Half of the mortality in diabetes is seen in individuals <50 years of age and commonly predicted by the early onset of diabetic kidney disease (DKD). In type 1 diabetes, increased urinary albumin-to-creatinine ratio (uACR) during adolescence defines this risk, but the pathological factors responsible remain unknown. We postulated that early in diabetes, glucose variations contribute to kidney injury molecule-1 (KIM-1) release from circulating T cells, elevating uACR and DKD risk. DKD risk was assigned in youth with type 1 diabetes (n = 100; 20.0 ± 2.8 years; males/females, 54:46; HbA1c 66.1 [12.3] mmol/mol; diabetes duration 10.7 ± 5.2 years; and BMI 24.5 [5.3] kg/m2) and 10-year historical uACR, HbA1c, and random blood glucose concentrations collected retrospectively. Glucose fluctuations in the absence of diabetes were also compared with streptozotocin diabetes in apolipoprotein E−/− mice. Kidney biopsies were used to examine infiltration of KIM-1–expressing T cells in DKD and compared with other chronic kidney disease. Individuals at high risk for DKD had persistent elevations in uACR defined by area under the curve (AUC; uACRAUC0–10yrs, 29.7 ± 8.8 vs. 4.5 ± 0.5; P < 0.01 vs. low risk) and early kidney dysfunction, including ∼8.3 mL/min/1.73 m2 higher estimated glomerular filtration rates (modified Schwartz equation; Padj < 0.031 vs. low risk) and plasma KIM-1 concentrations (∼15% higher vs. low risk; P < 0.034). High-risk individuals had greater glycemic variability and increased peripheral blood T-cell KIM-1 expression, particularly on CD8+ T cells. These findings were confirmed in a murine model of glycemic variability both in the presence and absence of diabetes. KIM-1+ T cells were also infiltrating kidney biopsies from individuals with DKD. Healthy primary human proximal tubule epithelial cells exposed to plasma from high-risk youth with diabetes showed elevated collagen IV and sodium–glucose cotransporter 2 expression, alleviated with KIM-1 blockade. Taken together, these studies suggest that glycemic variations confer risk for DKD in diabetes via increased CD8+ T-cell production of KIM-1.

The presence of diabetic kidney disease (DKD) is the strongest predictor of both cardiovascular disease (CVD) and premature mortality in all individuals with diabetes (13). In particular, recent evidence highlights that premature mortality seen in individuals less than ∼45 years of age who develop diabetes at a younger age is not being alleviated by modern treatment regimens (4,5). In type 1 diabetes, a greater future risk of DKD and CVD may be evident as early as childhood and adolescence (6,7), detected as high-normal urinary albumin-to-creatinine ratio (uACR) preceding micro- and macroalbuminuria. This rise in uACR is seen in adolescents with type 1 diabetes, predicting some 85% of future case subjects with DKD (6).

The underlying reasons for early elevation in uACR in those at risk for kidney and CVD remain unclear, but dramatically influence mortality in adults <45 years of age with type 1 diabetes (8). Recent longitudinal studies from childhood suggest that uACR increases do not directly relate to traditional risk markers of vascular disease, such as HbA1c in children and adolescents (6,7). HbA1c does not necessarily capture fluctuations in blood glucose concentrations over the course of each day (glycemic variability), postulated as more potent activators of pathological pathways leading to DKD (9), although this remains hotly debated. Continuous blood glucose monitoring does show that individuals with diabetes average ∼50% of their time with glycemic variations outside ranges seen in healthy subjects (10), and those with greatest variability have increased atherosclerotic plaque formation (11). Indeed, there is some evidence that in the long term, glycemic variability is an independent risk factor for DKD progression (1214).

This initial elevation in uACR also commonly precedes changes in other associated risk factors, including hypertension and dyslipidemia (15), but these become evident as kidney disease progresses. Further, early use of ACE inhibition and/or statin therapy, which are best practice in adults, have failed to significantly lower uACR in children and adolescents “at future risk” in a recent phase III clinical trial, the Adolescent Type 1 Diabetes Cardio-Renal Trial (AdDIT) (16). Another study has also shown that lowering blood pressure using renin-angiotensin system blockade in normotensive, normoalbuminuric individuals did not influence the progression of early kidney lesions in young adults with glomerular filtration rates >90 mL/min/1.73 m2 (17).

Taken together, these previous findings suggest that other pathological factors may be at play early during the development of DKD. Circulating markers of inflammation such as hs-CRP in youth (7) and kidney injury molecule-1 (KIM-1 [18]) are elevated early in DKD progression. KIM-1, also known as hepatitis A virus cellular receptor 1 (HAVCR1) and T-cell Ig mucin 1 (TIM-1), is a transmembrane glycoprotein receptor primarily found on T cells (19) and significantly injured proximal tubules of the kidney (20,21). Activation of KIM-1 activates potent stimulatory signals for T-cell proliferation and cytokine production (22), and renal T-cell infiltration is reported in established DKD (23), sparking speculation that activation of T-cell activation, at least in murine models and type 2 diabetes, may play a major role in the initiation of DKD (24,25). Cleaved KIM-1 is also released into urine (26) and used as a biomarker for established chronic kidney disease (CKD) (18), but one study has shown that increases in circulating KIM-1 precede elevations in urinary KIM-1 and predict early progressive decline in kidney function in type 1 (18) and potentially type 2 diabetes (27). The source of circulating KIM-1 early in DKD remains to be elucidated.

Hence, the objective of this study was to examine if there was an early link among glucose variation, T-cell KIM-1 expression, and increased risk of kidney disease, which is examined both in individuals with type 1 diabetes and by using kidney biopsies from individuals with DKD.

Study Design and Participants

A cross-sectional cohort of healthy adolescents/young adults (aged 15–25 years; N = 100) with diagnosed type 1 diabetes of >2 years’ duration were recruited during routine visits to the transitional diabetes clinic at the Mater Young Adult Health Centre. Exclusion criteria were: 1) previous diagnosis of micro/macroalbuminuria by uACR >2.5 mg/mmol in males and >3.5 mg/mmol in females (28) or DKD as defined by the Kidney Health Australia–Caring for Australasians with Renal Impairment algorithm for the early detection of CKD (29); 2) uncontrolled diabetes (defined as HbA1c >9.5% [80 mmol/mol]) or more than two episodes of ketoacidosis in the preceding 12 months; 3) history of severe familial hypercholesterolemia; 4) previous myocardial infarction, stroke, or preexisting kidney disease; 5) pregnancy; 6) any existing medication other than insulin or hormonal contraceptive agents; 7) autoimmune diseases including uncontrolled celiac disease or Addison disease; 8) any congenital condition resulting in insulin-requiring diabetes; 9) diagnosed eating disorders; and 10) acutely unwell at recruitment visit.

This research was approved by the Human Research Ethics Committees of Mater Misericordiae Limited (HREC_15_MHS_35T1DM) and the University of Queensland (UQ 2015–000–958). Written informed consent was obtained from all participants and, if under 18 years of age, from their legal guardian in addition to the participant assent, prior to inclusion in the study. All investigation was conducted according to the principles of the Declaration of Helsinki.

Data Collection and Biochemistry

Data obtained at recruitment included chronological age, sex, height, weight, medications, year of diabetes diagnosis, and resting seated mean systolic blood pressure. Historical HbA1c, nonfasted plasma glucose, and uACR were collated from patient histories back to diabetes diagnosis if available. The mean observation periods for HbA1c and plasma glucose were: low risk, 7.6 ± 2.7 years and high risk, 7.0 ± 3.4 years. For uACR, the mean observation period for the low-risk group was 5.0 ± 1.7 years and for the high-risk group 4.8 ± 2.5 years. HbA1c, fructosamine albumin, serum creatinine, uACR, and total cholesterol were assessed in a centralized Mater Pathology laboratory. Urine albumin (range 2–4,400 mg/L) and creatinine were measured by a Cobas 6000 Analyzer (Roche/Hitachi, Indianapolis, IN) by turbidimetry or crep-2 (range 5–1,290 mg/dL), respectively. Quantikine ELISA kits (R&D Systems, Minneapolis, MN) were used for plasma cystatin C, KIM-1, tumor necrosis factor receptor-2, and fibroblast growth factor-21 according to the manufacturer’s instructions.

Calculations

Estimated glomerular filtration rate (eGFR; in milliliters per minute per 1.73 m2) was calculated from plasma creatinine using the Chronic Kidney Disease Epidemiology Collaboration eGFR (eGFRCKD-EPI) equation, eGFR = 133 × min (SCr/0.8, 1)−0.49 × max (SCr/0.8, 1)−1.328 × 0.996Age × 0.932 (if female), and by the modified Schwartz equation, eGFRSCHWARTZ = 42 × height (centimeters)/SCr (micromoles per liter), for all participants irrespective of age given their previous discordance in young adults (30). Hyperfiltration was defined if the average of eGFRCKD-EPI and eGFRSCHWARTZ was >95th percentile for a healthy age-matched population (31). In all individuals, the second morning urine void was collected and averaged with the two previous consecutive clinic visits to calculate mean uACR values for DKD risk. Albuminuria was defined according to guidelines from the Australasian Proteinuria Consensus Working Group and the International Society of Pediatric and Adolescent Diabetes as uACR >2.5 mg/mmol in males and >3.5 mg/mmol in females (28,32).

Proximal Tubule Epithelial Cells and Patient Plasma Experiments

Primary human renal proximal tubule epithelial cells (PCS400010; ATCC, Manassas, VA) at passages 3–5 in triplicate were exposed to 4% patient plasma added into RPMI/F12 media as previously described (33), with a final d-glucose concentration of 5 mmol/L and FCS concentration of 0.5% in the presence and absence of anti–KIM-1 antibody pretreatment (5 μg/mL; R&D Systems). Sodium–glucose cotransporter 2 (SGLT2) (5 μg/mL; ab37296, Abcam, Cambridge, MA) and collagen IV (10 μg/mL; AB769, Merck Millipore, Darmstadt, Germany) were quantified using flow cytometry or IN Carta Image Analysis Software (General Electric, Boston, MA), respectively.

Human Peripheral Blood Mononuclear Cell Flow Cytometry

Peripheral blood mononuclear cells (PBMC) were incubated with anti-CD3/CD28 MACSiBeads (4:1) and then stained with antibodies against KIM-1 (219211; R&D Systems), CD3 (OKT3; BioLegend, San Diego, CA), CD4 (RPA-T4; BD Horizon; BD Biosciences, Piscataway, NJ), CD8 (SK1; BioLegend), CD25 (2A3; BD Horizon), CD127 (HIL-7R-M21; BD Horizon), CD45RA (H100; BD Horizon), HLA-DR (L243; BioLegend), and FOXP3 (POCH101, RPA-T4, and BC96; eBioscience, Waltham, MA; fixed panel only). Dead cells were excluded using a Fixable Viability Stain (BD Horizon). Cells were characterized using BD LSR Fortessa X-20 (SORP; BD Biosciences) and analyzed using FlowJo (>99% regulatory T [Treg] cell and conventional T [Tconv] cell purity). Treg cells were visualized using an Amnis ImageStreamX (Luminex Corporation, Austin, TX).

Human Kidney Tissue Immunofluorescence

Tissue sections were obtained from the healthy portion of a tumor nephrectomy (N = 4) or from patients with minimal change disease (N = 4), primary focal segmental glomerulosclerosis (FSGS; N = 4), or DKD (N = 4), following informed patient consent and approval by the Royal Brisbane and Women’s Hospital Human Research Ethics Committee (2002/011 and 2006/072). DKD was confirmed by a renal histopathologist by the presence of classical lesions (i.e., Kimmelstiel-Wilson nodules and mesangial matrix expansion). Pathological features of other renal diseases were not identified in renal tissue from donors with DKD. Frozen 7-μm human tissue sections were fixed with 25% ethanol/75% acetone at room temperature for 5 min, followed by a protein block with 10% donkey serum for 30 min. Sections were subsequently probed with primary antibodies against KIM-1 (goat polyclonal IgG; R&D Systems), aquaporin-1 (monoclonal mouse IgG1; Santa Cruz Biotechnology, Dallas, TX), and CD3 (rabbit polyclonal IgG; Agilent Technologies, Santa Clara, CA) at room temperature for 2 h. Fluorescent detection was obtained by secondary incubation with Alexa Fluor Plus 488 anti-goat IgG, Alexa Fluor Plus 555 anti-rabbit IgG, and Alexa Fluor Plus 647 anti-mouse IgG (all from Life Technologies, Grand Island, NY) at room temperature for 30 min. Nuclei were stained with DAPI (Sigma-Aldrich, St. Louis, MO). Slides were coverslipped in fluorescence mounting medium (Agilent Technologies), visualized using a Zeiss 780 NLO confocal microscope (Carl Zeiss, Hamburg, Germany), and quantified using ImageJ (v2.0.0; National Institutes of Health, Bethesda, MD) and QuPath (v0.2.0; University of Edinburgh, Edinburgh, U.K.).

Preclinical Model of Glycemic Variability

Transient intermittent hyperglycemia in the absence of diabetes was induced by four intraperitoneal d-glucose bolus doses (2 g/kg) at 2-h intervals with sham control mice administered 0.9% saline as described previously (34). This regimen was repeated weekly from adolescence at week 6 of age for 10 weeks in male apolipoprotein E–deficient (ApoE−/−; C57BL/6J background) mice prone to DKD and atherosclerosis. Immunofluorescence was performed using rabbit anti–Kim-1 antibody (10 μg/mL; ab47635; Abcam) at 4°C overnight and then secondary antibody donkey anti-rabbit IgG Highly Cross-Absorbed (0.5 μg/mL; AF568-A10042; Thermo Fisher Scientific) for 1 h at room temperature on fixed mouse tissue sections post–citrate buffer, pH 6.0, antigen retrieval. Sections were then washed, counterstained with DAPI (2 μg/mL), and visualized using an Olympus FV3000 confocal microscope. At the study end, circulating CD4+ and CD8+ T lymphocyte surface Kim-1 expression was assessed by a flow cytometry panel consisting of Ghost Dye 510 Viability dye (13–0870-T500; Tonbo Biosciences), Kim-1 (CD365; RMTI-4; BioLegend), CD3 (17A2; BioLegend), CD4 (RM4–5; BD Biosciences), and CD8b (Ly-3; YTS156.7.7; BioLegend). uACR was calculated following measurement using albumin ELISA (R&D Systems) and creatinine enzyme assay (Crystal Chem). Serum cystatin C was assayed by ELISA (R&D Systems). Renal cortical gene expression of Kim-1 (Havcr1), collagen IV (Col4a1), and SGLT2 (Slc5a2) was measured using quantitative PCR.

Preclinical Model of Diabetes and Early Kidney Disease

Diabetes was induced in male ApoE−/− (C57BL/6J background) mice at 5 weeks of age using multiple low-dose intraperitoneal (i.p.) injections of the β-cell toxin streptozotocin (55 mg/kg) (35). All mice (N = 10) with plasma glucose concentrations of >15 mmol/L 1 week later at 6 weeks of age were included in the study. Mice were followed concurrently for 10 weeks from week 6 to 16 of life, with those given chronic glucose injections as described above until week 16 of life.

Statistical Analyses

Data are shown as mean ± SD or median (interquartile range) unless otherwise stated. Normality testing (Shapiro-Wilk) was performed on all data prior to analysis. Parametric data were analyzed by one-way ANOVA with Tukey post hoc for multiple comparisons or Kruskal-Wallis with Dunn post hoc testing for low- and high-risk groups compared using Student t test or Mann-Whitney U. Univariate modeling with Holm correction was used to determine interdependence of variables for inclusion in eGFR linear models and to generate Pearson coefficients for mouse uACR/T-cell KIM-1 associations. General linear models in R were used to examine the relationships between uACR and eGFR surrogates or plasma KIM-1 with correction for covariates age, diabetes duration, sex, and HbA1c. All statistical analyses were performed using GraphPad Prism (v8.2.1; GraphPad Software, San Diego, CA). P values ≤0.05 were considered statistically significant.

Data and Resource Availability

The data set from Heidkamp’s group analyzed during the current study is available in the Stemformatics repository (https://www.stemformatics.org). The data sets and resources generated and analyzed during this study are available from the corresponding author upon reasonable request.

Youth With Type 1 Diabetes and High-Risk uACR Have Evidence of Early Kidney Disease

The uACR was examined in a cohort of 100 youth with type 1 diabetes, with a mean age of 20.0 ± 2.8 years of age, a ratio of male to female patients of 54:46, HbA1c of 66.1 ± 12.3 mmol/mol (8.2 ± 0.7%), BMI of 24.5 (5.3) kg/m2, and a diabetes duration of 10.7 ± 5.2 years (Table 1). Those with previously diagnosed kidney disease were excluded from this cohort. The study population was plotted in order of increasing uACR (Fig. 1A) and divided by uACR tertiles (Table 1) aligning with low (uACR ≤0.66 mg/mmol; n = 33), medium (uACR 0.67–1.16; n = 33), or high risk for DKD (uACR ≥1.17; n = 34). The risk calculations were performed on uACRs collected over a mean of 3.6 ± 1.0 years for the low-risk group and 3.9 ± 0.9 years for the high-risk group with ∼90% of participants having three uACR measurements taken during that time. Despite those with previously diagnosed kidney disease being excluded from our study, 11% (∼13% of male and 8.7% of female patients) of young people with diabetes in our cohort had early clinical evidence of kidney disease when their past three uACR results were combined (microalbuminuria in Table 1). This small subgroup with microalbuminuria also had higher systolic blood pressure than other high-risk subjects without albuminuria, but not increased lipids (Table 1).

Kidney function using eGFR showed that many individuals with diabetes in our human cohort had significantly increased glomerular filtration (hyperfiltration). This was most pronounced in the high-risk group in which 52.9% of youth had hyperfiltration compared with 30.3% of youth in both the low- and medium-risk groups (Table 1 and Supplementary Fig. 1AC). The relationship between DKD risk factors log uACR and eGFR corrected for age, sex, diabetes duration, and HbA1c,was examined using both adult eGFRCKD-EPI (Fig. 1B) and pediatric eGFRSCHWARTZ (Fig. 1C) equations and the eGFR surrogate serum cystatin C (Supplementary Fig. 1C). eGFR consistently showed a strong positive relationship with log uACR in individuals at low and medium risk for DKD with adjustment for age, diabetes duration, sex and HbA1c, but this was lost in high-risk individuals, evident using either the adult (eGFRCKD-EPI; low, r = 0.53, Padj = 0.002; high, r = −0.09, Padj = 0.63) (Fig. 1B) or pediatric eGFR (eGFRSCHWARTZ; low, r = 0.58, Padj = 0.0007; high, r = −0.28, Padj = 0.12) (Fig. 1C) formulas. In high-risk individuals, lower eGFR was seen in the context of greater log uACR, reminiscent of changes seen in early CKD (Fig. 1B and C and Supplementary Table 1). This was also seen when the relationship between eGFR by the surrogate cystatin C and log uACR was examined (Supplementary Fig. 1D).

Figure 1

Youth with type 1 diabetes at high risk for DKD have early kidney dysfunction and greater glycemic variability. Youth (20.0 ± 2.8 years old) with type 1 diabetes but not previously diagnosed with DKD were stratified by future risk for DKD using tertiles of uACR (low risk, N = 33; medium [Med] risk, N = 33; and high risk, N = 34). AD: Measures of kidney function. A: All subjects in the cohort (N = 100) plotted by increasing uACR. General linear regression plot of log uACR vs. eGFR using the adult eGFRCKD-EPI formula (B) or the pediatric modified eGFRSCHWARTZ equation (C), corrected for age, sex, diabetes duration and HbA1c. D: Historical uACR. Mean uACR observation periods were 5.0 ± 1.7 years and 4.8 ± 2.5 years for the low- versus high-risk group, respectively. Historical long-term glucose control (HbA1c) (E) and nonfasted plasma glucose concentrations (F) from the time of diabetes diagnosis (green arrows) according to DKD risk. Mean HbA1c/plasma glucose observation periods were 7.6 ± 2.7 years and 7.0 ± 3.4 years for the low- vs. high-risk group, respectively. G: AUC for retrospective uACR over the previous 10 years (AUCuACR 0-10) (other AUC plots in Supplementary Fig. 1). HJ: Cohort glycemic control at the time of recruitment. Long-term markers of glucose control, HbA1c (H) and fructosamine albumin (Alb) (I), and blood glucose variability in 1,5 anhydroglucitol (AG) (J). **P < 0.01 vs. low risk or medium risk. No., number.

Figure 1

Youth with type 1 diabetes at high risk for DKD have early kidney dysfunction and greater glycemic variability. Youth (20.0 ± 2.8 years old) with type 1 diabetes but not previously diagnosed with DKD were stratified by future risk for DKD using tertiles of uACR (low risk, N = 33; medium [Med] risk, N = 33; and high risk, N = 34). AD: Measures of kidney function. A: All subjects in the cohort (N = 100) plotted by increasing uACR. General linear regression plot of log uACR vs. eGFR using the adult eGFRCKD-EPI formula (B) or the pediatric modified eGFRSCHWARTZ equation (C), corrected for age, sex, diabetes duration and HbA1c. D: Historical uACR. Mean uACR observation periods were 5.0 ± 1.7 years and 4.8 ± 2.5 years for the low- versus high-risk group, respectively. Historical long-term glucose control (HbA1c) (E) and nonfasted plasma glucose concentrations (F) from the time of diabetes diagnosis (green arrows) according to DKD risk. Mean HbA1c/plasma glucose observation periods were 7.6 ± 2.7 years and 7.0 ± 3.4 years for the low- vs. high-risk group, respectively. G: AUC for retrospective uACR over the previous 10 years (AUCuACR 0-10) (other AUC plots in Supplementary Fig. 1). HJ: Cohort glycemic control at the time of recruitment. Long-term markers of glucose control, HbA1c (H) and fructosamine albumin (Alb) (I), and blood glucose variability in 1,5 anhydroglucitol (AG) (J). **P < 0.01 vs. low risk or medium risk. No., number.

Close modal
Table 1

Characteristics of youth stratified for DKD risk using tertiles of uACR

ParametersType 1 diabetes, low risk (<0.66 mg/mmol)Type 1 diabetes, medium risk (0.67–1.16 mg/mmol)*Type 1 diabetes, high risk (>1.16 mg/mmol)*#
No microalbuminuria (1.6 ± 0.3 mg/mmol*)Microalbuminuria (7.6 ± 4.7 mg/mmol*#)Combined (3.5 ± 3.8 mg/mmol*#)
Age (years) 20.1 ± 2.7 20.0 ± 2.8 19.7 ± 2.7 20.6 ± 3.9 20.0 ± 3.1 
Sex, N (% female) 33 (36) 33 (51) 23 (57) 11 (36) 34 (50) 
BMI (kg/m226.2 ± 4.1 24.7 ± 3.5 25.7 ± 6.5 26.2 ± 7.5 25.7 ± 6.6 
Random BG (mmol/L) 11.4 ± 4.5 11.6 ± 3.9 11.8 ± 5.4 12.6 ± 3.4 12.3 ± 4.7 
Diabetes duration (years) 9.9 ± 4.9 11.4 ± 4.9 10.3 ± 6.5 12.0 ± 3.9 10.7 ± 5.8 
HbA1c (%, mmol/mol) 8.0 ± 0.7, 64.0 ± 7.9 8.1 ± 0.8, 64.8 ± 8.3 8.4 ± 0.8, 68.2 ± 8.6* 8.4 ± 1.3, 68.8 ± 13.9* 8.4 ± 0.9, 68.7 ± 10.3* 
SBP (mmHg) 116.5 ± 15.4 115.6 ± 8.8 113.9 ± 12.3 127.0 ± 18.6 117.3 ± 16.1 
Total cholesterol (mmol/L) 4.4 ± 0.9 4.7 ± 1.0 4.6 ± 0.7 4.5 ± 1.1 4.6 ± 0.8 
eGFRCKD-EPI (mL/min/1.73 m2134.7 ± 9.5 136.3 ± 8.1 138.4 ± 10.9 134.7 ± 11.1 137.8 ± 11.4 
eGFRSCHWARTZ (mL/min/1.73 m2110.5 ± 16.1 116.5 ± 14.8 120.5 ± 20.0 112.0 ± 15.9 120.1 ± 22.6 
ParametersType 1 diabetes, low risk (<0.66 mg/mmol)Type 1 diabetes, medium risk (0.67–1.16 mg/mmol)*Type 1 diabetes, high risk (>1.16 mg/mmol)*#
No microalbuminuria (1.6 ± 0.3 mg/mmol*)Microalbuminuria (7.6 ± 4.7 mg/mmol*#)Combined (3.5 ± 3.8 mg/mmol*#)
Age (years) 20.1 ± 2.7 20.0 ± 2.8 19.7 ± 2.7 20.6 ± 3.9 20.0 ± 3.1 
Sex, N (% female) 33 (36) 33 (51) 23 (57) 11 (36) 34 (50) 
BMI (kg/m226.2 ± 4.1 24.7 ± 3.5 25.7 ± 6.5 26.2 ± 7.5 25.7 ± 6.6 
Random BG (mmol/L) 11.4 ± 4.5 11.6 ± 3.9 11.8 ± 5.4 12.6 ± 3.4 12.3 ± 4.7 
Diabetes duration (years) 9.9 ± 4.9 11.4 ± 4.9 10.3 ± 6.5 12.0 ± 3.9 10.7 ± 5.8 
HbA1c (%, mmol/mol) 8.0 ± 0.7, 64.0 ± 7.9 8.1 ± 0.8, 64.8 ± 8.3 8.4 ± 0.8, 68.2 ± 8.6* 8.4 ± 1.3, 68.8 ± 13.9* 8.4 ± 0.9, 68.7 ± 10.3* 
SBP (mmHg) 116.5 ± 15.4 115.6 ± 8.8 113.9 ± 12.3 127.0 ± 18.6 117.3 ± 16.1 
Total cholesterol (mmol/L) 4.4 ± 0.9 4.7 ± 1.0 4.6 ± 0.7 4.5 ± 1.1 4.6 ± 0.8 
eGFRCKD-EPI (mL/min/1.73 m2134.7 ± 9.5 136.3 ± 8.1 138.4 ± 10.9 134.7 ± 11.1 137.8 ± 11.4 
eGFRSCHWARTZ (mL/min/1.73 m2110.5 ± 16.1 116.5 ± 14.8 120.5 ± 20.0 112.0 ± 15.9 120.1 ± 22.6 

Low risk, N = 33; medium risk, N = 33; and high risk, N = 34. The high-risk tertile is also divided for the presence or absence of microalbuminuria. Data are mean ± SD and analyzed by Kruskal-Wallis with Dunn post hoc testing. BG, blood glucose.

*

P < 0.05 vs. low-risk tertile.

#

P < 0.05 vs. medium-risk tertile, Mann-Whitney U test.

P < 0.05 vs. high-risk tertile with no microalbuminuria.

Risk for DKD Is Evident Early After Type 1 Diabetes Diagnosis

Diabetes duration did not differ between risk groups (Table 1). uACR was elevated at or soon after diabetes diagnosis in those individuals at high compared with low risk for DKD (Fig. 1D) (area under the curve [AUC], low vs. high risk, P < 0.01) (Fig. 1G). Historically, both the long-term marker of glucose control HbA1c and point of care random blood glucose concentrations did not differ among risk groups for DKD (Fig. 1E and F and as AUC in Supplementary Fig. 1E and F). However, at recruitment (∼10 years after diabetes diagnosis), poorer glucose control as demonstrated by higher HbA1c (Fig. 1H) and fructosamine albumin (Fig. 1I) and greater glucose variability using 1,5 anhydroglucitol (36) (Fig. 1J) were evident in high- compared with low-risk subjects for DKD. If 1,5 anhydroglucitol is <2, as seen in the high-risk group (Fig. 1J), this translates to mean average daily glucose excursions >16.1 mmol/L (290 mg/dL).

Circulating KIM-1 Is Elevated in Youth at Greater DKD Risk

Youth at high risk of DKD had elevated plasma KIM-1 concentrations (Fig. 2A). Other biomarkers of early kidney injury did not differ among DKD risk groups, including plasma tumor necrosis factor receptor-2, fibroblast growth factor-21, and neutrophil gelatinase-associated lipocalin (Supplementary Fig. 2AC). Plasma KIM-1 concentrations also increased across risk groups (Fig. 2A) (Ptrend = 0.0382) and were positively associated with log uACR (Fig. 2B) in both medium- (r = 0.11; Padj = 0.002) and high-risk individuals (r = 0.28; Padj = 0.0003).

When healthy human kidney cells (PTECs) were exposed to 4% plasma from high-risk youth, they showed damage akin to DKD, including greater production of the fibrosis marker collagen IV (Fig. 2C and E-H) and cell surface expression of SGLT2 (Fig. 2D and E–H and Supplementary Fig. 2D and E), but viability was unaffected (Supplementary Fig. 2F and G). These DKD-like changes were not seen when PTECs were exposed to plasma from low-risk individuals pairwise matched for age, sex, diabetes duration, BMI, random blood glucose, and HbA1c (Supplementary Fig. 2I). Further, preincubation with anti–KIM-1–neutralizing antibodies alleviated the increases seen in collagen IV (Fig. 2C and E–H) and SGLT2 (Fig. 2D and E–H and Supplementary Fig. 2D and E) following exposure to high-risk plasma, suggesting that plasma KIM-1 was responsible. Urinary KIM-1 was not different between DKD risk groups (Fig. 2J).

Figure 2

Circulating KIM-1 increases with DKD risk and damages healthy primary human kidney cells. Youth (20.0 ± 2.8 years old) with type 1 diabetes were stratified by risk for DKD using tertiles of uACR (low risk, N = 33; medium [Med] risk, N = 33; and high risk, N = 34). A: Plasma KIM-1 measured by ELISA. B: Linear regression plot of log uACR vs. plasma KIM-1 in the entire cohort. C and D: Healthy PTECs were incubated for 24 h with plasma (4%) from matched pairs of participants (N = 10, low/high-risk pairs matched for age, sex, diabetes duration, HbA1c, and BMI) in the presence and absence of preincubation with KIM-1–neutralizing antibody (Ab). C: Collagen IV PTEC content by IN Carta Image Analysis Software. D: Upregulation of cell surface SGLT2 on PTECs by flow cytometry. EH: Representative photomicrographs of patient plasma-exposed PTECs stained for collagen IV (Coll IV; red), SGLT2 (green), and nuclei (DAPI; blue). E: Low risk plus control Ab. F: Low risk plus KIM-1–blocking Ab. G: High risk plus control Ab. H: High risk plus KIM-1– blocking Ab. Scale bar = 20 μm. I: Characteristics of paired low- and high-risk participants whose 4% plasma was used in PTEC experiments (N = 10/group). J: Urinary KIM-1 concentrations in all subjects per milligram of creatinine (Cr). *P < 0.05 vs. low risk; **P < 0.01 vs. low risk plus KIM-1 blockade. BG, blood glucose; MFU, mean fluorescence unit.

Figure 2

Circulating KIM-1 increases with DKD risk and damages healthy primary human kidney cells. Youth (20.0 ± 2.8 years old) with type 1 diabetes were stratified by risk for DKD using tertiles of uACR (low risk, N = 33; medium [Med] risk, N = 33; and high risk, N = 34). A: Plasma KIM-1 measured by ELISA. B: Linear regression plot of log uACR vs. plasma KIM-1 in the entire cohort. C and D: Healthy PTECs were incubated for 24 h with plasma (4%) from matched pairs of participants (N = 10, low/high-risk pairs matched for age, sex, diabetes duration, HbA1c, and BMI) in the presence and absence of preincubation with KIM-1–neutralizing antibody (Ab). C: Collagen IV PTEC content by IN Carta Image Analysis Software. D: Upregulation of cell surface SGLT2 on PTECs by flow cytometry. EH: Representative photomicrographs of patient plasma-exposed PTECs stained for collagen IV (Coll IV; red), SGLT2 (green), and nuclei (DAPI; blue). E: Low risk plus control Ab. F: Low risk plus KIM-1–blocking Ab. G: High risk plus control Ab. H: High risk plus KIM-1– blocking Ab. Scale bar = 20 μm. I: Characteristics of paired low- and high-risk participants whose 4% plasma was used in PTEC experiments (N = 10/group). J: Urinary KIM-1 concentrations in all subjects per milligram of creatinine (Cr). *P < 0.05 vs. low risk; **P < 0.01 vs. low risk plus KIM-1 blockade. BG, blood glucose; MFU, mean fluorescence unit.

Close modal
Figure 3

Glucose variability even in the absence of diabetes increases T-cell Kim-1 expression in preclinical models. A: Male adolescent ApoE−/− mice (6 weeks [Wks] of age) received four i.p. injections of glucose (2 g/kg; blue diamonds; N = 10) or isovolumetric saline injections (saline; white diamonds; N = 10), delivered 2 h apart, to achieve plasma glucose variations that peaked between ∼15 and 20 mmol/L. B: Plasma glucose concentrations after four bihourly glucose injections are shown. This was repeated weekly for 10 weeks and compared with age-matched streptozotocin diabetic mice followed over the same period (red diamonds; N = 10). B: Daily plasma glucose concentrations in saline- and glucose-injected mice. C: Long-term glucose control measured by glycated hemoglobin. D: uACR at week 16. E and F: Flow cytometry analysis of live peripheral blood Kim-1+ cells. E: Non-T cells (Kim-1+CD3). F: CD4+ (Kim-1+CD3+CD4+CD8) and CD8+ (Kim-1+CD3+CD8+CD4) subsets as proportion of CD3+ T cells. G: Linear regression of CD3+CD8+CD4Kim-1+ T cells and uACR. H: Contour plot of CD4+ and CD8+ separation of murine live (Ghost 510-nm viability) CD3+ Kim-1+ T cells from peripheral blood. Data are shown as mean SD or median (interquartile range) and tested using one-way ANOVA/Tukey post hoc or Kruskal Wallis/Dunn post hoc testing. *P < 0.05, **P < 0.01, ***P < 0.001 vs. saline group. Diab, diabetes.

Figure 3

Glucose variability even in the absence of diabetes increases T-cell Kim-1 expression in preclinical models. A: Male adolescent ApoE−/− mice (6 weeks [Wks] of age) received four i.p. injections of glucose (2 g/kg; blue diamonds; N = 10) or isovolumetric saline injections (saline; white diamonds; N = 10), delivered 2 h apart, to achieve plasma glucose variations that peaked between ∼15 and 20 mmol/L. B: Plasma glucose concentrations after four bihourly glucose injections are shown. This was repeated weekly for 10 weeks and compared with age-matched streptozotocin diabetic mice followed over the same period (red diamonds; N = 10). B: Daily plasma glucose concentrations in saline- and glucose-injected mice. C: Long-term glucose control measured by glycated hemoglobin. D: uACR at week 16. E and F: Flow cytometry analysis of live peripheral blood Kim-1+ cells. E: Non-T cells (Kim-1+CD3). F: CD4+ (Kim-1+CD3+CD4+CD8) and CD8+ (Kim-1+CD3+CD8+CD4) subsets as proportion of CD3+ T cells. G: Linear regression of CD3+CD8+CD4Kim-1+ T cells and uACR. H: Contour plot of CD4+ and CD8+ separation of murine live (Ghost 510-nm viability) CD3+ Kim-1+ T cells from peripheral blood. Data are shown as mean SD or median (interquartile range) and tested using one-way ANOVA/Tukey post hoc or Kruskal Wallis/Dunn post hoc testing. *P < 0.05, **P < 0.01, ***P < 0.001 vs. saline group. Diab, diabetes.

Close modal

Variations in Plasma Glucose Increase uACR Even in Absence of Diabetes

Glucose variability in the absence of diabetes was compared with type 1 diabetes (Fig. 3A) to examine if this was sufficient to elevate DKD risk measured as uACR and Kim-1. Adolescent ApoE−/− mice (6 weeks of age) who were susceptible to kidney and CVD received four i.p. injections of glucose (2 g/kg) or isovolumetric saline, delivered 2 h apart (Fig. 3A and B), achieving plasma glucose variations peaking between ∼15 and 20 mmol/L (Fig. 3B). No changes in body or kidney weight, random blood glucose (Supplementary Fig. 3), or HbA1c concentrations (Fig. 3C) were seen in saline- or glucose-injected groups, but as expected, HbA1c was elevated in the diabetes group by week 16 of life (Fig. 3C). uACR, which defined DKD risk in our human cohort, was also elevated by glucose variations in the absence of diabetes by week 16 (Fig. 3D). As expected, diabetes induced hyperfiltration, seen by decreases in the GFR surrogate plasma cystatin C (Supplementary Fig. 3) and structural injury seen as increased kidney collagen IV and Kim-1 expression compared with saline control mice (Supplementary Fig. 3).

Variations in Plasma Glucose Increase T-Cell KIM-1 Expression Independent of Diabetes

Kidney Kim-1 protein expression in the group given chronic glucose injections was not increased (Supplementary Fig. 3) but was increased on the cell surface of circulating T cells (Fig. 3E and F and gating strategy in Supplementary Fig. 4). Kim-1 was particularly increased in CD8+ T cells (Fig. 3F and H), and this was strongly associated with uACR in the glucose-injected group (Fig. 3G). There was no relationship between CD4+ T-cell Kim-1 expression and uACR in any group (Supplementary Fig. 3).

T Cells Are a Source of Circulating KIM-1 in Youth at High Risk for DKD

Using the data set from Heidkamp et al. (37) acquired from healthy human donors, we showed that peripheral blood cell expression of the KIM-1 gene HAVCR1 is highest in CD8+ T cells but is also present in CD4+ T cells and various dendritic cell subsets (Supplementary Fig. 5).

In our young cohort with diabetes, we found no differences in KIM-1 expression on circulating mononuclear cells (PBMC) that were CD3 (non-T cells) between individuals with different risk for DKD (gating strategy in Supplementary Figs. 6 and 7AC). However, when just T cells were studied, greater cell surface KIM-1 expression in individuals at high risk for DKD compared with low risk was evident (CD3+) (Supplementary Fig. 7B). T-cell KIM-1 was greatest on CD8+ T cells when compared with CD4+ T cells in youth with type 1 diabetes (Fig. 4A and B). With further characterization of Tconv subsets based on CD45RA and HLA-DR cell surface expression, youth at high risk for DKD had greater cell surface KIM-1 expression on naive Tconv cell subsets (CD45RA+HLA-DR) in both CD8+ (Fig. 4C) and CD4+ (Supplementary Fig. 7G) compartments. KIM-1 expression was also increased in CD8+ Tconv double-positive (CD45RA+HLA-DR+) cells, but not any other CD8+ (Supplementary Figure 7DF) or CD4+ Tconv subsets (Supplementary Figure 7GI).

Figure 4

T-cell KIM-1 expression is increased in youth at high risk for DKD. Youth (20.0 ± 2.8 years old) with type 1 diabetes were stratified by risk for DKD using tertiles of uACR (low risk, N = 33; medium risk, N = 33; and high risk, N = 34). KIM-1 expression on live PBMCs is shown for low- and high-risk groups. Control subjects are a reference group of healthy individuals but are not age- and sex-matched nor used in statistical analyses. Circulating KIM-1+ live T-cell (CD3+) populations (A) as a contour plot for all KIM-1+CD4 (green) and CD8+ (blue) and plotted as individual subjects (B). C: Circulating KIM-1+ Tconv (CD3+) cells expressing KIM-1 sorted for the specific subset of naive CD8+ Tconv cells (CD3+CD8+CD4CD45RA+HLA-DR). DG: Circulating Treg cell (CD3+CD25+CD127lo/−) populations expressing KIM-1 plotted as a proportion of all CD4+ (green) and CD8+ (blue) Treg (D) or all CD3+KIM-1+ Treg (CD3+CD8+CD25+CD127lo/−) (E). F: CD8+ Treg cells expressing KIM-1 sorted for the naive CD8+ Treg (CD8+CD25+CD127lo/−CD45RA+HLA-DR) subset. G: Single-cell imaging of two representative naive KIM-1+CD8+ Treg cells during flow cytometry showing channels for cell surface markers bright field (BF), CD8 (red), CD25 (green), CD45RA (violet), KIM-1 (blue), and intracellular FOXP3 (yellow). All other KIM-1+ T-cell subsets are presented in Supplementary Figs. 7 and 8. Data are shown as mean ± SD or median (interquartile range) and analyzed using Student t test or Mann-Whitney U test. C, control.

Figure 4

T-cell KIM-1 expression is increased in youth at high risk for DKD. Youth (20.0 ± 2.8 years old) with type 1 diabetes were stratified by risk for DKD using tertiles of uACR (low risk, N = 33; medium risk, N = 33; and high risk, N = 34). KIM-1 expression on live PBMCs is shown for low- and high-risk groups. Control subjects are a reference group of healthy individuals but are not age- and sex-matched nor used in statistical analyses. Circulating KIM-1+ live T-cell (CD3+) populations (A) as a contour plot for all KIM-1+CD4 (green) and CD8+ (blue) and plotted as individual subjects (B). C: Circulating KIM-1+ Tconv (CD3+) cells expressing KIM-1 sorted for the specific subset of naive CD8+ Tconv cells (CD3+CD8+CD4CD45RA+HLA-DR). DG: Circulating Treg cell (CD3+CD25+CD127lo/−) populations expressing KIM-1 plotted as a proportion of all CD4+ (green) and CD8+ (blue) Treg (D) or all CD3+KIM-1+ Treg (CD3+CD8+CD25+CD127lo/−) (E). F: CD8+ Treg cells expressing KIM-1 sorted for the naive CD8+ Treg (CD8+CD25+CD127lo/−CD45RA+HLA-DR) subset. G: Single-cell imaging of two representative naive KIM-1+CD8+ Treg cells during flow cytometry showing channels for cell surface markers bright field (BF), CD8 (red), CD25 (green), CD45RA (violet), KIM-1 (blue), and intracellular FOXP3 (yellow). All other KIM-1+ T-cell subsets are presented in Supplementary Figs. 7 and 8. Data are shown as mean ± SD or median (interquartile range) and analyzed using Student t test or Mann-Whitney U test. C, control.

Close modal
Figure 5

Kidney biopsies from individuals with DKD show infiltration of KIM-1+ T cells. Representative immunofluorescent staining of kidney tissue in frozen sections taken from research tissue donors with renal pathologist–confirmed healthy kidney (A), minimal change disease (MCD) (B), FSGS (C), and DKD (D). Kidney tissue sections were stained for Aquaporin-1 (white), T cells (CD3; red), KIM-1 (green), and nuclei (DAPI; blue). A representative KIM-1+ T cell is circled in D. Scale bars = 100 μm; 10 μm for the inset image in D. E: Morphometric quantification of CD3+ T cells (cells per mm2) from kidney tissues (N = 4 donors/group) with values for individual donors presented. Data are shown as median with interquartile range. F: Proportion of CD3+ T cells expressing KIM-1 from healthy kidney tissue (N = 4), MCD (n = 4), FSGS (n = 4), and DKD tissue (n = 4).

Figure 5

Kidney biopsies from individuals with DKD show infiltration of KIM-1+ T cells. Representative immunofluorescent staining of kidney tissue in frozen sections taken from research tissue donors with renal pathologist–confirmed healthy kidney (A), minimal change disease (MCD) (B), FSGS (C), and DKD (D). Kidney tissue sections were stained for Aquaporin-1 (white), T cells (CD3; red), KIM-1 (green), and nuclei (DAPI; blue). A representative KIM-1+ T cell is circled in D. Scale bars = 100 μm; 10 μm for the inset image in D. E: Morphometric quantification of CD3+ T cells (cells per mm2) from kidney tissues (N = 4 donors/group) with values for individual donors presented. Data are shown as median with interquartile range. F: Proportion of CD3+ T cells expressing KIM-1 from healthy kidney tissue (N = 4), MCD (n = 4), FSGS (n = 4), and DKD tissue (n = 4).

Close modal

KIM-1 was more highly expressed by Treg compared with Tconv cells, predominantly in the CD8+ Treg cell population (Fig. 4D). KIM-1 was further increased in both CD8+ and CD4+ Treg cells in youth at greater risk for DKD, in whom increased frequencies of KIM-1+CD8+ Treg cells were present in high-risk youth as compared with those at low risk for DKD (Fig. 4D and E). Treg cells (either CD4+ or CD8+CD25+CD127lo/−) each had >90% FOXP3 expression (Supplementary Fig. 8AD). As for Tconv cells, KIM-1 was predominantly expressed by naive Treg cells in both the CD8+ (Fig. 4F) and CD4+ (Supplementary Fig. 8H) compartments and was significantly elevated in youth with high DKD risk. Coexpression of cell surface KIM-1 and intracellular FOXP3 by naive CD8+ Treg cells was confirmed by single-cell imaging (Fig. 4G). CD8+ double-positive (CD45RA+HLA-DR+) Treg cells also had higher KIM-1 expression in youth at high risk for DKD as compared with low risk (Supplementary Fig. 8G). Other circulating Treg subsets did not differ in KIM-1 expression according to DKD risk (Supplementary Fig. 8EJ). For all T-cell subsets examined, there were no differences in frequencies between high- and low-risk subjects (Supplementary Fig. 9AX).

KIM-1+ T Cells Infiltrate Kidney Tissue in Established DKD

Kidney tissue taken from individuals with impairment in kidney function (Supplementary Table 1) (i.e., those with either primary FSGS or DKD) had increased proximal tubule KIM-1 when compared with healthy kidney or kidney tissue taken from subjects with minimal change nephropathy (Fig. 5A–D). Kidney tissue from individuals with FSGS or DKD had greater T-cell infiltration (Fig. 5E), consistent with previous reports (38,39). However, the proportion of T cells expressing KIM-1 was significantly greater in those tissue donors with established DKD when compared with all other groups (Fig. 5F), including those with FSGS who had a similar degree of functional impairment (Supplementary Table 1).

Historically, strict glucose control shows a legacy effect to decrease the incidence of chronic diabetes complications (40,41), but this has been challenged by more recent large-scale clinical trials (42,43). Further, youth with diabetes often cannot achieve the strict clinical targets required to reduce their risk for DKD and CVD (44) due to factors including puberty, insulin resistance, and physical growth. This has shifted the focus to blood glucose fluctuations, which are now clearly evident with continuous blood glucose monitoring and show a strong relationship with risk for DKD (45) and mortality (11). Certainly, in the current study, youth at high risk for DKD had increased glucose variability. In both youth at higher risk for DKD and in susceptible mice with glucose fluctuations or diabetes, glucose variability was associated with elevations in uACR and increased expression of KIM-1 on circulating T cells. Further, T-cell KIM-1 was a likely source of measured increases in plasma KIM-1, an early biomarker of progressive DKD in previous studies in high-risk youth (18,27,46). In the future, it would be interesting to track variability in blood glucose in real time using continuous blood glucose monitoring and examine acute and chronic effects on KIM-1 expression in T cells. This would allow better ascertainment of causality in a clinical context.

Glucose variability in the absence of diabetes can activate innate immune cells, resulting in atherosclerosis (34,47). In type 2 diabetes, dynamic modulation of T-cell subsets during oral glucose tolerance testing, irrespective of baseline glucose tolerance, has also been shown (48). In the current study, glucose variations stimulated the expression of the immunomodulatory molecule KIM-1 on circulating T cells in youth at high risk for DKD. While DKD is not considered as a primarily immune-mediated form of CKD (49), extensive evidence does support involvement of many components of the immune system in DKD progression (23). Certainly, CD4+ and CD8+ T cells infiltrate the kidney parenchyma in diabetes (24), and modest associations between urinary albumin excretion and fewer Treg cells, commonly beneficial in disease, have been demonstrated in type 2 diabetes (25). Dysfunctional CD8+ cytotoxic T cells (part of the Tconv subset) are definitely seen in youth with type 1 diabetes (50,51), but also in obese individuals with glucose intolerance at risk for kidney and CVD (52). In our study, there were fewer circulating KIM-1+CD4+ T cells, but a greater proportion of CD8+ T cells in youth at high risk for DKD, the majority of which were naive Treg cells. Given the heterogeneity of Treg cells, a more detailed study of their functionality in populations at risk for DKD is warranted. Interestingly, in a preclinical model of diabetes, short-term interruption of Treg cell function exacerbates kidney damage (53). This is consistent with other contexts, in which interruption of CD25 either via loss-of-function mutations in humans (54) or via specific knockout in humanized mice (55) resulted in loss of self-tolerance/autoimmunity and enhanced proliferation of CD8+ cells, as seen in the current study in high DKD risk youth. These findings might suggest that glucose variability in high-risk youth is the result of persistent autoimmunity and that CD8+ Treg cells adaptively increase in an attempt to combat this. The greater proportion of KIM-1+ naive CD8+ Treg cells to both activated or memory CD8+ Treg cells could also allude to a functional inability of this pathway to regulate and suppress autoimmunity or chronic inflammation, supporting this postulate. Although beyond the scope of the current study, the reasons for this compensatory or alternatively dysfunctional CD8+ Treg cell response in those more susceptible to DKD should be examined in the future, since previous studies show that particular MHC haplotypes that associate with autoimmunity also confer greater risk for developing vascular complications in diabetes (56).

Circulating concentrations of the immunomodulatory molecule KIM-1 were also elevated in the plasma of subjects at high risk of DKD, which increased collagen IV and SGLT2 expression when exposed to healthy kidney cells (renal PTECs), and this was prevented with KIM-1 blockade. KIM-1 is hypothesized to act as a double-edged sword in the injured kidney (57), where it acutely facilitates repair, removing cell debris from tubule lumens, and elicits anti-inflammatory antigen presentation to control immune responses (58). By contrast in CKD, persistent kidney KIM-1 expression stimulates inflammation and fibrosis, eventually spilling over into the urine (21). In this study, supported by our clinical and murine findings, we postulate that increases in circulating KIM-1 and T-cell KIM-1 expression precede changes in kidney or urinary KIM-1, which are known to occur later in DKD (59). In agreement, two previous studies in individuals with type 1 diabetes also demonstrated that circulating soluble KIM-1 was elevated prior to urinary KIM-1 concentrations (18,27,46). However, the source of these circulating increases in KIM-1 in diabetes has remained elusive. As supported by data in the current study, we hypothesize that KIM-1 is released into the circulation by T cells in response to glucose fluctuations. Certainly, neutralization of soluble KIM-1 in plasma from high-risk youth alleviated subsequent injury upon exposure to human primary PTECs. This does not, though, prove that the source of plasma KIM-1 was from T cells, although this is highly likely given that only T cells and injured kidney cells highly express this molecule. It is also possible that soluble KIM-1 from the bloodstream and produced locally by infiltrating T cells, as seen in our kidney biopsies, each compete with kidney cell KIM-1 for ligands (e.g., phosphatidylserine and TIM-4), thereby inhibiting local anti-inflammatory responses. Indeed, a recent study indicated that surprisingly, healthy kidneys had a high proportion of T cells in which physiological function remains relatively unknown (60). It would be interesting in the future to examine what specific T-cell subsets are present in healthy kidneys and if they express KIM-1 under specific conditions. If local or infiltrating kidney T cells chronically express and release soluble KIM-1 in response to glucose variations, this might interfere with the ability of PTECs to phagocytose other damaged kidney cells, blocking a known protective response by sequestering self-antigens from presentation to the immune system. PTECs can also directly present antigens to the immune system as seen in other auto-immune kidney diseases such as crescentic glomerulonephritis, in which KIM-1 is known to play a role (61). This might explain the paradox seen between the seemingly protective KIM-1+CD8+ and CD4+ regulatory profile and the increases in uACR, indicating changes in kidney function in our high-risk youth.

There were of course, limitations to our research. The first is that we do not have prospective data to indicate whether the youth categorized as high risk go on to develop overt DKD, since these young adults were between 20 and 29 years of age at recruitment. However, there is strong evidence from previous prospective studies that >85% of those youth with type 1 diabetes in the upper tertile of uACR do develop DKD. This lack of prospective follow-up is offset somewhat by our historical uACR data, in which persistent elevation in uACRs was seen from close to diabetes diagnosis until recruitment (with a mean observation period of ∼5 years), in youth at high risk for DKD. We also did not measure autoantibodies at the time of recruitment to assess autoimmunity, given that our youth had type 1 diabetes for 10 years on average, but maturity-onset diabetes of the young had been previously excluded. In addition, we did not examine the functionality of the KIM-1+ Tconv and Treg cells, which were increased in youth at risk for DKD, to see if this aligned with either autoimmunity or uACR. The kidney biopsies studied were also from older individuals with established DKD and represent a later stage of disease than present in our youth with type 1 diabetes. Therefore, future studies should examine T-cell infiltration and KIM-1 expression in early DKD in kidney biopsies such as those previously studied by Caramori et al. (62). However, many pathological pathways to DKD appear common to type 1 and type 2 diabetes (49,63). Reverse causality must also be considered for the association between increased KIM-1 in T cells and high risk for DKD in patients, but this is offset somewhat by the murine and PTEC studies. Despite these limitations, we have presented evidence of a novel pathway, by which kidney injury seen as increased uACR can be initiated via recruitment and expansion of KIM-1+ T cells as the result of glucose variations in early diabetes.

The data presented in this study suggest that glycemic variations align with risk for DKD in diabetes and increased CD8+ T-cell production of KIM-1. This highlights other pathways for consideration when designing therapies for the prevention of DKD and highlights the clinical importance of considering glycemic variability rather than purely relying on HbA1c.

H.L.B. and T.O’M.-S. have contributed equally.

See accompanying article, p. 1617.

This article contains supplementary material online at https://doi.org/10.2337/figshare.14186459.

Acknowledgments. The authors thank the outstanding Mater Young Adult Health Centre diabetes clinic nursing and administration staff as well as the flow cytometry and microscopy core facility staff at the Translational Research Institute (Brisbane, Queensland, Australia).

Funding. This work was supported by the National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases (DIACOMP 25034-61), Kidney Health Australia, National Health and Medical Research Council of Australia (GNT1102935 and GNT1160428), and the Mater Foundation (Equity Trustees and the Laurie Gertrude McCallum Estate and George Weaber Foundation Trusts). A.K.F. was supported by an Australian Government Research Training Program Scholarship. The Translational Research Institute is supported by a grant from the Australian Government.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. J.M.F. designed the experiments, analyzed and interpreted data, prepared the manuscript, and gained funding. D.A.M., A.J.K., A.K.F., K.T.K.G., A.G., M.A.S., R.W., K.J.R., N.F., and S.S.B. completed experiments including the ELISAs, flow cytometry, and PTEC experiments, analyzed and interpreted data, and assisted in manuscript preparation. D.A.M., A.J.M., M.C.F., and P.C. completed the mouse experiments. T.B., N.D’S., J.N., A.M., S.T., H.L.B., and T.O’M.-S. assisted type 1 diabetes clinical cohort design and collation. A.R., N.I., T.J., J.C., H.H., M.H., K.D., D.W.J., A.C., and H.L.B. assisted in study conceptualization and design and gaining financial support. All authors edited and approved the final manuscript. J.M.F. is the guarantor of this work and, as such, had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. This study was presented at the Australasian Diabetes Congress 2020, 11–13 November 2020.

1.
Huo
L
,
Harding
JL
,
Peeters
A
,
Shaw
JE
,
Magliano
DJ
.
Life expectancy of type 1 diabetic patients during 1997-2010: a national Australian registry-based cohort study
.
Diabetologia
2016
;
59
:
1177
1185
2.
Miller
RG
,
Mahajan
HD
,
Costacou
T
,
Sekikawa
A
,
Anderson
SJ
,
Orchard
TJ
.
A contemporary estimate of total mortality and cardiovascular disease risk in young adults with type 1 diabetes: the Pittsburgh Epidemiology of Diabetes Complications Study
.
Diabetes Care
2016
;
39
:
2296
2303
3.
Raghavan
S
,
Vassy
JL
,
Ho
YL
, et al
.
Diabetes mellitus-related all-cause and cardiovascular mortality in a national cohort of adults
.
J Am Heart Assoc
2019
;
8
:
e011295
4.
Gregg
EW
,
Cheng
YJ
,
Srinivasan
M
, et al
.
Trends in cause-specific mortality among adults with and without diagnosed diabetes in the USA: an epidemiological analysis of linked national survey and vital statistics data
.
Lancet
2018
;
391
:
2430
2440
5.
Rawshani
A
,
Sattar
N
,
Franzén
S
, et al
.
Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study
.
Lancet
2018
;
392
:
477
486
6.
Amin
R
,
Widmer
B
,
Prevost
AT
, et al
.
Risk of microalbuminuria and progression to macroalbuminuria in a cohort with childhood onset type 1 diabetes: prospective observational study
.
BMJ
2008
;
336
:
697
701
7.
Marcovecchio
ML
,
Chiesa
ST
,
Armitage
J
, et al.;
Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT) Study Group
.
Renal and cardiovascular risk according to tertiles of urinary albumin-to-creatinine ratio: the Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT)
.
Diabetes Care
2018
;
41
:
1963
1969
8.
Orchard
TJ
,
Secrest
AM
,
Miller
RG
,
Costacou
T
.
In the absence of renal disease, 20 year mortality risk in type 1 diabetes is comparable to that of the general population: a report from the Pittsburgh Epidemiology of Diabetes Complications Study
.
Diabetologia
2010
;
53
:
2312
2319
9.
Hirsch
IB
.
Glycemic variability and diabetes complications: does it matter? Of course it does!
Diabetes Care
2015
;
38
:
1610
1614
10.
Hall
H
,
Perelman
D
,
Breschi
A
, et al
.
Glucotypes reveal new patterns of glucose dysregulation
.
PLoS Biol
2018
;
16
:
e2005143
11.
Yoshida
N
,
Yamamoto
H
,
Shinke
T
, et al
.
Impact of CD14++CD16+ monocytes on plaque vulnerability in diabetic and non-diabetic patients with asymptomatic coronary artery disease: a cross-sectional study
.
Cardiovasc Diabetol
2017
;
16
:
96
12.
Lachin
JM
,
Bebu
I
,
Bergenstal
RM
, et al.;
DCCT/EDIC Research Group
.
Association of glycemic variability in type 1 diabetes with progression of microvascular outcomes in the Diabetes Control and Complications Trial
.
Diabetes Care
2017
;
40
:
777
783
13.
Penno
G
,
Solini
A
,
Bonora
E
, et al.;
Renal Insufficiency And Cardiovascular Events Study Group
.
HbA1c variability as an independent correlate of nephropathy, but not retinopathy, in patients with type 2 diabetes: the Renal Insufficiency And Cardiovascular Events (RIACE) Italian multicenter study
.
Diabetes Care
2013
;
36
:
2301
2310
14.
Wadén
J
,
Forsblom
C
,
Thorn
LM
,
Gordin
D
,
Saraheimo
M
;
Finnish Diabetic Nephropathy Study Group
.
A1C variability predicts incident cardiovascular events, microalbuminuria, and overt diabetic nephropathy in patients with type 1 diabetes
.
Diabetes
2009
;
58
:
2649
2655
15.
Marcovecchio
ML
,
Dalton
RN
,
Daneman
D
, et al.;
Adolescent type 1 Diabetes cardio-renal Intervention Trial (AdDIT) study group
.
A new strategy for vascular complications in young people with type 1 diabetes mellitus
.
Nat Rev Endocrinol
2019
;
15
:
429
435
16.
Marcovecchio
ML
,
Chiesa
ST
,
Bond
S
, et al.;
AdDIT Study Group
.
ACE inhibitors and statins in adolescents with type 1 diabetes
.
N Engl J Med
2017
;
377
:
1733
1745
17.
Mauer
M
,
Zinman
B
,
Gardiner
R
, et al
.
Renal and retinal effects of enalapril and losartan in type 1 diabetes
.
N Engl J Med
2009
;
361
:
40
51
18.
Nowak
N
,
Skupien
J
,
Niewczas
MA
, et al
.
Increased plasma kidney injury molecule-1 suggests early progressive renal decline in non-proteinuric patients with type 1 diabetes
.
Kidney Int
2016
;
89
:
459
467
19.
Mariat
C
,
Degauque
N
,
Balasubramanian
S
, et al
.
Tim-1 signaling substitutes for conventional signal 1 and requires costimulation to induce T cell proliferation
.
J Immunol
2009
;
182
:
1379
1385
20.
Han
WK
,
Bailly
V
,
Abichandani
R
,
Thadhani
R
,
Bonventre
JV
.
Kidney Injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury
.
Kidney Int
2002
;
62
:
237
244
21.
Vaidya
VS
,
Ramirez
V
,
Ichimura
T
,
Bobadilla
NA
,
Bonventre
JV
.
Urinary kidney injury molecule-1: a sensitive quantitative biomarker for early detection of kidney tubular injury
.
Am J Physiol Renal Physiol
2006
;
290
:
F517
F529
22.
Meyers
JH
,
Chakravarti
S
,
Schlesinger
D
, et al
.
TIM-4 is the ligand for TIM-1, and the TIM-1-TIM-4 interaction regulates T cell proliferation
.
Nat Immunol
2005
;
6
:
455
464
23.
Pichler
R
,
Afkarian
M
,
Dieter
BP
,
Tuttle
KR
.
Immunity and inflammation in diabetic kidney disease: translating mechanisms to biomarkers and treatment targets
.
Am J Physiol Renal Physiol
2017
;
312
:
F716
F731
24.
Moon
JY
,
Jeong
KH
,
Lee
TW
,
Ihm
CG
,
Lim
SJ
,
Lee
SH
.
Aberrant recruitment and activation of T cells in diabetic nephropathy
.
Am J Nephrol
2012
;
35
:
164
174
25.
Zeng
C
,
Shi
X
,
Zhang
B
, et al
.
The imbalance of Th17/Th1/Tregs in patients with type 2 diabetes: relationship with metabolic factors and complications
.
J Mol Med (Berl)
2012
;
90
:
175
186
26.
Zhang
Z
,
Humphreys
BD
,
Bonventre
JV
.
Shedding of the urinary biomarker kidney injury molecule-1 (KIM-1) is regulated by MAP kinases and juxtamembrane region
.
J Am Soc Nephrol
2007
;
18
:
2704
2714
27.
Colombo
M
,
Looker
HC
,
Farran
B
, et al.;
SUMMIT Investigators
.
Serum kidney injury molecule 1 and β2-microglobulin perform as well as larger biomarker panels for prediction of rapid decline in renal function in type 2 diabetes
.
Diabetologia
2019
;
62
:
156
168
28.
Johnson
DW
,
Jones
GR
,
Mathew
TH
, et al.;
Australasian Proteinuria Consensus Working Group
.
Chronic kidney disease and measurement of albuminuria or proteinuria: a position statement
.
Med J Aust
2012
;
197
:
224
225
29.
Johnson
DW
,
Atai
E
,
Chan
M
, et al.;
KHA-CARI
.
KHA-CARI guideline: Early chronic kidney disease: detection, prevention and management
.
Nephrology (Carlton)
2013
;
18
:
340
350
30.
Webster-Clark
M
,
Jaeger
B
,
Zhong
Y
, et al
.
Low agreement between modified-Schwartz and CKD-EPI eGFR in young adults: a retrospective longitudinal cohort study
.
BMC Nephrol
2018
;
19
:
194
31.
Turer
CB
,
Baum
M
,
Dubourg
L
,
Selistre
LS
,
Skinner
AC
.
Prevalence of hyperfiltration among US youth/young adults with overweight and obesity: A population-based association study
.
Obes Sci Pract
2019
;
5
:
570
580
32.
Donaghue
KC
,
Marcovecchio
ML
,
Wadwa
RP
, et al
.
ISPAD Clinical Practice Consensus Guidelines 2018: Microvascular and macrovascular complications in children and adolescents
.
Pediatr Diabetes
2018
;
19
(
Suppl. 27
):
262
274
33.
Vesey
DA
,
Qi
W
,
Chen
X
,
Pollock
CA
,
Johnson
DW
.
Isolation and primary culture of human proximal tubule cells
.
Methods Mol Biol
2009
;
466
:
19
24
34.
Flynn
MC
,
Kraakman
MJ
,
Tikellis
C
, et al
.
Transient intermittent hyperglycemia accelerates atherosclerosis by promoting myelopoiesis
.
Circ Res
2020
;
127
:
877
892
35.
Coughlan
MT
,
Higgins
GC
,
Nguyen
TV
, et al
.
Deficiency in apoptosis-inducing factor recapitulates chronic kidney disease via aberrant mitochondrial homeostasis
.
Diabetes
2016
;
65
:
1085
1098
36.
Dungan
KM
,
Buse
JB
,
Largay
J
, et al
.
1,5-anhydroglucitol and postprandial hyperglycemia as measured by continuous glucose monitoring system in moderately controlled patients with diabetes
.
Diabetes Care
2006
;
29
:
1214
1219
37.
Heidkamp
GF
,
Sander
J
,
Lehmann
CHK
, et al
.
Human lymphoid organ dendritic cell identity is predominantly dictated by ontogeny, not tissue microenvironment
.
Sci Immunol
2016
;
1
:
eaai7677
38.
Benz
K
,
Büttner
M
,
Dittrich
K
,
Campean
V
,
Dötsch
J
,
Amann
K
.
Characterisation of renal immune cell infiltrates in children with nephrotic syndrome
.
Pediatr Nephrol
2010
;
25
:
1291
1298
39.
Moriya
R
,
Manivel
JC
,
Mauer
M
.
Juxtaglomerular apparatus T-cell infiltration affects glomerular structure in type 1 diabetic patients
.
Diabetologia
2004
;
47
:
82
88
40.
Nathan
DM
,
Genuth
S
,
Lachin
J
, et al.;
Diabetes Control and Complications Trial Research Group
.
The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus
.
N Engl J Med
1993
;
329
:
977
986
41.
UK Prospective Diabetes Study (UKPDS) Group
.
Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34)
.
Lancet
1998
;
352
:
854
865
42.
Patel
A
,
MacMahon
S
,
Chalmers
J
, et al.;
ADVANCE Collaborative Group
.
Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes
.
N Engl J Med
2008
;
358
:
2560
2572
43.
Gerstein
HC
,
Miller
ME
,
Byington
RP
, et al.;
Action to Control Cardiovascular Risk in Diabetes Study Group
.
Effects of intensive glucose lowering in type 2 diabetes
.
N Engl J Med
2008
;
358
:
2545
2559
44.
Miller
KM
,
Foster
NC
,
Beck
RW
, et al.;
T1D Exchange Clinic Network
.
Current state of type 1 diabetes treatment in the U.S.: updated data from the T1D Exchange clinic registry
.
Diabetes Care
2015
;
38
:
971
978
45.
Bergenstal
RM
.
Glycemic variability and diabetes complications: does it matter? simply put, there are better glycemic markers!
Diabetes Care
2015
;
38
:
1615
1621
46.
Colombo
M
,
Valo
E
,
McGurnaghan
SJ
, et al.;
FinnDiane Study Group and the Scottish Diabetes Research Network (SDRN) Type 1 Bioresource Collaboration
.
Biomarker panels associated with progression of renal disease in type 1 diabetes
.
Diabetologia
2019
;
62
:
1616
1627
47.
Nagareddy
PR
,
Murphy
AJ
,
Stirzaker
RA
, et al
.
Hyperglycemia promotes myelopoiesis and impairs the resolution of atherosclerosis
.
Cell Metab
2013
;
17
:
695
708
48.
Miya
A
,
Nakamura
A
,
Miyoshi
H
, et al
.
Impact of glucose loading on variations in CD4+ and CD8+ T cells in Japanese participants with or without type 2 Diabetes
.
Front Endocrinol (Lausanne)
2018
;
9
:
81
49.
Forbes
JM
,
Cooper
ME
.
Mechanisms of diabetic complications
.
Physiol Rev
2013
;
93
:
137
188
50.
Faustman
DL
,
Davis
M
.
The primacy of CD8 T lymphocytes in type 1 diabetes and implications for therapies
.
J Mol Med (Berl)
2009
;
87
:
1173
1178
51.
Garyu
JW
,
Uduman
M
,
Stewart
A
, et al
.
Characterization of diabetogenic CD8+ T cells: immune therapy with metabolic blockade
.
J Biol Chem
2016
;
291
:
11230
11240
52.
Wagner
NM
,
Brandhorst
G
,
Czepluch
F
, et al
.
Circulating regulatory T cells are reduced in obesity and may identify subjects at increased metabolic and cardiovascular risk
.
Obesity (Silver Spring)
2013
;
21
:
461
468
53.
Eller
K
,
Kirsch
A
,
Wolf
AM
, et al
.
Potential role of regulatory T cells in reversing obesity-linked insulin resistance and diabetic nephropathy
.
Diabetes
2011
;
60
:
2954
2962
54.
Sharfe
N
,
Dadi
HK
,
Shahar
M
,
Roifman
CM
.
Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor
.
Proc Natl Acad Sci USA
1997
;
94
:
3168
3171
55.
Goudy
K
,
Aydin
D
,
Barzaghi
F
, et al
.
Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity
.
Clin Immunol
2013
;
146
:
248
261
56.
Söderlund
J
,
Forsblom
C
,
Ilonen
J
, et al.;
FinnDiane Study Group
.
HLA class II is a factor in cardiovascular morbidity and mortality rates in patients with type 1 diabetes
.
Diabetologia
2012
;
55
:
2963
2969
57.
Ichimura
T
,
Brooks
CR
,
Bonventre
JV
.
Kim-1/Tim-1 and immune cells: shifting sands
.
Kidney Int
2012
;
81
:
809
811
58.
Yang
L
,
Brooks
CR
,
Xiao
S
, et al
.
KIM-1-mediated phagocytosis reduces acute injury to the kidney
.
J Clin Invest
2015
;
125
:
1620
1636
59.
Panduru
NM
,
Sandholm
N
,
Forsblom
C
, et al.;
FinnDiane Study Group
.
Kidney injury molecule-1 and the loss of kidney function in diabetic nephropathy: a likely causal link in patients with type 1 diabetes
.
Diabetes Care
2015
;
38
:
1130
1137
60.
Park
J
,
Shrestha
R
,
Qiu
C
, et al
.
Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease
.
Science
2018
;
360
:
758
763
61.
Nozaki
Y
,
Nikolic-Paterson
DJ
,
Snelgrove
SL
, et al
.
Endogenous Tim-1 (Kim-1) promotes T-cell responses and cell-mediated injury in experimental crescentic glomerulonephritis
.
Kidney Int
2012
;
81
:
844
855
62.
Caramori
ML
,
Parks
A
,
Mauer
M
.
Renal lesions predict progression of diabetic nephropathy in type 1 diabetes
.
J Am Soc Nephrol
2013
;
24
:
1175
1181
63.
Forbes
JM
,
Fotheringham
AK
.
Vascular complications in diabetes: old messages, new thoughts
.
Diabetologia
2017
;
60
:
2129
2138
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.