcient power to run
anal yses on this outcome.
GFR was determined wi th the use of
the CKD-EPI serum creatinine, serum
cystatin C, and combined equations re-
cently published by Inker et al. (2 0) for
the CKD-EPI Investigators Group. Cystatin
C was measured in the University of Col-
orado Hospital clinical laboratory accord-
ing to the package insert instructions
of a commercially available assay (Dade-
Behring) on a BN II or Prospec nephelom-
eter. The coefcient of variation was 3.3%.
Intraassay precision is 2.34.1%, and in-
terassay precision is 2.63.3% according
to the package insert. Results are reported
in milligrams per liter, with a sensitivity
cutoff of 0.23 mg/L. Because of a system-
atic shift in the Dade-Behring cystatin C
assay over the period of the study, cystatin
C levels were standardized to V3 levels by
Deming regression equations, as previ-
ously described (26).
Serum cr eati nine level was measured
according to package insert instructions
on a Roche Mira Plus II an alyzer until
2006 and then an Olympus AU400e (r =
0.9999 between methodologies) trace-
able to the National Institute of Standards
and Technol ogy standard reference mate-
rial at the U niversity of Colorado Clinical
Translational Res earch L aboratory. Intra-
and interassay precision are 1.6 and 3.3%,
respect ively. Because of the longitudinal
design of CACTI and to assess for any po-
tential c hang e over time in the serum cre-
atinine assay, 100 samples each from all
three visits w ere remeasured as a single
batch. Deming regression equations
were u sed to standardize serum creatinine
level by the same methodology as previ-
ously described for cystatin C (26).
With the use of the CKD-EPI creati-
nine equation to estimate GFR, 122 sub-
jects with type 1 diabetes experienced a
GFR loss .3 mL/min/1.73 m
2
per year
compared with 327 who had a stable re -
nal function (GFR loss ,3 mL/min/1.73
m
2
per year). In contrast, by CKD-EPI
cystatin C equa tion, rapid GFR decline
developed in only 58 subjects with type
1 diabetes compared with 391 with stable
renal function. By combined CKD-EPI
creatin ine and cyst atin C equation, rapid
GFR decline developed in 64 subjects
with type 1 diabetes compared with 385
with stabl e renal function on foll ow-up.
We compared subjects with rapid GFR
decl ine with those with stable renal func-
tion over the 6 years of the study. Rapi d
GFR decline developed in only 29 control
subjects per CKD-EPI cystatin C equation
compared with 135 and 4 7 control sub-
jects per CKD-EPI creatinine and combined
creatinine and cystatin C equations,
respective ly.
CACTI clamp cohort: ISI
The ISI is derived from previous euglycemic-
hyperinsulinemic clamp studies in a sub-
set of 87 subjects (40 with and 47 without
diabetes frequency matched for age, sex,
and weight) of the CACTI cohort, as
previously described i n detail (18). The
best-t ISI model for subjects with type
1 diabetes comprises waist circumfer-
ence, daily insulin dose per kilogram
body weight, triglyceride levels, and
DBP and explained 63% of the variance
in GDR. The equation to calculate the ISI
is as follows: exp(4.1075 0.01299 3
waist [cm] 1.05819 3 insuli n dos e
[daily units/kg] 0.00354 3 triglycer-
ide level [mg/dL] 0.00802 3 DBP
[mmHg]) (19).
Statistical analysis
Anal yses were perfor med with SAS ver-
sion 9.2 for Windows (SAS Institute,
Cary, NC) software. Differences between
type 1 diabetic subjects and control sub-
jects at baseline and follow-up were as-
sessed by x
2
test for categorical variables
and t test for continuous v ariables. Non -
parametric variables (e.g., ACR and tri-
glyceride leve l) w ere log transforme d to
compute geometric means. U nivariate lo-
gistic regres sion was perfo rmed to evalu-
ate the associations between variables at
baseline and the development of micro-
albuminuria and rapid renal function de-
cline c alculated by the CKD-EPI
crea tinine, cystatin C, and combined cre-
atinine and cystatin C equ ations. The fol-
lowing variables wer e tested for univariate
associations with the dependent out-
comes: LDL cholesterol, HDL cholesterol,
total cholester ol, SBP, HbA
1c
, type 1 di-
abetes duration, and age. DBP and triglyc-
eride level were not included because t hey
are part of the ISI. Only variables s igni-
cantly associated with the dependent var-
iables in univariate analyses were eligible
for selection in the multivariable models,
with a signicance level of P , 0.05 for
inclusion. Stepwise logistic regression
care. diabetesjournals.org DIABETES CARE, VOLUME 36, NOVEMBER 2013 3679
Bjornstad and Associates
was used to determine which variables re-
mained in multivariable models predicting
microalbuminuria and rapid GFR de-
cline. Only variables with P , 0.1 in step-
wiseselectionwereincludedinthe
models. We further evaluated the inde-
pendence of the association of ISI to renal
outcomes by adjusting the multivariable
models for the dichotomous variabl es of
sex, smoking status, and ACEi/ARB use.
All analyses were stratied by type 1 dia-
betes status, and signicance was based on
an a of 0.05.
RESULTSdData, including ISI, were
available for 449 subjects with type 1
diabetes and 565 control subjects at base-
line (V1) and follow-up (V3). Among type
1 diabetic subjects, 76 had an ACR $30
mg/g, and 1521 (depending on which
eGFR calculation was used) had a GFR
,60 mL/min/1.73 m
2
at V1; all were ex-
cluded from the analyses. Twenty-ve
subjects progressed from normoalbumi-
nuria at V1 to microalbuminuria at V3,
and there were 1214 new cases of GFR
,60 mL/min/1.73 m
2
.Conversely,
among control subjects, ACR $30 mg/g
developed in only six, and GFR ,60 mL/
min/1.73 m
2
developed in only two t o
four at V3.
The mean ISI at V1 among type 1 di-
abetic subjects was 4.4 6 1.6 mg z kg
21
z
min
21
vs. 15.4 6 6.7 mg z kg
21
z min
21
in control subjects (P , 0.0001). Simi-
larly at V3, the mean was 4.7 6 1.8 and
15.6 6 6.6 mg z kg
21
z min
21
among type
1 diabetic and cont rol subjects, respec-
tively (P , 0.0001). In addition, among
both subject groups, the mean differences
in HbA
1c
; age; total cholesterol, HDL cho-
lesterol, LDL cholesterol, and triglyceride
levels; SBP and DBP; and the proportions
of hypertension, ACEi/ARB use, and non-
Hispanic whites were all statistically sig-
nicantly different (Table 1). There was a
signicant overall decline in eGFR over 6
years i n the enti re cohort as calculated by
CKD-EPI creatinine (P , 0.000 1), cysta-
tin C (P , 0.0001), and combined creat-
inine a nd cystatin C (P , 0.0001).
In univariate analysis, only ISI at
baseline predicted a lower odds of de-
veloping ACR $30 mg/g (od ds ratio 0.65
[95% CI 0.490.85], P = 0.003), and
HbA
1c
predicted a higher od ds of devel-
oping ACR $30 mg/g (1.45 [1.101.92],
P = 0.009). ISI remained in the model af-
ter adjusting for HbA
1c
in a multivar iable
logistic regression (0.69 [0.510.93], P =
0.013) (Table 2). Moreover, the associa-
tion rem ained signicant with additional
adjustments for sex, smok ing status, and
ACEi /ARB use (P =0.018).
In univariate analysis, ISI was associ-
ated with a d ecrease d odds (0.77 [0.64
0.92], P = 0.004) and HbA
1c
(P ,
0.0001), age (P = 0.04), SBP (P = 0.02),
and BMI (P = 0.01 ) w ere associated with
an increased odds of developing r apid
GFR decline as calculated by CKD-EPI
cystatin C. In the ste pwise model, ISI re-
mained signicant after adjusting for
HbA
1c
and age (0.80 [0.670.97], P =
0.02) (Table 3). Moreover, the association
rema ine d unchanged with additional ad-
justments for both sex and smoking sta-
tus, but a djusting for ACEi/ARB use at
baseline a ttenuated the s ignicance of
the association (0.831 [0.6861.006],
P = 0.058). We found no associations
Table 1dSubject characteristics
V1 V3
Variable
Type 1 diabetes
(n = 449)
No diabetes
(n =565) P value
Type 1 diabetes
(n =449)
No diabetes
(n = 565) P value
Age (years) 36.7 6 8.7 40.2 6 8.8 ,0.0001 43.3 6 8.7 46.8 6 8.8 ,0.0 001
Sex 0.31 0.12
Male 47 51 46 50
Female 53 49 54 50
Non-Hispa nic whites 95 85 ,0.0001 95 85 ,0.0 001
Type 1 diabetes duration (years) 22.9 6 8.9 NA NA 29.2 6 8.9 NA NA
HbA
1c
(%) 7.9 6 1.2 5.5 6 0.4 ,0.0001 7.9 6 1.2 5.5 6 0.5 ,0.0001
HbA
1c
(mmol/mol) 63 6 10.8 37 6 2 ,0.0001 63 6 10.8 37 6 3.1 ,0.0001
BMI (kg/m
2
)26.16 4 .3 26.0 6 4.8 0.75 26.7 6 4.6 26.5 6 4.9 0.47
ISI (mg z kg
21
z min
21
)4.46 1 .6 15.4 6 6.7 ,0.0001 4.7 6 1.8 15.6 6 6.6 ,0.0001
Cystatin C (mg/L) 0.81 6 0.21 0.78 6 0.10 0.03 0.85 6 0.34 0.78 6 0.12 ,0.0 001
Serum creatinine (mg/dL) 0.83 6 0.34 0.83 6 0.24 0.89 0.86 6 0 .38 0.83 6 0.18 0.13
CKD-EPI equation
Creatinine (mL/m in/1.73 m
2
) 105 6 24 102 6 20 0.08 97.9 6 21.4 96.5 6 14.5 0.22
Cystatin C (mL/min/1.73 m
2
) 108 6 18 108 6 12 0.84 102 6 22 106 6 14 0.006
Combined (mL/min/1.73 m
2
) 103 6 23 101 6 18 0.14 101 6 22 102 6 13 0.39
MA or greater 18 3 ,0.0001 17 2 ,0.0 001
ACEi/ARB use 32 3 ,0.0001 46 8 ,0.0 001
Lipid medications 16 7 ,0.0001 47 19 ,0.0 001
Hypertension 39 16 ,0.0001 54 24 ,0.0001
SBP (mmHg) 116 6 13 114 6 12 0.03 116 6 13 116 6 13 0.35
DBP (mmHg) 77 6 9796 9 0.00 03 74 6 9786 9 ,0.0 001
Total cholesterol (mg/dL) 174 6 33 192 6 37 ,0.0001 163 6 33 186 6 33 ,0.0 001
HDL choleste rol (mg/dL) 56 6 16 51 6 15 ,0.0001 61 6 20 56 6 19 ,0.0001
LDL cholesterol (mg/dL) 99 6 28 116 6 33 ,0.0001 86 6 30 108 6 31 ,0.0001
Triglycerides (mg/dL)* 81 (41190) 111 (52298) ,0.0001 68 (35168) 98 (47263) ,0.0001
Data are mean 6 SD or % unless otherwise indicate d. MA, microalbuminuria; NA, not applicable. *Geometric mean (5th95th percentile).
3680 DIABETES CARE, VOLUME 36, NOVEMBER 201 3 care.diabetesjournals.org
Reduced insulin resistance and DN
between ISI or HbA
1c
and rapid G FR de-
cline when calculated by CKD-EP I creat-
inine (P = 0.38) or combined creatinine
and cystatin C (P = 0.50).
CONCLUSIONSdA major challenge
in preventing DN is the difculty in
accurately identifyin g high-risk patients
and the need for additional therapeutic
tar gets. The aim of this study was to
evaluate the association of insulin sensi-
tivi ty and DN. The data show that greater
insulin sensitivity at baseline indepen-
dently predicts lower risk of developing
ACR $30 mg/g and confers protection
from a rapid decline of GFR as calculated
by CKD-EPI cysta tin C.
The association betw een reduced in-
sulin sensitivity and diabetes complica-
tions is increasingly recogn ized, but it is
not a recent discovery. In 1968, Martin
and Stocks (27) showed that microvascu-
lar complicat ions were associ ate d with re-
duced insulin sensitivity in long-standing
type 1 diabetes. In 1993, Yip et al. (28)
explored insulin resistance as an underlying
factor in type 1 diabetes and found reduced
insulin sensitivity, measured by GDR, in
asmallgroupwithmicroalbuminuria,
whereas Orchard et al. (13) later demon-
strated that eGDR (a marker of insulin sen-
sitivity) predicted overt nephropathy in
type 1 diabetic subjects in the Pittsburgh
EDC cohort. The utility of eGDR as a
marker of microvascular complications
in type 1 diabetes was also conrmed by
Chillarón et al. (29) and Girgis et al. (30) in
two smaller cross-sectional studies in 91
and 61 subjects, respectively.
The exact mechanism of reduced in-
sulin sensitivity in type 1 diabetes is
poorly understoo d. One hypothesis sug-
gests that reduced insulin sensitivity is
secondary to prolonged exposure to su-
praphysiologic levels of exogenous insulin,
which has been shown to be associated
with increased ectopic fat accumulation in
the liver and skeletal muscles, increased
oxidative stress, and decreased mitochon-
drial biogenesis (31,32). One possible
mechanistic pathway linking reduced insu-
lin sensitivity to DN in type 1 diabetes is
through reduced insulin sensitivity effects
on overall nonessential fatty acid exposure
and lipotoxicity in the development of
macro- and microangiopathy.
The present study is similar to that of
Orchard et al. (13) but with a larger
sample and contemporary equations for
estimating GFR and insulin sensitivity
and the inclusion of nondiabetic subjects
in the analyses. The present study popu-
lation was also 10 years older than the
Pittsburgh EDC study population and
had a lower mean HbA
1c
of 7.9%. In ad-
dition, the present study uses the ISI, an
equation derived from the study of Snell-
Bergeon et al. (19), which is the largest
euglycemic-hyperinsulinemic clamp
study in adults with and without type 1
diabetes and improves on the adjusted R
2
increase from the Pittsbur gh EDC study
(from 0.570.63). Microalbuminuria is
still recognized as the earliest stage of
DN in type 1 diabetes (33), but evidence
shows that ACR $30 mg/g does not in-
exorably lead to prog ression of DN (34).
For that reason, we decided to add a sec-
ond recognized phenotype of early DN:
rapid G FR decline, which some believe
to be a better marker of trajectory toward
impaired renal fun ction (25). Annual
screening for albuminuria a nd eGFR is
recommended by the American Diabetes
Association (35).
We calculated a rapid decline in
kidney function with all three of the
CKD-EPI equations (20). Concordance
between CKD-EPI cystatin C and creati-
nine eGFR has been reported to be as low
as 62% (36), and CKD-EPI cystatin C
eGFR is considered to be less biased by
age and weight than creatinine-based
measurements (22). Consistent with the
present data, cystatin C has been shown
to be more accurate in detecting rapid
GFR d ecline than creatinine-based mea-
surements in type 1 diabetic subjects with
normal GFR levels (37). Furthermore,
rapid GFR decline esti mated by cystatin
C has also been shown to be associated
with a higher risk for cardiovascular com-
plications and mortality than GFR esti-
mated by creatinine (21,36). This is also
consistent with our ndings from the
CACTI cohort, where eGFR calculated
by cystatin C predicted coronary art ery
calcication progress ion better than
eGFR calculated by the CKD-EPI creatinine
or combined equations (8). These ndings
agree with the present data, where only
rapid GFR decline calculated by CKD-EPI
cystatin C eGFR was associated with ISI in
subjects with type 1 diabetes.
There are limitations to the current
study, including the observational design
and no direct measurements of GFR or
insulin sensitivity because of the large co-
hort size. However, we used all three of
the recently published CKD-EPI equations.
Table 2dMultivariable analysis predicting development of microalbuminuria
Microalbuminuria (ACR $30 mg/g) P value
LDL cholesterol Did not enter model d
HDL choleste rol Did not enter model d
Total cholesterol Did not enter model d
SBP Did not enter model d
BMI Did not enter model d
HbA
1c
1.31 (0.981.75) 0.07
Duration of diabetes Did not enter model d
Age Did not enter model d
ISI 0.69 (0.510.93) 0.01
Data are odds ratio (95% CI).
Table 3dMultivariable analysis predicting development of rapid GFR decline
(>3 mL/min/1.73 m
2
/year)
CKD-EPI cystatin C P value
LDL cholesterol Did not enter model d
HDL choleste rol Did not enter model d
Total cholesterol Did not enter model d
SBP Did not enter model d
BMI Did not enter model d
HbA
1c
1.49 (1.201.87) 0.0004
Duration of diabetes Did not enter model d
Age 1.05 (1.011.08) 0.006
ISI 0.80 (0.670.97) 0.02
Data are odds ratio (95% CI).
care. diabetesjournals.org DIABETES CARE, VOLUME 36, NOVEMBER 2013 3681
Bjornstad and Associates
It should be noted that eGFR at higher
levels is associated with greater variability;
however, these equations are state of the
art and used by such studies as the Di-
abetes Control and Complications Trial/
Epidemiology of Diabetes Interventions
and Complications (DCCT/EDIC) (38).
Moreover, cystatin Cbased equations
have been shown to estimate GFR well in
diabetic subjects with normal and elevated
renal function (37,39). In addition, the ISI
has strong agreement with GDR measured
by the gold standard method in the CACTI
clamp study (R
2
= 0.63), thereby support-
ing its role as a true reection of insulin
sensitivity. There was insufcient power
to assess the ability of ISI to predict risk
of developing microalbuminuria in con-
trol subjects and the categorical develop-
ment of eGFR ,60 mL/min/1.73 m
2
in
both groups because these end points de-
veloped in very few subjects in the cohort.
The subjects with type 1 diabetes without
complete data at baseline had worse renal
function and lipid panel ndings, which
may bias the results to the null because less
healthy subjects with type 1 diabetes were
not included in the analyses.
In summary, greater insulin sensitiv-
ity as measured by ISI at baseline appears
to be protective against the development
of microalbuminuria and rapid eGFR de-
cline as estimated b y cystatin C. Des pit e
the Bypass Angi opla sty Revasculari zati on
Investigati on 2 Diabetes (BARI 2D) study
(40) showing no benet of an insulin sen-
sitizing strategy on nephropathy in sub -
jects with type 2 diabetes, modication of
insu lin sensi tivity holds promise as a ther-
apeutic target to reduce vascular compli-
cations in type 1 diabetes because both
lifestyle c hanges (diet and exercise) and
medications, such as metformin, can im-
prove insulin sensitivity. Further research
is requ ired to bett er assess the role of in-
sulin sensitivity in DN i n typ e 1 diabetes,
especially given the increased incidence of
obesity in people with type 1 diabetes.
AcknowledgmentsdSupport for this study
was provided by National Heart, Lung, and
Blood Institute grant R01 HL113029, Diabetes
Endocrinology Research Center Clinical
Investigation Core grant P30-DK-57516,
and JDRF grant 17-2013-313. The study was
performed at the Adult Clinical and Trans-
lational Research Center at the University of
Colorado Denver with support from National
Institutes of Health grant M01-RR-00051, at
the Barbara Davis Center for Childhood Di-
abetes, and at the Colorado Heart Imaging
Center in Denver. J.K.S.-B. was supported by
an American Diabetes Association Junior Fac-
ulty Award (1-10-JF-50). D.M.M. was sup-
ported by a grant from the National Institute of
Diabetes and Digestive and Kidney Diseases
(DK-075360).
R.J.J. has patent applications related to th e
lowering of uric acid and blocking fructose
metabolism as a me ans for slowing DN or
improving insulin resistanc e and has shares
with XORT Therapeutic s related to these pat-
ents. No othe r potential conicts of interest
relevant to this article were reported.
P.B. contributed to the research and dis-
cussion and wrote, reviewed, and edited the
manuscript. J.K.S.-B. formulated the I SI
model, contributed to the research and dis-
cussion, and reviewe d and edited the manu-
script. M.R. designed the CACTI study,
contributed to the research and discussion,
and reviewed and edited the manus cript. D.J.,
M.B.C., and R.J.J. contributed to the disc us-
sion and reviewed and edited the ma nuscript.
D.M.M. contributed to th e research and dis-
cussion and revie wed and edited the manu-
script. D.M.M. is the guarantor of this work
and, as such, had full access to all the data in
the study and takes responsibility f or the in-
tegrity of the data and the accuracy of the data
analysis.
Parts of this study were presented in ab stract
form at the 73rd Scientic Sessions of the
American Diabetes Association, Chicago, Illinois,
2125 June 2013.
References
1. Guilbert JJ. The World Health Report
2006: working together for health. Educ
Health (Abingdon) 2006;19:385387
2. Molitch ME, DeFronzo RA, Franz MJ,
Keane WF, Mogensen CE, Parving HH.
Diabetic nephropathy. Diabetes Care
2003;26(Suppl. 1):S94S98
3. Collins AJ, Foley RN, Herzog C, et al. US
Renal Data System 2010 annual data re-
port. Am J Kidney Dis 2011;57(1 Suppl.
1):A8, e1e526
4. Maahs DM, Rewers M. Editorial: mortal-
ity and renal disea se in type 1 diabetes
mellitusprogress made, more to be do ne.
J Clin Endocrinol Metab 2006;91:3757
3759
5. Orchard TJ, Secrest AM, Miller RG, Costacou
T. In the absence of renal disease, 20 year
mortality risk in type 1 diabetes is com-
parable to that of the general population:
a report from the Pittsburgh Epidemiology
of Diabetes Complications Study. Dia-
betologia. 2010;53:23122319
6. Borch-Johnsen K, Kreiner S. Proteinuria:
value as pr edictor of cardiovascular mor-
tality in insulin dependent diabetes mel-
litus. Br Med J (Clin R es Ed) 1987;294:
16511654
7. Orchard TJ, Olson JC, Er bey JR, et al.
Insulin resistance-related factors, but not
glycemia, predict coronary artery disease
in type 1 diabetes: 10-year follow-up data
from the Pittsburgh Epidemiology of Di-
abetes Complications Study. Diabetes
Care 2003;26:13741379
8. Maahs D, Jalal D, Chonchol M, Johnson R,
Rewers M, Snell-Bergeon JK. Impaired
renal function further increases odds of
6-year coronary artery calcication pro-
gression in adults with type 1 diabetes: the
CACTI study. Diabetes Care 2013;36:
26072614
9. Diabetes Epidemiology Research
International Mortality Study Group.
International evaluation of cause-specic
mortality and IDDM. Diabetes Care
1991;14:5560
10. Maahs DM. Ca rdiov ascular disease (CVD)
limbo: how soon and low should we go to
prevent CVD in diabetes? Diabetes Tech-
nol Ther 2012;14:449452
11. Soedamah-Muthu SS, Chaturvedi N, Toeller
M, et al. Risk factors for coronary heart
disease in type 1 diabetic patients in Europe:
the EURODIAB Prospective Complications
Study. Diabetes Care 2004;27:530537
12. Kilpatrick ES, Rigby AS, Atkin SL. Insulin
resistance, the metabolic syndro me, and
compl ication risk in type 1 diabetes:
double diabetes in the Diabetes Control
and Complications Trial. Diabetes Care
2007;30:70771 2
13. Orchard TJ, Cha ng YF, Ferr ell RE, Petro
N, Ellis DE. Nephropathy in type 1 di-
abetes: a manifestation of insulin resistance
and multiple genetic susceptibilities? Fur-
ther evidence from the Pittsburgh Epide-
miology of Diabetes Complication Study.
Kidney Int 2002;62:963970
14. Yki-Jarvinen H, Koivisto VA. Natural
course of insulin resistance in type I di-
abetes. New Engl J Med 1986;315:224
230
15. Amiel SA, Sherwin RS, Simonson DC,
Lauritano AA, Tamborlane WV. Impaired
insulin action in puberty. A contributing
factor to poor glycemic control in ado-
lesce nts with diabetes. New Engl J Med
1986;315:215219
16. Nadeau KJ, Regensteiner JG, Bauer TA,
et al. Insulin resistance in adolescents with
type 1 diabetes and its relation ship to
cardiovascular function. J Clin Endocrinol
Metab 2010;95:513521
17. Rodrigues TC, Veyna AM, Haarhues MD,
Kinney GL, Rewers M, Snell-Bergeon JK.
Obesity and coronary artery calcium in
diabetes: the Coronary Artery Calcica-
tion in Type 1 Diab etes (CACTI) study.
Diabetes Technol Ther 201 1;13:991996
18. Schauer IE, Snell-Bergeon JK, Bergman
BC, et a l. Insulin resis tance, d efective
insulin-mediated fatty acid suppres-
sion , and coronary artery calcication in
subjects with and without type 1 diabetes:
the CACTI study. Diabetes 2011;60:306
314
19. Snell-Bergeon JK, Maahs DM, Schauer IE,
Bergm an BC, Rewers M. A method for
estimating insulin sensitivity in adul ts
3682 DIABETES CARE, VOLUME 36, NOVEMBER 201 3 care.diabetesjournals.org
Reduced insulin resistance and DN
with type 1 diabetes. Presented at the 70th
Annual Meeting of the American Diabetes
Association, 2529 June 2010, Orlando,
Florida
20. Inker LA, Schmid CH, Tighiouart H, et al.;
CKD-EPI Investigators. Estimating glo-
merular l tration rate from serum creati-
nine and cystatin C. New Engl J Med
2012;367:2029
21. Shlipak MG, Katz R, Kestenbaum B, et al.
Rapid decline of kidney function increases
cardiovascular risk in the elderly. J Am
Soc Nephrol 2009;20:26252630
22. Shlipak MG, Katz R, Kestenbaum B, et al.
Rate of kidney function decline in older
adults: a comparison using creatinine and
cystatin C. Am J Nephrol 2009;30:171
178
23. Rifkin DE, Shlipak MG, Katz R, Fri ed LF,
Siscovick D, Chonchol M, et al. Rapid
kidney function decline a nd mortality risk
in older adults. Arch Intern Med 2008;
168:22122218
24. Maahs DM, Kinney GL, Wadwa P, et al.
Hypertension prev alence, awareness,
treatment, and control in an adult type 1
diabetes population and a comparable
general popul ation. Diabetes Care 2005;
28:301306
25. Perkins BA, Fi cociello LH, Ostrander BE,
et al. Microalbuminuria and the risk for
early progressive renal function decline in
type 1 diabetes. J Am Soc Nephrol 2007;
18:13531361
26. Maahs DM, Jalal D, McFann K, Rewers M,
Snell-Bergeon JK. Systematic shifts in
cystatin C between 2006 and 2010. Clin J
Am Soc Nephrol 2011;6:19521955
27. Martin FI, Stocks AE. Insulin sensitivity
and vascular disease in ins ulin-dependent
diabetics. Br Med J 1968;2:8182
28. Yip J, Mattock MB, Moroc utti A, Sethi M,
Trevisan R, Viberti G. Insulin resistance in
insulin-dependent diabetic patients with
microalbuminuria. Lancet 1993;342:
883887
29. Chilla rón JJ, Goday A, Flores-Le-Roux JA,
et al. Estimated glucose disposal rate i n
assessment of the metabolic syndrome
and microvascular complications in pa-
tients with type 1 diabetes. J Clin Endo-
crinol Metab 2009;94:3530 3534
30. Girgis CM, Scalley BD, Park KE. Utility of
the estimated glucose disposal rate as a
marker of microvascular complications in
young adults with type 1 diabetes. Di-
abetes Res Clin Pract 2012;96:e7 0e72
31. Perseghin G, Lattuada G, Danna M, et al.
Insulin resistance, intramyocellul ar lipid
content, and plasma adiponec tin in pa-
tients with type 1 diabetes. Am J Physiol
Endocrino l Metab 2003;285:E1174
E1181
32. Houstis N, Rosen ED, Lander ES. Reactive
oxyge n spec ies have a causal role in mul-
tiple forms of insulin resistance. Nature
2006;440:944948
33. Standa rds of medic al care in diabetes
2013. Diabetes Care 2013;36 (Suppl. 1):
S11S66
34. de Boer IH, Rue TC, Cleary PA, et al.;
Diabetes Control and Complications Trial/
Epidemiology of Diabetes Interventions
and Complications Study Researc h
Group. Long-term renal outcomes of pa-
tients with type 1 diabetes mellitus and
microalbuminuria: an analysis of the Di-
abetes Control and Complications Trial/
Epidemiology of Diabetes Interventions
and Complications cohort. Arch Intern
Med 2011;171:412420
35. Ceriello A, Novials A, Ortega E, et al. Ev-
idence that hyperglycemia after recovery
from hypoglycemia worsens endothelia l
function and increases oxidative stress
and inammati on in hea lthy control
subje cts and subjects with type 1 diabetes.
Diabetes 2012;61:2993
2997
36. Krolewski AS, Warram JH, Forsbl om C,
et al. Serum concentration of cystatin C
and risk of end-stage renal disease in di-
abetes. Diabetes Care 2012;35:2311
2316
37. Premaratne E, MacIsaac RJ, Finch S,
Panagiotopoulos S, Ekinci E, Jerums G.
Serial measurements of cystatin C are
more accurate than creatinine-based meth-
ods in detecting declining renal fu nction
in type 1 diabetes. Di abetes Care 2008;31:
971973
38. de Boer IH, Sun W, Clear y PA, et al.;
DCCT/EDIC Research Group. Intensive
diabetes therapy and glomerular ltration
rate in type 1 diabetes. N Engl J Med 2011;
365:23662376
39. Macisaac RJ, Tsalamandris C, Thomas MC,
et al. Estimating glomerular ltration rate in
diabetes: a comparison of cystatin-C- and
creatinine-based methods. Diabetologia
2006;49:16861689
40. Frye RL, August P, Brooks MM, et al.
A randomiz ed trial of th erapies for type
2 diabetes and coronary artery disease.
N Engl J Med 2009;360:2 5032515
care. diabetesjournals.org DIABETES CARE, VOLUME 36, NOVEMBER 2013 3683
Bjornstad and Associates
... Type 2 diabetes mellitus (T2DM) is closely associated with insulin resistance (IR), and DKD is no exception [5,6]. IR refers to the decreased or impaired responsiveness of insulin-targeted tissues to physiological levels of endogenous and exogenous insulin [5]. ...
Article
Full-text available
Purpose This retrospective study aimed to investigate the relationship between alternative insulin resistance (IR) indexes not reliant on insulin and diabetic kidney disease (DKD) incidence in a newly diagnosed cohort of individuals with type 2 diabetes mellitus (T2DM). Methods We conducted a retrospective analysis of baseline characteristics in a cohort of 521 individuals with T2DM, then followed up on the outcome of DKD. To assess the predictive ability of IR indexes, we compared the performance of four non-insulin-based IR indexes and the homeostasis model for insulin resistance (HOMA-IR) using logistic regression and consistency-statistics (C-statistics). Furthermore, we computed the net reclassification index (NRI) and integrated discrimination improvement (IDI) to evaluate the additional effects of the indexes. Results The four alternative IR indexes of DKD patients were significantly higher than those of non-DKD. After adjustment for other variables, the highest tertile of all indexes was significantly related to DKD incidence, compared with the lowest tertile. Furthermore, the C-statistics for the triglyceride-glucose index (TyG index) and triglyceride to high-density lipoprotein ratio (TG/HDL) were all 0.652, while triglyceride glucose-body mass index (TyG-BMI) and metabolic score for insulin resistance (METS-IR) were 0.639 and 0.651, respectively. The incorporation of the alternative IR indexes into the baseline model revealed positive additional effects, leading to an improved prediction of the risk for DKD. Conclusions It was discovered that the alternative IR indexes served as independent risk factors of DKD. Among the four alternative indexes, TyG index and TG/HDL had the best prediction performance for DKD, followed by METS-IR.
... 8 Various studies have observed correlations of reduced IS with microalbuminuria and predicted glomerular function decline over 6 years and have also predicted remission of albuminuria in adults with T1DM following improvement in insulin sensitivity. [9][10][11] Similarly, the odds of developing diabetic retinopathy and neuropathy are predicted by baseline estimated insulin sensitivity. 10,11 Methodology In this narrative review article, we focus our attention on pathophysiology of IR in T1DM and techniques for identifying those at risk to put into action various strategies for management of the same. ...
Article
Full-text available
Though insulin resistance (IR) was previously considered a feature of only type 2 Diabetes (T2DM), its development in type 1 Diabetes (T1DM) is not an uncommon occurrence, the causes of which are multifactorial (gender, pubertal status, diabetes duration, ethnicity, genetics, adiposity, glycemic control, chronic inflammation). Despite improvements in glucose, blood pressure and lipid profile, vascular complications (coronary artery disease and nephropathy) continue to remain common causes of morbidity and mortality in T1DM. Aggressive glycemic control reduces but does not eliminate the risk of IR. IR accelerates the development of micro and macrovascular complications, many of which can be potentially reversed if diagnosed and managed early. Lack of endogenous insulin production makes estimation of insulin sensitivity in T1DM difficult. As hyperinsulinemic-euglycemic clamp studies are cumbersome and invasive, the use of prediction equations for calculating estimated insulin sensitivity may prove to be useful. Along with intensive insulin therapy, dietary modifications and increasing physical activity, the role of Metformin in managing IR in T1DM is becoming increasingly popular. Metformin adjunct therapy in T1DM has been shown to improve insulin sensitivity, glycemic control, lipid profile, body composition, vascular smooth muscle function, thereby reducing the risk of vascular complications, as well as reversal of early vascular dysfunction. However, further studies to assess long-term efficacy and safety of Metformin use in adolescents and youth with T1DM are needed. This review aims at revisiting the pathophysiology of IR in T1DM and techniques of identifying those at risk so as to put into action various strategies for management of the same.
... Furthermore, an evening exercise bout with its insulin sensitising effects may improve insulin sensitivity at a time in which it is typically worsened (41). Increased insulin sensitivity is an important therapeutic target to reduce the risk of macro- (42) and microvascular (43) complications in type 1 diabetes. ...
Article
Full-text available
The scientific literature shows that exercise has many benefits for individuals with type 1 diabetes. Yet, several barriers to exercise in this population exist, such as post-exercise hypoglycaemia or hyperglycaemia. Several studies suggest that the timing of exercise may be an important factor in preventing exercise-induced hypoglycaemia or hyperglycaemia. However, there is a paucity of evidence solely focused on summarising findings regarding exercise timing and the impact it has on glucose metabolism in type 1 diabetes. This report suggests that resistance or high-intensity interval exercise/training (often known as HIIT) may be best commenced at the time of day when an individual is most likely to experience a hypoglycaemic event (i.e., afternoon/evening) due to the superior blood glucose stability resistance and HIIT exercise provides. Continuous aerobic-based exercise is advised to be performed in the morning due to circadian elevations in blood glucose at this time, thereby providing added protection against a hypoglycaemic episode. Ultimately, the evidence concerning exercise timing and glycaemic control remains at an embryonic stage. Carefully designed investigations of this nexus are required, which could be harnessed to determine the most effective, and possibly safest, time to exercise for those with type 1 diabetes.
Article
Background Diabetic nephropathy (DN) is a complication caused by diabetes. Circular RNAs (circRNAs) are a kind of RNA with a closed circular structure, which has high stability and is involved in many disease‐related processes. The mechanism of circRNA TAO kinase 1 (circTAOK1) in the pathogenesis and development of DN is unclear. Methods CircTAOK1, microRNA (miR)‐142‐3p, and sex‐determining region Y‐box transcription factor 6 (SOX6) mRNA levels were analyzed by real‐time quantitative polymerase chain reaction (RT‐qPCR). Cell counting kit‐8 (CCK8) and 5‐ethynyl‐2′‐deoxyuridine (EdU) assays were used to analyze cell proliferation. Cell cycle distribution was detected by flow cytometry. Western blot assay was performed to test B‐cell lymphoma 2 (Bcl‐2), Bcl‐2 associated X (Bax), cleaved‐caspase 3, and fibronectin (FN), collagen I (Col I), and collagen IV (Col IV) protein levels. ELISA assay was used to measure interleukin 1β (IL‐1β), interleukin 6 (IL‐6), and tumor necrosis factor (TNF‐α) levels. The reactive oxygen species (ROS) and malondialdehyde (MDA) levels and the superoxide dismutase (SOD) activity were assessed by the corresponding kits. And the correlation between miR‐142‐3p and circTAOK1 or SOX6 was confirmed by dual luciferase reporter assay, RNA immunoprecipitation assay and RNA pull down assay. Results CircTAOK1 and SOX6 expression levels were up‐regulated, while miR‐142‐3p expression was down‐regulated in DN serum and HG‐treated HK‐2 cells. Knockdown of circTAOK1 could inhibit cell injury of HG‐induced HK‐2 cells. The inhibitory effect of circTAOK1 knockdown on HG‐induced HK‐2 cell injury was restored by miR‐142‐3p downregulation. CircTAOK1 acted as a sponge for miR‐142‐3p, and SOX6 was targeted by miR‐142‐3p. The overexpression of SOX6 could recover the effect of miR‐142‐3p overexpression on HG‐induced HK‐2 cell injury. CircTAOK1 regulated the expression of SOX6 by targeting miR‐142‐3p. Conclusion CircTAOK1 knockdown inhibited HG‐induced HK‐2 cell damage in DN by the miR‐142‐3p/SOX6 axis.
Article
Aim: This study aimed to determine whether the insulin resistance (IR) and lipid profiles in Type 1 Diabetes (T1D) offspring are associated with IR and other cardiovascular risk factors in their parents. Methods: This study included 99 T1D patients (19.6 ± 4.0 yrs.), 85 mothers and 60 fathers. Parents' IR was assessed by HOMA-IR, and the insulin sensitivity in T1D patients was assessed by the estimated Glucose Disposal Rate (eGDR). Results: The eGDR in the T1D offspring was negatively related to age (p = 0.023), weight (p = 0.004), LDL (p = 0.026), and microalbuminuria (p = 0.019). Maternal Type 2 Diabetes (p < 0.001) and HOMA-IR (p = 0.029) were negatively related to eGDR in their T1D offspring. The maternal HOMA-IR and the proband's eGDR were positively (p = 0.012) and negatively (p = 0.042) associated with the birth weight of the T1D offspring, respectively. We didn't find an association with the fathers' profiles. Conclusions: In a cohort of offspring with T1D the insulin sensitivity was related to the IR, lipid profile, and the presence of T2D only in their mothers. Precocious screening and treatment of these risk factors beyond glycemic control will benefit T1D with this background.
Chapter
The majority of patients with type 2 diabetes (T2DM) eventually succumb to a cardiovascular event, and dyslipidemia, present in more than half of all patients, is one of the causes of cardiovascular morbidity and mortality. In addition to lifestyle changes, a number of lipid-lowering medications have been shown to be effective in decreasing cardiovascular risk. In this review, we discuss several classes of medications that primarily target LDL cholesterol and have been shown to reduce cardiovascular events. These include the statins, ezetimibe, PCSK9 inhibitors, and bempedoic acid. Agents that do not target LDL cholesterol such as fibrates and omega-3 fatty acids also show promise, although they need additional study. Finally, a neglected area of diabetic dyslipidemia management is the role of diabetes-specific pharmacotherapy. Several drug classes, including metformin, GLP-1 receptor agonists, and thiazolidinediones, target lipoprotein abnormalities and have been shown to reduce cardiovascular risk. Judicious use of these medications in conjunction with lipid-specific agents will contribute to cardiovascular risk reduction.KeywordsCholesterolTriglyceridesHDL cholesterolLDL cholesterolFibratesStatinsPCSK9 inhibitorsOmega-3 fatty acids
Chapter
Diabetes is the most common cause of end-stage kidney disease (ESKD) in the USA and other developed countries. About 50% of the patients with type 1 diabetes (T1D) and 30% of those with type 2 diabetes (T2D) develop chronic kidney disease (CKD) (Afkarian, et al., J Am Soc Nephrol. 24:302–8, 2013; Fox, et al., Lancet. 380:1662–73, 2012). Diabetic nephropathy is a chronic condition that develops over many years, and it is characterized by a gradual increase in urinary albumin excretion, blood pressure levels and cardiovascular risk, declining glomerular filtration rate (GFR), and eventual ESKD. Annual screening for CKD should include both albuminuria measurements and estimates of GFR. Diabetic nephropathy manifests as a constellation of kidney structural changes considered unique to this disease, which closely correlate with kidney function. Multiple factors are associated with CKD in diabetes, and patients with diabetes often require multiple therapies aimed at prevention of progressive CKD and its associated comorbidities and mortality. Management of cardiorenal risk factors, including lifestyle modifications (diet, exercise, and cessation of smoking); glucose, blood pressure, and lipid control; use of agents blocking the renin-angiotensin-aldosterone system; and use of SGLT2 inhibitors in patients with T2D and other agents with proven renal or cardiovascular benefit are the cornerstones of therapy.KeywordsDiabetic nephropathy diabetic kidney diseaseChronic kidney disease in diabetesRenal function lossGlomerular filtration rate declineAlbuminuriamicroalbuminuriaProteinuria
Article
Background: We compared plasma metabolites of amino acid oxidation and the tricarboxylic acid (TCA) cycle in youth with and without type 1 diabetes mellitus (T1DM) and related the metabolites to glomerular filtration rate (GFR), renal plasma flow (RPF), and albuminuria. Metabolites associated with impaired kidney function may warrant future study as potential biomarkers or even future interventions to improve kidney bioenergetics. Methods: Metabolomic profiling of fasting plasma samples using a targeted panel of 644 metabolites and an untargeted panel of 19,777 metabolites was performed in 50 youth with T1DM ≤ 10 years and 20 controls. GFR and RPF were ascertained by iohexol and p-aminohippurate clearance, and albuminuria calculated as urine albumin to creatinine ratio. Sparse partial least squares discriminant analysis and moderated t tests were used to identify metabolites associated with GFR and RPF. Results: Adolescents with and without T1DM were similar in age (16.1 ± 3.0 vs. 16.1 ± 2.9 years) and BMI (23.4 ± 5.1 vs. 22.7 ± 3.7 kg/m2), but those with T1DM had higher GFR (189 ± 40 vs. 136 ± 22 ml/min) and RPF (820 ± 125 vs. 615 ± 65 ml/min). Metabolites of amino acid oxidation and the TCA cycle were significantly lower in adolescents with T1DM vs. controls, and the measured metabolites were able to discriminate diabetes status with an AUC of 0.82 (95% CI: 0.71, 0.93) and error rate of 0.21. Lower glycine (r:-0.33, q = 0.01), histidine (r:-0.45, q < 0.001), methionine (r: -0.29, q = 0.02), phenylalanine (r: -0.29, q = 0.01), serine (r: -0.42, q < 0.001), threonine (r: -0.28, q = 0.02), citrate (r: -0.35, q = 0.003), fumarate (r: -0.24, q = 0.04), and malate (r: -0.29, q = 0.02) correlated with higher GFR. Lower glycine (r: -0.28, q = 0.04), phenylalanine (r:-0.3, q = 0.03), fumarate (r: -0.29, q = 0.04), and malate (r: -0.5, q < 0.001) correlated with higher RPF. Lower histidine (r: -0.28, q = 0.02) was correlated with higher mean ACR. Conclusions: In conclusion, adolescents with relatively short T1DM duration exhibited lower plasma levels of carboxylic acids that associated with hyperfiltration and hyperperfusion. Trial registration: ClinicalTrials.gov NCT03618420 and NCT03584217 A higher resolution version of the Graphical abstract is available as Supplementary information.
Article
Full-text available
OBJECTIVE To determine whether baseline estimated glomerular filtration rate (eGFR) and albumin-to-creatinine ratio (ACR) independently predict coronary artery calcification (CAC) progression, and to determine how eGFR changes over 6 years in adults with type 1 diabetes compared with nondiabetic adults.RESEARCH DESIGN AND METHODS The Coronary Artery Calcification in Type 1 Diabetes study participants (n = 1,066) with complete data for eGFR assessment at baseline and 6 years were included. Three Chronic Kidney Disease Epidemiology Collaboration equations (serum creatinine, cystatin C, and both) were used to estimate eGFR. The association of baseline ACR and eGFR with CAC progression was analyzed using multiple logistic regression.RESULTSIncreasing categorical baseline ACR (<10, 10-30, and >30 µg/mg) predicted CAC progression in participants with type 1 diabetes (odds ratio [OR], 2.15; 95% CI, 1.50-3.09; 7.19 [3.90-13.26]; and 18.09 [8.48-38.62]), respectively, compared with nondiabetic subjects. Baseline eGFR <60 mL/min/1.73 m(2) also predicted CAC progression (OR, 5-7, compared with nondiabetic participants). ORs for CAC progression were higher in women than in men when using the cystatin C-based Chronic Kidney Disease Epidemiology Collaboration equations. Participants with type 1 diabetes had greater eGFR decreases over 6 years than nondiabetic participants using cystatin C-based equations.CONCLUSIONS Although increasing ACR or decreasing eGFR predicts CAC progression, coronary atherosclerosis progresses faster in people with type 1 diabetes even in the absence of diabetic kidney disease. These findings emphasize the interaction between kidney disease and cardiovascular disease in type 1 diabetes and highlight the public health importance of lowering cardiorenal risk in people with type 1 diabetes.
Article
Full-text available
Currently there is debate on whether hypoglycemia is an independent risk factor for atherosclerosis, but little attention has been paid to the effects of recovery from hypoglycemia. In normal control individuals and in people with type 1 diabetes, recovery from a 2-h induced hypoglycemia was obtained by reaching normoglycemia or hyperglycemia for another 2 h and then maintaining normal glycemia for the following 6 h. Hyperglycemia after hypoglycemia was also repeated with the concomitant infusion of vitamin C. Recovery with normoglycemia is accompanied by a significant improvement in endothelial dysfunction, oxidative stress, and inflammation, which are affected by hypoglycemia; however, a period of hyperglycemia after hypoglycemia worsens all of these parameters, an effect that persists even after the additional 6 h of normoglycemia. This effect is partially counterbalanced when hyperglycemia after hypoglycemia is accompanied by the simultaneous infusion of vitamin C, suggesting that when hyperglycemia follows hypoglycemia, an ischemia-reperfusion-like effect is produced. This study shows that the way in which recovery from hypoglycemia takes place in people with type 1 diabetes could play an important role in favoring the appearance of endothelial dysfunction, oxidative stress, and inflammation, widely recognized cardiovascular risk factors.
Article
Full-text available
OBJECTIVE Patients with diabetes have a high risk of end-stage renal disease (ESRD). We examined whether prediction of this outcome, according to chronic kidney disease (CKD) staging by creatinine-based estimates of the glomerular filtration rate (eGFRcreat), is improved by further staging with serum cystatin C–based estimates (eGFRcyst). RESEARCH DESIGN AND METHODS Patients with diabetes in CKD stages 1–3 were selected from three cohorts: two from Joslin Diabetes Center, one with type 1 diabetes (N = 364) and one with type 2 diabetes (N = 402), and the third from the Finnish Diabetic Nephropathy (FinnDiane) Study of type 1 (N = 399). Baseline serum concentrations of creatinine and cystatin C were measured in all patients. Follow-up averaged 8–10 years and onsets of ESRD (n = 246) and death unrelated to ESRD (n = 159) were ascertained. RESULTS Although CKD staging by eGFRcyst was concordant with that by eGFRcreat for 62% of Joslin patients and 73% of FinnDiane patients, those given a higher stage by eGFRcyst than eGFRcreat had a significantly higher risk of ESRD than those with concordant staging in all three cohorts (hazard ratio 2.3 [95% CI 1.8–3.1]). Similarly, patients at a lower stage by eGFRcyst than by eGFRcreat had a lower risk than those with concordant staging (0.30 [0.13–0.68]). Deaths unrelated to ESRD followed the same pattern, but differences were not as large. CONCLUSIONS In patients with diabetes, CKD staging based on eGFRcyst significantly improves ESRD risk stratification based on eGFRcreat. This conclusion can be generalized to patients with type 1 and type 2 diabetes and to diabetic patients in the U.S. and Finland.
Article
Objective A cross-cultural study was completed to evaluate differences in mortality patterns in four population cohorts in Japan; Israel; Allegheny County, Pennsylvania; and Finland. Research Design and Methods Cases were diagnosed between 1 January 1965 and 31 December 1979. Mortality was determined as of 1 January 1985. There were 147 deaths occurring in the 8212 insulin-dependent diabetes mellitus (IDDM) patients in the four countries. A standardized protocol for assessing causes of death (cause-specific mortality) was developed; in this article, we report the causes. Results Major overall mortality differences by country appeared, with IDDM subjects in Japan much more likely to die than in the other countries. In Japan, the elevated mortality was the result of acute diabetes-related complications and kidney disease. For all countries, mortality from acute diabetes-related complications accounted for a surprisingly high percentage of deaths (> 25% in each country). A larger percentage of cases in Finland died as a result of suicide than for the other three countries. Conclusions The results suggest that there are major cross-country differences in cause-specific mortality and that much of the premature mortality associated with diabetes is potentially preventable.
Article
Estimates of glomerular filtration rate (GFR) that are based on serum creatinine are routinely used; however, they are imprecise, potentially leading to the overdiagnosis of chronic kidney disease. Cystatin C is an alternative filtration marker for estimating GFR. Using cross-sectional analyses, we developed estimating equations based on cystatin C alone and in combination with creatinine in diverse populations totaling 5352 participants from 13 studies. These equations were then validated in 1119 participants from 5 different studies in which GFR had been measured. Cystatin and creatinine assays were traceable to primary reference materials. Mean measured GFRs were 68 and 70 ml per minute per 1.73 m(2) of body-surface area in the development and validation data sets, respectively. In the validation data set, the creatinine-cystatin C equation performed better than equations that used creatinine or cystatin C alone. Bias was similar among the three equations, with a median difference between measured and estimated GFR of 3.9 ml per minute per 1.73 m(2) with the combined equation, as compared with 3.7 and 3.4 ml per minute per 1.73 m(2) with the creatinine equation and the cystatin C equation (P=0.07 and P=0.05), respectively. Precision was improved with the combined equation (interquartile range of the difference, 13.4 vs. 15.4 and 16.4 ml per minute per 1.73 m(2), respectively [P=0.001 and P<0.001]), and the results were more accurate (percentage of estimates that were >30% of measured GFR, 8.5 vs. 12.8 and 14.1, respectively [P<0.001 for both comparisons]). In participants whose estimated GFR based on creatinine was 45 to 74 ml per minute per 1.73 m(2), the combined equation improved the classification of measured GFR as either less than 60 ml per minute per 1.73 m(2) or greater than or equal to 60 ml per minute per 1.73 m(2) (net reclassification index, 19.4% [P<0.001]) and correctly reclassified 16.9% of those with an estimated GFR of 45 to 59 ml per minute per 1.73 m(2) as having a GFR of 60 ml or higher per minute per 1.73 m(2). The combined creatinine-cystatin C equation performed better than equations based on either of these markers alone and may be useful as a confirmatory test for chronic kidney disease. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases.).