Heat shock proteins (HSPs) are induced by cellular stress and function as molecular chaperones that regulate protein folding. Diabetes impairs the function/expression of many HSPs, including HSP70 and HSP90, key regulators of pathological mechanisms involved in diabetes complications. Therefore, we investigated whether pharmacological HSP90 inhibition ameliorates diabetes-associated renal damage and atheroprogression in a mouse model of combined hyperglycemia and hyperlipidemia (streptozotocin-induced diabetic apolipoprotein E–deficient mouse). Treatment of diabetic mice with 17-dimethylaminoethylamino-17-demethoxygeldanamycin (DMAG, 2 and 4 mg/kg, 10 weeks) improved renal function, as evidenced by dose-dependent decreases in albuminuria, renal lesions (mesangial expansion, leukocyte infiltration, and fibrosis), and expression of proinflammatory and profibrotic genes. Furthermore, DMAG significantly reduced atherosclerotic lesions and induced a more stable plaque phenotype, characterized by lower content of lipids, leukocytes, and inflammatory markers, and increased collagen and smooth muscle cell content. Mechanistically, the renoprotective and antiatherosclerotic effects of DMAG are mediated by the induction of protective HSP70 along with inactivation of nuclear factor-κB (NF-κB) and signal transducers and activators of transcription (STAT) and target gene expression, both in diabetic mice and in cultured cells under hyperglycemic and proinflammatory conditions. In conclusion, HSP90 inhibition by DMAG restrains the progression of renal and vascular damage in experimental diabetes, with potential implications for the prevention of diabetes complications.

The prevalence of diabetes has reached epidemic proportions. Nephropathy and atherosclerosis are the major diabetes-driven complications, resulting in disability and increased risk of mortality in diabetic patients. In fact, diabetic nephropathy is the leading cause of end-stage renal disease and also a macrovascular disease risk factor, and atherosclerosis is the main reason for impaired life expectancy in diabetic patients (1,2). Therefore, the prevention of micro- and macrovasculature damage associated with diabetes to improve patient quality of life and life expectancy is a major public health concern. Owing to the fact that current treatment of diabetes is insufficient to avert the complications in a significant proportion of patients, diabetes-associated renal and vascular damage might be counteracted by blocking injury mechanisms and promoting protective and reparative factors. Based on both observational clinical studies and functional evidence from animal models, inflammation is considered a key contributor to the onset and progression of diabetic nephropathy and atherosclerosis, with promising therapeutic opportunities (2,3).

Cellular stress response is directed by the conserved family of heat shock proteins (HSPs) to promote cytoprotection through the transcriptional regulation by heat shock transcription factor 1 (HSF1) (4). Given the ubiquity of HSP, it seems unsurprising that these chaperone proteins have been related to many metabolic abnormalities. Evidence from patients and animal models indicate that diabetes differentially modulates the expression of HSP25/27, HSP60, HSP70/72, and HSP90 (57), and this may affect the ability of cells to mount an effective cytoprotective response. HSP90, one of the most conserved HSPs, uses the energy generated by ATP binding and hydrolysis to allow folding, maturation, and stability of the so-called “client” proteins involved in cell survival, signal transduction, transcriptional regulation, and innate and adaptive immunity (4). To do so, HSP90 interacts with other cochaperones, forming a multichaperone complex that recognizes client proteins and modulates their activities (4). HSP90 machinery in association with its cochaperone HSP70 is involved in folding and activation of newly synthesized protein kinases, including IκB kinase (IKK) and Janus kinase (JAK) (8,9). IKK and JAK play, respectively, a key role in the activation of nuclear factor-κB (NF-κB) and signal transducers and activators of transcription (STAT), major transcription factors that regulate the expression of many genes involved in inflammation (10,11). This overall prompted the notion of HSP90 as a molecular target of interest for the treatment of several immunological and inflammatory disorders (12,13). So far, most clinical trials of HSP90 inhibitors have focused on cancer therapy (14).

Furthermore, HSP90 inhibitors effectively disrupt HSP90-HSF1 complexes, allowing HSF1 activation and subsequent nuclear translocation to transactivate HSP genes (4,14). Preclinical data implicate HSP90 as a promising anti-inflammatory target in rheumatoid arthritis, systemic lupus erythematosus, uveitis, liver injury, and cardiovascular disease models (12,1518). In diabetic animals, HSP90 inhibition improved insulin sensitivity (19), high-fat diet–induced renal failure (20), and neurodegeneration (21), but the underlying mechanisms involved in these antidiabetic actions are not well defined. Hence, in this work we investigate whether targeting HSP90 delays the progression of diabetes complications in a diabetes mouse model of concomitant renal and macrovascular disease.

Diabetes Model

Animal studies were performed according to the Directive 2010/63/EU of the European Parliament and were approved by the Institutional Animal Care and Use Committee (IIS-Fundacion Jimenez Diaz). Experimental diabetes model of insulin deficiency was induced in 10-week-old male apolipoprotein E–deficient (apoE−/−) mice by two daily intraperitoneal injections of streptozotocin (STZ, 125 mg/kg/day; Sigma-Aldrich, St. Louis, MO) (2225). Animals maintained on standard diet were monitored every 2–3 days for body weight and nonfasting blood glucose. Severely hyperglycemic mice (blood glucose >29 mmol/L) received insulin (1–1.5 IU) to maintain blood glucose levels within a more tolerable range. Mice with overt diabetes (glucose >19.4 mmol/L) were randomized to receive 10 weeks of treatment with vehicle (200 μL saline i.p., every other day; n = 9) and 17-dimethylaminoethylamino-17-demethoxygeldanamycin (DMAG; InvivoGen, San Diego, CA) at 2 mg/kg/day (n = 9) and 4 mg/kg/day (n = 6). Age-matched apoE−/− mice were used as nondiabetic controls (vehicle, n = 4; DMAG 4 mg/kg/day, n = 3). At the end of the study, 16 h–fasted mice were anesthetized (100 mg/kg ketamine and 15 mg/kg xylazine), saline perfused, and killed. Blood and urine samples were collected for biochemistry.

Histological Analysis

Dissected kidneys were snap-frozen for expression studies or stored in 4% paraformaldehyde for histology. Aortas were divided into two parts: the upper aortic root was embedded in Tissue-Tek O.C.T. Compound (Sakura Finetek Europe, the Netherlands) for histological analysis, and the abdominal/thoracic aorta was processed for mRNA analysis.

Paraffin-embedded kidney sections (3 μm) were stained with periodic acid Schiff (PAS). Renal lesions were semiquantitatively graded (0–3 scale) in a blinded manner according to the extent of lesions in glomeruli (hypertrophy, hypercellularity, and mesangial expansion; 30 glomeruli per sample), tubules (atrophy and degeneration), and interstitium (fibrosis and infiltration; 20 fields at ×40 per sample). Glomerular area and PAS+ mesangial area were quantified by computerized morphometry. Immunodetection of macrophages (F4/80; AbD Serotec, Oxford, U.K.), T cells (CD3; DAKO, Glostrup, Denmark), HSP70 (Abcam), and tyrosine-phosphorylated STAT1 (P-STAT1; Life Technologies, Rockville, MD) and STAT3 (P-STAT3; Santa Cruz Biotechnology, Inc., Santa Cruz, CA) was assessed by immunoperoxidase. Renal fibrosis was determined by picrosirius red staining. Positive staining (>10 fields at ×20 magnification) was quantified using Image Pro-Plus (Media Cybernetics, Bethesda, MD) and expressed as percentage of total area and number of positive cells per glomerular cross-section or interstitial area measured (mm2).

Atherosclerotic lesions in serial 8-μm aortic sections (covering ∼1,000 μm from valve leaflets) were quantified by morphometry after Oil Red O/hematoxylin staining. Individual lesion area was determined by averaging the maximal values (approximately three sections). Collagen content was determined by picrosirius red staining. Lesional macrophages (MOMA-2; AbD Serotec, Oxford, U.K.), CD3 T cells, vascular smooth muscle cells (VSMCs) (α-actin-Cy3; Sigma-Aldrich), chemokine (C-C motif) ligand (CCL) 2 (Santa Cruz Biotechnology), CCL5 (Antibodies Online), and tumor necrosis factor α (TNF-α; Santa Cruz Biotechnology) were detected by immunoperoxidase or immunofluorescence. Positive staining was expressed as percentage of total plaque area or number of positive cells per lesion area (26,27). Activated NF-κB in kidney and aorta was detected by Southwestern histochemistry using digoxigenin-labeled probes (26,28).

Cell Cultures

Primary mesangial cells (MCs) from mouse kidneys were cultured in RPMI 1640 with 25 mmol/L HEPES, pH 7.4, supplemented with 10% FBS (22). Murine MC line (SV40 MES 13, CRL-1927; American Type Culture Collection [ATCC], Manassas, VA) was maintained in Dulbecco’s modified Eagle’s medium (DMEM):F12 medium containing 5% FBS. Murine proximal tubuloepithelial MCT line (29) was cultured in RPMI 1640 containing 10% FBS. Mouse vascular endothelial cell line (MILE SVEN 1, CRL-2279; ATCC) was maintained in DMEM with 5% FBS. Murine bone marrow–derived macrophages were cultured for 7 days in RPMI 1640 with 10% FBS containing 10% L929-cell conditioned medium (23). Murine monocyte/macrophage cell line (RAW 264.7, TIB-71; ATCC) was maintained in DMEM with 10% FBS. All culture media were supplemented with 100 units/mL penicillin, 100 g/mL streptomycin, and 2 mmol/L l-glutamine (Life Technologies). Quiescent cells were pretreated for 4 h with DMAG (5–50 nmol/L) before stimulation with high glucose (HG; 30 mmol/L d-glucose; Sigma-Aldrich) or cytokines (interleukin-6 [IL-6] 102 units/mL plus interferon-γ [IFN-γ] 103 units/mL; PeproTech, Rocky Hill, NJ). Cell viability was assessed by the 1-(4,5-dimethylthiazol2-yl)-3,5-diphenylformazan thiazolyl blue formazan (MTT) method.

Transfection of Small Interfering RNA

Cells grown to 60–70% confluence were transfected with 20–30 nmol/L of small interfering RNA (siRNA) targeting HSP70 or negative control scramble siRNA (Ambion) using Lipofectamine RNAiMAX reagent (Life Technologies). Transfected cells (silencing efficiency 50–75%) were pretreated with DMAG for 4 h before 24 h of cytokine stimulation.

mRNA Expression Analysis

Total RNA from mouse tissues (kidney and aorta) and cultured cells was extracted with TRIzol (Life Technologies) and analyzed by real-time quantitative PCR using Taqman gene expression assays (Applied Biosystem, Foster City, CA). Target gene expression was normalized to housekeeping gene (18S).

Protein Expression Analysis

Total cell or kidney whole lysate proteins were resolved on SDS-PAGE gels and immunoblotted for fibronectin (Millipore Corporation, Billerica, MA), HSP32, HSP70 (Enzo Life Sciences, Farmingdale, NY), transforming growth factor-β (TGF-β), HSP27, HSP90, IκBα (Santa Cruz Biotechnology), P-STAT1 (Life Technologies), and P-STAT3 (Cell Signaling, Beverly, MA), using β-actin (Santa Cruz Biotechnology) or α-tubulin (Sigma-Aldrich) as loading controls. CCL2 and CCL5 protein levels were measured by ELISA (BD Biosciences; R&D Systems, Minneapolis, MN).

Cell Migration Assay

Renal cell chemoattractant capacity was determined using a transwell coculture system (28). In brief, RAW 264.7 cells (1 × 105) were seeded on the upper transwell inserts (8.0-µm pore size; Merck Millipore, Billerica, MA) and then placed onto a 24-well plate containing MC or MCT cells (5 × 104 per well), which were treated for 4 h with DMAG prior to cytokine stimulation (IL-6 + IFN-γ, 20 h). Cocultures were incubated for a further 8 h, nonmigratory cells were removed from the upper surface of the membrane, and migrated cells were fixed (4% paraformaldehyde), stained (0.2% crystal violet), and counted on 10 randomly selected ×200 fields.

Statistics

Values are expressed as mean ± SEM. Statistical analyses were performed using Prism 5 (GraphPad Software, Inc., La Joya, CA). Differences across groups were considered significant at P < 0.05 using either nonparametric Mann-Whitney U test or one-way ANOVA followed by post hoc Bonferroni pairwise comparison test.

Metabolic and Biochemical Effects of HSP90 Inhibitor in Mice

STZ-induced pancreatic injury in apoE−/− mice represents an insulin-deficient model that combines hyperglycemia and hyperlipidemia. Diabetic apoE−/− mice develop accelerated renal and vascular injury, with similarities to human diabetic nephropathy and atherosclerosis (24,30). We compared the evolution of diabetes in apoE−/− mice after treatment with either vehicle or DMAG at two different concentrations (2 and 4 mg/kg) for 10 weeks. Throughout the study, blood glucose curves were similar in the three diabetic groups (Supplementary Fig. 1). At the end of the study, levels of blood glucose, GHbA1c, total cholesterol, HDL and LDL cholesterol, and triglycerides were all significantly higher in diabetic apoE−/− mice compared with nondiabetic controls, but they were not affected by DMAG treatment (Table 1). Serum transaminase activities were also similar across the groups (Table 1), indicating preserved liver function. As expected, diabetic mice had evidence of enhanced kidney disease, with significant increases in serum creatinine, kidney-to-body weight ratio, and urinary albumin-to-creatinine ratio (Table 1). Interestingly, DMAG treatment in diabetic animals prevented body weight loss and also significantly decreased serum creatinine, relative kidney weight, and albuminuria (Table 1), reflecting an improved renal function by DMAG.

Table 1

Metabolic and renal data in normoglycemic control and diabetic apoE−/− mice after 10 weeks of treatment

Control
Diabetes
Vehicle (n = 4)DMAG 4 mg/kg (n = 3)Vehicle (n = 9)DMAG 2 mg/kg (n = 9)DMAG 4 mg/kg (n = 6)
ΔBW (g) 2.5 ± 0.5 2.3 ± 0.3 −3.3 ± 0.3 −2.9 ± 0.1 −1.3 ± 0.1†† 
Blood glucose (mmol/L) 9.2 ± 0.5 8.7 ± 0.8 29.3 ± 0.7*** 31.1 ± 1.5** 29.6 ± 2.4*** 
GHbA1c, % (mmol/mol) ND ND 3.3 ± 0.4 (12 ± 2) 3.3 ± 0.9 (13 ± 1) 3.2 ± 0.6 (11 ± 4) 
Total chol (mmol/L) 8.5 ± 0.4 9.4 ± 0.5 15.6 ± 0.4*** 17.9 ± 1.1*** 14.3 ± 1.4* 
LDL chol (mmol/L) 8.0 ± 0.4 8.9 ± 0.5 15.1 ± 0.4*** 17.3 ± 1.1*** 13.7 ± 1.4* 
HDL chol (mmol/L) 0.33 ± 0.02 0.30 ± 0.02 0.29 ± 0.03 0.20 ± 0.01**, 0.26 ± 0.02 
TG (mmol/L) 0.62 ± 0.03 0.62 ± 0.07 0.86 ± 0.07 0.86 ± 0.06 0.62 ± 0.06 
ALT (units/L) 78 ± 8 68 ± 9 117 ± 11 95 ± 18 80 ± 19 
AST (units/L) 185 ± 10 110 ± 16 209 ± 15 170 ± 18 176 ± 34 
KBWR (g/kg) 14.8 ± 0.4 14.7 ± 0.4 20.5 ± 1.1** 15.0 ± 0.5††† 15.3 ± 0.8†† 
SCr (μmol/L) 11.1 ± 2.2 8.8 ± 0.0 36.2 ± 4.4*** 19.4 ± 1.8†† 20.3 ± 2.6 
UAC (μg/μmol) 7.2 ± 0.2 6.9 ± 0.3 22.6 ± 1.4*** 13.0 ± 0.8*,††† 16.5 ± 0.8***,†† 
PAS+ mesangial area (%) 4.6 ± 0.2 4.8 ± 0.4 14.3 ± 0.9*** 9.7 ± 1.1*,†† 8.8 ± 0.7†† 
Glomerular area (μm22,188 ± 107 2,309 ± 134 4,032 ± 229 3,080 ± 214 2,379 ± 213†† 
Control
Diabetes
Vehicle (n = 4)DMAG 4 mg/kg (n = 3)Vehicle (n = 9)DMAG 2 mg/kg (n = 9)DMAG 4 mg/kg (n = 6)
ΔBW (g) 2.5 ± 0.5 2.3 ± 0.3 −3.3 ± 0.3 −2.9 ± 0.1 −1.3 ± 0.1†† 
Blood glucose (mmol/L) 9.2 ± 0.5 8.7 ± 0.8 29.3 ± 0.7*** 31.1 ± 1.5** 29.6 ± 2.4*** 
GHbA1c, % (mmol/mol) ND ND 3.3 ± 0.4 (12 ± 2) 3.3 ± 0.9 (13 ± 1) 3.2 ± 0.6 (11 ± 4) 
Total chol (mmol/L) 8.5 ± 0.4 9.4 ± 0.5 15.6 ± 0.4*** 17.9 ± 1.1*** 14.3 ± 1.4* 
LDL chol (mmol/L) 8.0 ± 0.4 8.9 ± 0.5 15.1 ± 0.4*** 17.3 ± 1.1*** 13.7 ± 1.4* 
HDL chol (mmol/L) 0.33 ± 0.02 0.30 ± 0.02 0.29 ± 0.03 0.20 ± 0.01**, 0.26 ± 0.02 
TG (mmol/L) 0.62 ± 0.03 0.62 ± 0.07 0.86 ± 0.07 0.86 ± 0.06 0.62 ± 0.06 
ALT (units/L) 78 ± 8 68 ± 9 117 ± 11 95 ± 18 80 ± 19 
AST (units/L) 185 ± 10 110 ± 16 209 ± 15 170 ± 18 176 ± 34 
KBWR (g/kg) 14.8 ± 0.4 14.7 ± 0.4 20.5 ± 1.1** 15.0 ± 0.5††† 15.3 ± 0.8†† 
SCr (μmol/L) 11.1 ± 2.2 8.8 ± 0.0 36.2 ± 4.4*** 19.4 ± 1.8†† 20.3 ± 2.6 
UAC (μg/μmol) 7.2 ± 0.2 6.9 ± 0.3 22.6 ± 1.4*** 13.0 ± 0.8*,††† 16.5 ± 0.8***,†† 
PAS+ mesangial area (%) 4.6 ± 0.2 4.8 ± 0.4 14.3 ± 0.9*** 9.7 ± 1.1*,†† 8.8 ± 0.7†† 
Glomerular area (μm22,188 ± 107 2,309 ± 134 4,032 ± 229 3,080 ± 214 2,379 ± 213†† 

Data are mean ± SEM. ALT, alanine aminotransferase; AST, aspartate aminotransferase; ΔBW, body weight change (final − initial); chol, cholesterol; KBWR, kidney-to-body weight ratio; ND, not determined; SCr, serum creatinine; TG, triglyceride; UAC, urine albumin-to-creatinine ratio.

*P < 0.05;

**P < 0.01;

***P < 0.001 vs. control-vehicle;

P < 0.05;

††P < 0.01;

†††P < 0.001 vs. diabetes-vehicle.

DMAG Treatment Ameliorates Diabetic Kidney Disease in Mice

Histopathological examination of PAS-stained sections revealed that diabetes caused moderate to severe renal damage in apoE−/− mice, compared with nondiabetic controls (Fig. 1A–C). Furthermore, DMAG treatment dose-dependently ameliorated the renal pathologic changes associated with diabetes, including: 1) glomerular hypertrophy, hypercellularity (mesangial proliferation and infiltrating cells), mesangial matrix expansion, and capillary dilation; 2) tubular atrophy, epithelial dilation, and deposits of glycogen; and 3) interstitial fibrosis and inflammatory infiltrate (Fig. 1A–C and Table 1). DMAG had no significant effect on any parameters measured in nondiabetic controls (Fig. 1A–C and Table 1).

Figure 1

DMAG protects from diabetes-associated renal injury in apoE−/− mice. A: Representative images (scale bar, 20 μm) of PAS-stained kidney sections from nondiabetic control and diabetic apoE−/− mice treated with vehicle or DMAG (2 and 4 mg/kg) for 10 weeks. Semiquantitative assessments of lesions in glomeruli (B) and tubulointerstitial compartment (C). Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. control-vehicle group; ††P < 0.01 and †††P < 0.001 vs. diabetes-vehicle group.

Figure 1

DMAG protects from diabetes-associated renal injury in apoE−/− mice. A: Representative images (scale bar, 20 μm) of PAS-stained kidney sections from nondiabetic control and diabetic apoE−/− mice treated with vehicle or DMAG (2 and 4 mg/kg) for 10 weeks. Semiquantitative assessments of lesions in glomeruli (B) and tubulointerstitial compartment (C). Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. control-vehicle group; ††P < 0.01 and †††P < 0.001 vs. diabetes-vehicle group.

Close modal

Quantification of F4/80-positive macrophages (Fig. 2A) and CD3-positive T cells (Fig. 2B) demonstrated a marked decrease (∼60%) in the number of infiltrating cells in both glomeruli and interstitium of diabetic mice treated with DMAG, compared with the vehicle-treated group. Consistently, DMAG reduced the gene expression levels of monocyte and T-cell chemokines (CCL2 and CCL5) and proinflammatory cytokine TNF-α in diabetic kidneys, with significant effect at the highest dose (% decrease vs. vehicle: 59 ± 8, 45 ± 12, and 85 ± 3, respectively; P < 0.05) (Fig. 2C). Chemokine protein levels in diabetic kidneys were also found significantly decreased in DMAG groups (Fig. 2D).

Figure 2

Attenuated inflammation in kidneys from DMAG-treated diabetic mice. Immunoperoxidase images (scale bar, 20 μm) of F4/80+ macrophages (A) and CD3+ T-cell (B) staining in kidney samples from diabetic apoE−/− mice. Bottom panels: Quantification of mononuclear cell infiltration in glomeruli and interstitium. C: Real-time PCR analysis of inflammatory gene expression in renal cortex from diabetic mice. Values normalized by 18S are expressed in arbitrary units (a.u.). D: CCL2 and CCL5 protein expression in the renal cortical lysates from diabetic mice were measured by ELISA. The dashed line is a separation between CCL2 and CCL5 data. White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are mean ± SEM (n = 6–9 animals per group). *P < 0.05, **P < 0.01, and ***P < 0.001 vs. diabetes-vehicle group. gcs, glomerular cross-section.

Figure 2

Attenuated inflammation in kidneys from DMAG-treated diabetic mice. Immunoperoxidase images (scale bar, 20 μm) of F4/80+ macrophages (A) and CD3+ T-cell (B) staining in kidney samples from diabetic apoE−/− mice. Bottom panels: Quantification of mononuclear cell infiltration in glomeruli and interstitium. C: Real-time PCR analysis of inflammatory gene expression in renal cortex from diabetic mice. Values normalized by 18S are expressed in arbitrary units (a.u.). D: CCL2 and CCL5 protein expression in the renal cortical lysates from diabetic mice were measured by ELISA. The dashed line is a separation between CCL2 and CCL5 data. White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are mean ± SEM (n = 6–9 animals per group). *P < 0.05, **P < 0.01, and ***P < 0.001 vs. diabetes-vehicle group. gcs, glomerular cross-section.

Close modal

To assess the extent of renal fibrosis, we measured the collagen content by picrosirius red staining in kidney sections from nondiabetic and diabetic mice. DMAG effectively reduced the glomerular and tubulointerstitial fibrosis in diabetic apoE−/− mice without significant effect in nondiabetic controls (Fig. 3A and B). Furthermore, diabetic mice treated with DMAG exhibited significantly lower levels of extracellular matrix proteins (type I collagen and fibronectin) and profibrotic factor TGF-β than the vehicle-treated group (Fig. 3C and D). DMAG also suppressed the renal expression of kidney injury molecule-1 (Kim-1) (Fig. 3C), a sensitive biomarker of tubular injury in several kidney diseases.

Figure 3

DMAG reduces renal fibrosis in diabetic apoE−/− mice. A: Representative images (scale bar, 20 μm) of picrosirius red staining in renal sections from nondiabetic control and diabetic apoE−/− mice treated with vehicle or DMAG (2 and 4 mg/kg) for 10 weeks. B: Quantitative analysis of collagen-positive area in glomerular and tubulointerstitial compartments. C: Real-time PCR analysis of mRNA expression of type I collagen (Col I), fibronectin (Fn), profibrotic factor (Tgf-β), and tubular injury marker (Kim-1) in renal cortex from diabetic mice. Values normalized by 18S are expressed in arbitrary units (a.u.). D: Western blot analyses of fibronectin (FN) and TGF-β expression in renal cortical lysates from diabetic mice. Shown are representative blots and the summary of normalized densitometric quantification. Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05 and ***P < 0.001 vs. control-vehicle group; †P < 0.05, ††P < 0.01, and †††P < 0.001 vs. diabetes-vehicle group. Veh, vehicle.

Figure 3

DMAG reduces renal fibrosis in diabetic apoE−/− mice. A: Representative images (scale bar, 20 μm) of picrosirius red staining in renal sections from nondiabetic control and diabetic apoE−/− mice treated with vehicle or DMAG (2 and 4 mg/kg) for 10 weeks. B: Quantitative analysis of collagen-positive area in glomerular and tubulointerstitial compartments. C: Real-time PCR analysis of mRNA expression of type I collagen (Col I), fibronectin (Fn), profibrotic factor (Tgf-β), and tubular injury marker (Kim-1) in renal cortex from diabetic mice. Values normalized by 18S are expressed in arbitrary units (a.u.). D: Western blot analyses of fibronectin (FN) and TGF-β expression in renal cortical lysates from diabetic mice. Shown are representative blots and the summary of normalized densitometric quantification. Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05 and ***P < 0.001 vs. control-vehicle group; †P < 0.05, ††P < 0.01, and †††P < 0.001 vs. diabetes-vehicle group. Veh, vehicle.

Close modal

HSP90 Inhibition Modulates HSP70 Expression and Inflammatory Signaling Pathways in Diabetic Kidneys

To assess the efficiency of DMAG on HSP90 inhibition, we studied the effects on its cochaperone HSP70, which is upregulated as a result of the blockage of the ATP-binding site of HSP90 (14). As expected, DMAG upregulated the mRNA and protein expression of HSP70 (∼2–3.5-fold increase vs. vehicle), but not HSP90, in diabetic kidneys, with a wide distribution in both glomerular and tubular regions of DMAG-treated mice (Supplementary Fig. 2A–C).

Among the different HSP90-associated proteins, we further analyzed whether DMAG modulates renal activation of NF-κB and STAT, key intracellular pathways in the regulation of inflammatory and fibrotic genes (3). In situ Southwestern histochemistry revealed an intense nuclear localization of activated NF-κB in glomerular and tubulointerstitial cells of diabetic mice, and a dose-dependent inhibition by DMAG treatment (Fig. 4A–C). DMAG administration also prevented STAT1 and STAT3 activation in diabetic kidneys, as demonstrated by immunodetection of tyrosine-phosphorylated proteins (Fig. 4A–D).

Figure 4

HSP90 inhibition attenuates NF-κB and STAT in diabetic kidneys. In situ detection of activated NF-κB (Southwestern histochemistry) and phosphorylated STAT proteins (P-STAT1 and P-STAT3; immunoperoxidase) in kidney sections from diabetic apoE−/− mice after 10 weeks of treatment with vehicle or DMAG (2 and 4 mg/kg). Representative micrographs (A) (scale bar, 20 μm) and quantification of positive staining in glomerular (B) and tubulointerstitial (C) compartments are shown. D: Western blot analyses of P-STAT1 and P-STAT3 in renal cortical lysates from diabetic mice. Shown are representative images and the summary of normalized quantification, expressed in arbitrary units (a.u.). White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are mean ± SEM (n = 6–9 animals per group). *P < 0.05, **P < 0.01, and ***P < 0.001 vs. diabetes-vehicle group. gcs, glomerular cross-section; Veh, vehicle.

Figure 4

HSP90 inhibition attenuates NF-κB and STAT in diabetic kidneys. In situ detection of activated NF-κB (Southwestern histochemistry) and phosphorylated STAT proteins (P-STAT1 and P-STAT3; immunoperoxidase) in kidney sections from diabetic apoE−/− mice after 10 weeks of treatment with vehicle or DMAG (2 and 4 mg/kg). Representative micrographs (A) (scale bar, 20 μm) and quantification of positive staining in glomerular (B) and tubulointerstitial (C) compartments are shown. D: Western blot analyses of P-STAT1 and P-STAT3 in renal cortical lysates from diabetic mice. Shown are representative images and the summary of normalized quantification, expressed in arbitrary units (a.u.). White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are mean ± SEM (n = 6–9 animals per group). *P < 0.05, **P < 0.01, and ***P < 0.001 vs. diabetes-vehicle group. gcs, glomerular cross-section; Veh, vehicle.

Close modal

Impact of DMAG on Diabetes-Associated Atherosclerosis

Quantification of aortic root sections after Oil Red O/hematoxylin staining (Fig. 5A–C) revealed that the induction of diabetes exacerbates atherosclerosis in apoE−/− mice (2.5-fold increase in area compared with nondiabetic controls) and also demonstrates the atheroprotective effect of DMAG. In fact, DMAG attenuated atherosclerosis in nondiabetic apoE−/− mice (Fig. 5A and B), as previously reported (17). Remarkably, atherosclerotic plaques of diabetic mice with DMAG treatment exhibited a dose-dependent decrease in the lesion size (Fig. 5B), extension (Fig. 5C), and neutral lipid content (% Oil Red O–positive area: vehicle, 23.9 ± 3.9; DMAG 2 mg/kg, 15.1 ± 1.2; DMAG 4 mg/kg, 4.2 ± 1.2; P < 0.05 vs. vehicle; not shown).

Figure 5

DMAG therapy alters the size and composition of atherosclerotic plaques in diabetic mice. A: Representative images of Oil Red O/hematoxylin-stained aortic root sections from nondiabetic control and diabetic apoE−/− mice after 10 weeks of treatment with vehicle or DMAG (2 and 4 mg/kg). B: Average of maximal lesion area in each group. C: Extent of atherosclerotic lesions throughout the studied region in diabetic mice (vehicle, white circles; DMAG 2 mg/kg, white squares; DMAG 4 mg/kg, black squares). D and E: Representative micrographs and quantification of macrophages (MOMA-2) (D) and T cells (CD3) (E) in the atherosclerotic plaques of diabetic mice. F: Assessment of plaque stability by the detection of collagen (picrosirius red staining) and VSMC (α-actin immunofluorescence) in diabetic mouse aortas. Representative images and quantification of collagen-to-lipid ratio (picrosirius red area/Oil Red O area) and VSMC-to-macrophage ratio (α-actin area/MOMA-2 area) are shown. Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05 and **P < 0.01 vs. control-vehicle group; †P < 0.05, ††P < 0.01, and †††P < 0.001 vs. diabetes-vehicle group. Scale bars: 200 μm (A) and 50 μm (DF). Dashed lines, atheroma plaques. L, lumen.

Figure 5

DMAG therapy alters the size and composition of atherosclerotic plaques in diabetic mice. A: Representative images of Oil Red O/hematoxylin-stained aortic root sections from nondiabetic control and diabetic apoE−/− mice after 10 weeks of treatment with vehicle or DMAG (2 and 4 mg/kg). B: Average of maximal lesion area in each group. C: Extent of atherosclerotic lesions throughout the studied region in diabetic mice (vehicle, white circles; DMAG 2 mg/kg, white squares; DMAG 4 mg/kg, black squares). D and E: Representative micrographs and quantification of macrophages (MOMA-2) (D) and T cells (CD3) (E) in the atherosclerotic plaques of diabetic mice. F: Assessment of plaque stability by the detection of collagen (picrosirius red staining) and VSMC (α-actin immunofluorescence) in diabetic mouse aortas. Representative images and quantification of collagen-to-lipid ratio (picrosirius red area/Oil Red O area) and VSMC-to-macrophage ratio (α-actin area/MOMA-2 area) are shown. Vertical striped bars indicate control-vehicle mice (n = 4); hatched bars, control-DMAG 4 mg/kg (n = 3); white bars, diabetes-vehicle (n = 9); gray bars, diabetes-DMAG 2 mg/kg (n = 9); black bars, diabetes-DMAG 4 mg/kg (n = 6). Data are mean ± SEM. *P < 0.05 and **P < 0.01 vs. control-vehicle group; †P < 0.05, ††P < 0.01, and †††P < 0.001 vs. diabetes-vehicle group. Scale bars: 200 μm (A) and 50 μm (DF). Dashed lines, atheroma plaques. L, lumen.

Close modal

Accumulation of monocytes/macrophages (MOMA-2) (Fig. 5D) and T cells (CD3) (Fig. 5E) within the atherosclerotic plaques of diabetic mice was reduced by DMAG treatment, with significant effect at high dose (% inhibition vs. vehicle: 58 ± 9 and 56 ± 13, respectively; P < 0.02). Furthermore, analysis of collagen (picrosirius red staining) and VSMC (α-actin immunofluorescence) content in atherosclerotic plaques from DMAG-treated diabetic mice showed a more stable phenotype, with significant raises in the relative proportions of collagen to lipids and VSMCs to macrophages when compared with vehicle-treated diabetic mice (Fig. 5F).

The anti-inflammatory effect of DMAG in diabetic atherosclerosis was also evidenced by a marked decrease in the activation of NF-κB (Fig. 6A) and the expression of CCL2 (Fig. 6B), CCL5 (Fig. 6C), and TNF-α (Fig. 6D) in plaques of DMAG-treated compared with vehicle-treated mice. In addition, quantitative real-time PCR analysis on RNA from aorta presented notable reductions in gene expression of CCL2, CCL5, and TNF-α as a result of DMAG treatment (Fig. 6E). Furthermore, DMAG treatment resulted in a dose-dependent induction of HSP70 mRNA expression in mouse aorta (Fig. 6E).

Figure 6

Reduced inflammation in atherosclerotic plaques of DMAG-treated mice. A: In situ detection of activated NF-κB by Southwestern histochemistry in aortic sections of diabetic apoE−/− mice. Representative images (scale bar, 20 μm; arrows, positive staining) and quantification of positive cells per lesion area. B–D: Immunoperoxidase images (scale bar, 50 μm; L, lumen) of CCL2 (B), CCL5 (C), and TNF-α (D) staining in diabetic mouse aortas and quantification of positive staining per lesion area. E: Real-time PCR analysis of the indicated genes in aortic tissue. Values normalized by 18S are expressed in arbitrary units (a.u.). White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are the mean ± SEM (six to nine animals per group). *P < 0.05 and **P < 0.01 vs. diabetes-vehicle group.

Figure 6

Reduced inflammation in atherosclerotic plaques of DMAG-treated mice. A: In situ detection of activated NF-κB by Southwestern histochemistry in aortic sections of diabetic apoE−/− mice. Representative images (scale bar, 20 μm; arrows, positive staining) and quantification of positive cells per lesion area. B–D: Immunoperoxidase images (scale bar, 50 μm; L, lumen) of CCL2 (B), CCL5 (C), and TNF-α (D) staining in diabetic mouse aortas and quantification of positive staining per lesion area. E: Real-time PCR analysis of the indicated genes in aortic tissue. Values normalized by 18S are expressed in arbitrary units (a.u.). White bars indicate diabetes-vehicle mice; gray bars, diabetes-DMAG 2 mg/kg; black bars, diabetes-DMAG 4 mg/kg. Data are the mean ± SEM (six to nine animals per group). *P < 0.05 and **P < 0.01 vs. diabetes-vehicle group.

Close modal

In Vitro Analysis of DMAG in Renal and Vascular Cells

The in vitro effects of DMAG on NF-κB and STAT activation and target gene expression were investigated in renal cells, vascular endothelial cells, and macrophages stimulated under hyperglycemic (HG) or proinflammatory (IL-6 + IFN-γ) conditions, in an attempt to mimic the diabetic milieu.

In the tubuloepithelial MCT cell line and primary macrophages, DMAG prevented HG-induced NF-κB activation, as evidenced by increased protein levels of inhibitory IκBα subunit (Fig. 7A). Similarly, DMAG pretreatment inhibited the degradation of IκBα in cytokine-stimulated MCT (Supplementary Fig. 3A), whereas no effect was observed with DMAG alone. Confocal microscopy experiments further revealed that DMAG reduced the nuclear translocation of the p65 subunit in vascular endothelial cells exposed to HG (Fig. 7B). We also observed that DMAG prevented the phosphorylation of STAT1 and STAT3 proteins in both HG-stimulated macrophages (Fig. 7C) and cytokine-stimulated renal cells (Fig. 7D and Supplementary Fig. 3B). Cell viability remained unaffected under these experimental conditions (Supplementary Fig. 3C).

Figure 7

In vitro effects of DMAG treatment in renal and vascular cells. Quiescent cells were pretreated with DMAG (50 nmol/L, 4 h) prior to stimulation with either cytokines (IL-6 102 units/mL and IFN-γ 103 units/mL) or HG (d-glucose 30 mmol/L) at different time points. A: Western blot analysis in total cell extracts from MCT and macrophages (MΦ), showing that DMAG prevents IκBα degradation induced by 6 h of HG stimulation. B: Representative confocal images of p65 NF-κB subunit localization in endothelial cells and quantification of p65-positive nuclei after 2 h of HG stimulation. Western blot analyses of P-STAT1/P-STAT3 in total cell extracts from HG-stimulated macrophages (6 h) (C) and cytokine-stimulated MC and MCT (1 h) (D). E: Real-time PCR analysis of Ccl2 and type I collagen (Col I) gene expression in MC and MCT stimulated with cytokines at 8 h. Values were normalized to 18S. F: Transwell cell migration assay of RAW 264.7 macrophages to cytokine-stimulated renal MC and MCT. G: Real-time PCR analysis of Ccl2 gene expression in MCT cells transfected (20–30 nmol/L, 24 h) with either scrambled (scr) or specific siRNA for HSP70 (si70) and stimulated overnight with cytokines. A, C, and D: Representative immunoblots and summary of normalized densitometric quantification are shown. E and G: Real-time PCR data were normalized to 18S. Values represent the mean ± SEM of three to six independent experiments. White bars indicate basal condition; black bars, stimulus (cytokine or HG); gray bars, DMAG+stimulus; and hatched bars, DMAG alone. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. basal; †P < 0.05 and ††P < 0.01 vs. stimulus.

Figure 7

In vitro effects of DMAG treatment in renal and vascular cells. Quiescent cells were pretreated with DMAG (50 nmol/L, 4 h) prior to stimulation with either cytokines (IL-6 102 units/mL and IFN-γ 103 units/mL) or HG (d-glucose 30 mmol/L) at different time points. A: Western blot analysis in total cell extracts from MCT and macrophages (MΦ), showing that DMAG prevents IκBα degradation induced by 6 h of HG stimulation. B: Representative confocal images of p65 NF-κB subunit localization in endothelial cells and quantification of p65-positive nuclei after 2 h of HG stimulation. Western blot analyses of P-STAT1/P-STAT3 in total cell extracts from HG-stimulated macrophages (6 h) (C) and cytokine-stimulated MC and MCT (1 h) (D). E: Real-time PCR analysis of Ccl2 and type I collagen (Col I) gene expression in MC and MCT stimulated with cytokines at 8 h. Values were normalized to 18S. F: Transwell cell migration assay of RAW 264.7 macrophages to cytokine-stimulated renal MC and MCT. G: Real-time PCR analysis of Ccl2 gene expression in MCT cells transfected (20–30 nmol/L, 24 h) with either scrambled (scr) or specific siRNA for HSP70 (si70) and stimulated overnight with cytokines. A, C, and D: Representative immunoblots and summary of normalized densitometric quantification are shown. E and G: Real-time PCR data were normalized to 18S. Values represent the mean ± SEM of three to six independent experiments. White bars indicate basal condition; black bars, stimulus (cytokine or HG); gray bars, DMAG+stimulus; and hatched bars, DMAG alone. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. basal; †P < 0.05 and ††P < 0.01 vs. stimulus.

Close modal

We next examined the role of HSP90 inhibition on the expression of NF-κB/STAT-dependent genes (e.g., CCL2 and type I collagen) induced by hyperglycemia or inflammation. In cytokine-stimulated renal cells, DMAG pretreatment resulted in a significantly reduced mRNA expression of CCL2 and type I collagen (Fig. 7E). DMAG also prevented CCL2 protein secretion (Supplementary Fig. 3E) and led to a lower macrophage chemoattractant capacity of renal cells, as determined by a migration assay in a coculture system (% inhibition vs. cytokines: 56 ± 2 and 78 ± 4, respectively; P < 0.02) (Fig. 7F). Similarly, DMAG was also able to prevent HG-induced CCL2 expression in MCT, macrophages, and endothelial cells (Supplementary Fig. 3D).

The involvement of HSP70 in DMAG-mediated cellular effects was further explored by genetic silencing in renal cells. First, we observed that DMAG dose-dependently increased both gene (Supplementary Fig. 4A) and protein (Supplementary Fig. 4B) expression of HSP family members, predominantly HSP70, and to a lesser degree HSP32 and HSP27. In addition, cell transfection with HSP70 siRNA markedly reduced HSP70 expression, without affecting HSP90 levels (Supplementary Fig. 5). We also observed that HSP70 silencing increased the cellular susceptibility to inflammatory stimulation, as demonstrated by a 2.5-fold higher Ccl2 mRNA expression in HSP70-silenced cells compared with cells transfected with a control scrambled siRNA (Fig. 7G). Remarkably, DMAG was not able to significantly inhibit the cytokine-dependent Ccl2 mRNA expression in HSP70-silenced cells (Fig. 7G), providing evidence that the beneficial effect associated with HSP90 inhibition is partially due to the induction of HSP70 expression.

Alterations in cellular homeostasis, signaling pathways, and gene expression operate concurrently to develop diabetic vascular derangements, such as nephropathy and atherosclerosis. Diabetes is associated with defects in HSP function mediated by either transcriptional or posttranslational mechanisms that regulate cellular levels of HSP in a tissue-specific manner, and such impairments contribute to diabetes complications, thus resulting in a vicious cycle (31,32). The role of HSP90 extends beyond heat shock response owing to its chaperoning function to fold and stabilize many client proteins involved in cell proliferation, differentiation, apoptosis, and inflammation, and is therefore proposed as an important target in immunity and inflammation (4). In this report, we demonstrate that pharmacological HSP90 inhibition ameliorates diabetes-associated renal damage and atheroprogression through the attenuation of cellular processes regulated by NF-κB and STAT signaling pathways, therefore indicating that HSP90 is a therapeutic target in diabetes complications.

HSP90 inhibitors such as geldanamycin and its derivatives targeting HSP90 N terminus and blocking its ATPase activity have emerged as a clinically relevant strategy in cancer and are also promising drugs for immune and inflammatory diseases, including diabetes (14). Our results in a well-established mouse model of combined hyperglycemia and hyperlipidemia (STZ-induced diabetic apoE−/− mice) demonstrate that HSP90 inhibition by DMAG effectively improved renal function, as evidenced by reduced serum creatinine and urine albumin-to-creatinine ratio, without overt side effects and toxicity. Unlike a previous study that shows reversed hyperglycemia by HSP90 inhibitors in diet-induced obese mice (19), we observed that DMAG administration did not affect the metabolic severity of diabetes, with no changes in hyperglycemia, lipid profile, and body weight. Of note, DMAG treatment attenuated the structural pathologic changes in the diabetic kidney, including glomerular hyperplasia and mesangial matrix expansion, as well as tubular atrophy, interstitial fibrosis, and inflammation, hallmarks of end-stage renal failure. In line with the recently reported effect of HSP90 inhibitor on high-fat diet–induced renal failure in diabetes (20), our work adds differences in treatment dose and duration and study samples and provides mechanistic details about the renoprotective action of DMAG in diabetic mice.

Diabetes-associated inflammation promotes a progressive accumulation of kidney leukocytes (33) that contribute to the development and progression of diabetic nephropathy either by direct interaction with the renal cells or by releasing cytokines and growth factors involved in cell proliferation, migration, and extracellular matrix production (34,35). Besides renal histological improvement, we also found a marked anti-inflammatory and antifibrotic effect of HSP90 inhibition. In fact, DMAG-treated mice exhibited reduced macrophage and T-cell infiltration along with a decreased expression of proinflammatory genes (TNF-α, CCL2, and CCL5). Previous reports indicated that HSP90 inhibitors prevent inflammatory gene expression in different cell types, including cancer cells (36), leukocytes (17,37), and renal and vascular cells (15,17). In line with this, our studies in cultured mesangial and tubular cells mimicking hyperglycemic and inflammatory diabetic conditions demonstrated that DMAG inhibited the gene expression and protein secretion of CCL2 and also mitigated macrophage chemotaxis, further substantiating the in vivo findings. Moreover, we observed that DMAG protected from the development of renal fibrosis in diabetic mice, with significant reductions in the expression of extracellular matrix proteins and profibrotic factor TGF-β. These observations were further confirmed in vitro by analyzing the expression of an extracellular matrix protein (type I collagen) in renal cells. In agreement with the previously reported ability of DMAG to inhibit TGF-β signaling in vitro (38) and to reduce renal fibrosis in mice (39), our findings point toward HSP90 as an attractive target to attenuate renal inflammation and fibrosis in diabetic nephropathy.

Our data highlight a potential use of HSP90 inhibitors for prevention of diabetes-associated atherosclerosis. Indeed, DMAG administration dose-dependently reduced the size and extension of atherosclerotic lesions in STZ-induced diabetic apoE−/− mice and also altered plaque composition without any incidence in lipid profile. Increased HSP90 expression has been linked to vulnerability of human advanced atherosclerotic lesions, and therefore it has been suggested as a promising target to stabilize atheroma plaques through the modulation of inflammation and oxidative stress (17,18,40). Our observations in diabetic mice revealed that DMAG treatment promoted a less inflamed, more stable phenotype characterized by lower content of lipids, macrophages, and T cells and higher collagen and VSMC content. Additionally, atherosclerotic lesions of DMAG-treated mice displayed a lower amount of NF-κB–activated cells together with a downregulation in the aortic expression of cytokines and chemokines involved in proatherogenic activation of vessel cells. Our in vitro studies performed in macrophages and endothelial cells, two of the cell types involved in atherosclerosis progression, demonstrated that DMAG impaired inflammatory gene expression in a hyperglycemic environment. These results are in agreement with the recently reported antimigratory effect of HSP90 inhibitor during atherogenesis (41).

Our study provides in vivo and in vitro evidence that DMAG inhibits NF-κB and STAT, two representative HSP90-regulated pathways that control key pathological mechanisms involved in diabetes complications. Owing to its chaperoning function, HSP90 regulates the stability and the activation of NF-κB and STAT signaling pathways by direct association and stabilization of their respective activating kinases, IKK (42) and JAK2 (43). Accordingly, the efficacy of HSP90 inhibitors in animal models has been linked to the disruption of the IKK complex and JAK2 protein stability, further inhibiting downstream transcription factors such as p50/p65 NF-κB (44) and STAT1/STAT3/STAT5 (45). Dysregulated NF-κB and STAT pathways contribute to diabetic nephropathy and atherosclerosis by inducing the transcription of many genes associated with inflammation, renal fibrosis (4547), and the proatherogenic state (23,48). We have previously tested a number of potential novel approaches (e.g., kinase inhibitors, permeable peptides, and gene therapy) to tackle diabetic nephropathy and atherosclerosis by separately inhibiting the STAT and NF-κB pathways (23,26,28,29). Here, we demonstrated that DMAG treatment resulted in combined inhibition of the NF-κB and STAT pathways and subsequent downregulation of their inducible genes (e.g., cytokines, chemokines, and extracellular matrix proteins) both in diabetic mice and in vitro. Given that HSP90 inhibitors degrade many different client proteins, it is likely that the protective effects of DMAG on diabetic mice may result from inhibition of multiple target proteins in renal and vascular cells.

Another established effect of HSP90 inhibition is the induction of HS70 expression through the activation of HSF1, resulting from HSP90-HSF1 complex disruption (14,19). HSP70 is likely to confer protection against disturbed metabolic homeostasis via multiple modes of action, including reduced inflammation (49) and improved insulin sensitivity (19). In agreement with previous descriptions (38,50), we demonstrate that DMAG treatment dose-dependently induced HSP70 expression in kidneys and aorta of diabetic mice and in cultured cells. Interestingly, we also observed that HSP70 gene silencing enhanced cytokine responsiveness of cells and partially attenuated the inhibitory effect of DMAG, thus suggesting that DMAG-mediated HSP70 induction contributes to cytoprotection and counterbalance of diabetes-induced cell damage.

Collectively, our experimental data provide the first evidence that HSP90 inhibition simultaneously ameliorates diabetes-associated nephropathy and atherosclerosis through the induction of protective HSP70 and the attenuation of the NF-κB and STAT pathways, thus resulting in improved renal inflammation and fibrosis and stabilization of atherosclerotic plaques. Therefore, we propose HSP90 inhibition as a novel intervention to limit diabetes-associated complications.

Acknowledgments. The authors thank Dr. M.D. Sanchez-Niño (Nephrology Department, IIS-Fundacion Jimenez Diaz) for helpful advice on the siRNA experiments.

Funding. This work was supported by grants from the Spanish Ministry of Economy and Competitiveness (SAF2012-38830 and SAF2010-21852), the Spanish Ministry of Health (FIS PI14/00386 and DiabetesCancerConnect PIE13/00051), the Spanish Societies of Nephrology and Atherosclerosis, and the Iñigo Alvarez de Toledo Renal Foundation. I.L. was supported by a postdoctoral fellowship from FIS (Sara Borrell program).

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

Author Contributions. I.L. designed experiments, researched and analyzed data, and wrote the manuscript. A.O. and C.R. researched and analyzed data and critically revised the manuscript. B.M., J.M.-M., and J.B. researched, analyzed, or interpreted data. J.E. and J.-L.M.-V. critically revised the manuscript for important intellectual content. C.G.-G. conceived and designed the study, analyzed data, and drafted and reviewed the manuscript. C.G.-G. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Nathan
DM
.
Long-term complications of diabetes mellitus
.
N Engl J Med
1993
;
328
:
1676
1685
[PubMed]
2.
Rask-Madsen
C
,
King
GL
.
Vascular complications of diabetes: mechanisms of injury and protective factors
.
Cell Metab
2013
;
17
:
20
33
[PubMed]
3.
Fernandez-Fernandez
B
,
Ortiz
A
,
Gomez-Guerrero
C
,
Egido
J
.
Therapeutic approaches to diabetic nephropathy--beyond the RAS
.
Nat Rev Nephrol
2014
;
10
:
325
346
[PubMed]
4.
Taipale
M
,
Jarosz
DF
,
Lindquist
S
.
HSP90 at the hub of protein homeostasis: emerging mechanistic insights
.
Nat Rev Mol Cell Biol
2010
;
11
:
515
528
[PubMed]
5.
Kurucz
I
,
Morva
A
,
Vaag
A
, et al
.
Decreased expression of heat shock protein 72 in skeletal muscle of patients with type 2 diabetes correlates with insulin resistance
.
Diabetes
2002
;
51
:
1102
1109
[PubMed]
6.
Tessari
P
,
Puricelli
L
,
Iori
E
, et al
.
Altered chaperone and protein turnover regulators expression in cultured skin fibroblasts from type 1 diabetes mellitus with nephropathy
.
J Proteome Res
2007
;
6
:
976
986
[PubMed]
7.
Barutta
F
,
Pinach
S
,
Giunti
S
, et al
.
Heat shock protein expression in diabetic nephropathy
.
Am J Physiol Renal Physiol
2008
;
295
:
F1817
F1824
[PubMed]
8.
Caplan
AJ
,
Mandal
AK
,
Theodoraki
MA
.
Molecular chaperones and protein kinase quality control
.
Trends Cell Biol
2007
;
17
:
87
92
[PubMed]
9.
Li
J
,
Buchner
J
.
Structure, function and regulation of the hsp90 machinery
.
Biomed J
2013
;
36
:
106
117
[PubMed]
10.
Lawrence
T
.
The nuclear factor NF-kappaB pathway in inflammation
.
Cold Spring Harb Perspect Biol
2009
;
1
:
a001651
[PubMed]
11.
Adhikari
N
,
Charles
N
,
Lehmann
U
,
Hall
JL
.
Transcription factor and kinase-mediated signaling in atherosclerosis and vascular injury
.
Curr Atheroscler Rep
2006
;
8
:
252
260
[PubMed]
12.
Rice
JW
,
Veal
JM
,
Fadden
RP
, et al
.
Small molecule inhibitors of Hsp90 potently affect inflammatory disease pathways and exhibit activity in models of rheumatoid arthritis
.
Arthritis Rheum
2008
;
58
:
3765
3775
[PubMed]
13.
Kakeda
M
,
Arock
M
,
Schlapbach
C
,
Yawalkar
N
.
Increased expression of heat shock protein 90 in keratinocytes and mast cells in patients with psoriasis
.
J Am Acad Dermatol
2014
;
70
:
683
690.e1
[PubMed]
14.
Hong
DS
,
Banerji
U
,
Tavana
B
,
George
GC
,
Aaron
J
,
Kurzrock
R
.
Targeting the molecular chaperone heat shock protein 90 (HSP90): lessons learned and future directions
.
Cancer Treat Rev
2013
;
39
:
375
387
[PubMed]
15.
Shimp
SK
 3rd
,
Chafin
CB
,
Regna
NL
, et al
.
Heat shock protein 90 inhibition by 17-DMAG lessens disease in the MRL/lpr mouse model of systemic lupus erythematosus
.
Cell Mol Immunol
2012
;
9
:
255
266
[PubMed]
16.
Ambade
A
,
Catalano
D
,
Lim
A
,
Mandrekar
P
.
Inhibition of heat shock protein (molecular weight 90 kDa) attenuates proinflammatory cytokines and prevents lipopolysaccharide-induced liver injury in mice
.
Hepatology
2012
;
55
:
1585
1595
[PubMed]
17.
Madrigal-Matute
J
,
López-Franco
O
,
Blanco-Colio
LM
, et al
.
Heat shock protein 90 inhibitors attenuate inflammatory responses in atherosclerosis
.
Cardiovasc Res
2010
;
86
:
330
337
[PubMed]
18.
Madrigal-Matute
J
,
Fernandez-Garcia
CE
,
Gomez-Guerrero
C
, et al
.
HSP90 inhibition by 17-DMAG attenuates oxidative stress in experimental atherosclerosis
.
Cardiovasc Res
2012
;
95
:
116
123
[PubMed]
19.
Lee
JH
,
Gao
J
,
Kosinski
PA
, et al
.
Heat shock protein 90 (HSP90) inhibitors activate the heat shock factor 1 (HSF1) stress response pathway and improve glucose regulation in diabetic mice
.
Biochem Biophys Res Commun
2013
;
430
:
1109
1113
[PubMed]
20.
Zhang
HM
,
Dang
H
,
Kamat
A
,
Yeh
CK
,
Zhang
BX
.
Geldanamycin derivative ameliorates high fat diet-induced renal failure in diabetes
.
PLoS One
2012
;
7
:
e32746
[PubMed]
21.
Urban
MJ
,
Pan
P
,
Farmer
KL
,
Zhao
H
,
Blagg
BS
,
Dobrowsky
RT
.
Modulating molecular chaperones improves sensory fiber recovery and mitochondrial function in diabetic peripheral neuropathy
.
Exp Neurol
2012
;
235
:
388
396
[PubMed]
22.
Lopez-Parra
V
,
Mallavia
B
,
Lopez-Franco
O
, et al
.
Fcγ receptor deficiency attenuates diabetic nephropathy
.
J Am Soc Nephrol
2012
;
23
:
1518
1527
[PubMed]
23.
Recio
C
,
Oguiza
A
,
Lazaro
I
,
Mallavia
B
,
Egido
J
,
Gomez-Guerrero
C
.
Suppressor of cytokine signaling 1-derived peptide inhibits Janus kinase/signal transducers and activators of transcription pathway and improves inflammation and atherosclerosis in diabetic mice
.
Arterioscler Thromb Vasc Biol
2014
;
34
:
1953
1960
[PubMed]
24.
Chew
P
,
Yuen
DY
,
Stefanovic
N
, et al
.
Antiatherosclerotic and renoprotective effects of ebselen in the diabetic apolipoprotein E/GPx1-double knockout mouse
.
Diabetes
2010
;
59
:
3198
3207
[PubMed]
25.
Engler
FA
,
Zheng
B
,
Balthasar
JP
.
Investigation of the influence of nephropathy on monoclonal antibody disposition: a pharmacokinetic study in a mouse model of diabetic nephropathy
.
Pharm Res
2014
;
31
:
1185
1193
[PubMed]
26.
López-Franco
O
,
Hernández-Vargas
P
,
Ortiz-Muñoz
G
, et al
.
Parthenolide modulates the NF-kappaB-mediated inflammatory responses in experimental atherosclerosis
.
Arterioscler Thromb Vasc Biol
2006
;
26
:
1864
1870
[PubMed]
27.
Ortiz-Muñoz
G
,
Martin-Ventura
JL
,
Hernandez-Vargas
P
, et al
.
Suppressors of cytokine signaling modulate JAK/STAT-mediated cell responses during atherosclerosis
.
Arterioscler Thromb Vasc Biol
2009
;
29
:
525
531
[PubMed]
28.
Mallavia
B
,
Recio
C
,
Oguiza
A
, et al
.
Peptide inhibitor of NF-κB translocation ameliorates experimental atherosclerosis
.
Am J Pathol
2013
;
182
:
1910
1921
[PubMed]
29.
Ortiz-Muñoz
G
,
Lopez-Parra
V
,
Lopez-Franco
O
, et al
.
Suppressors of cytokine signaling abrogate diabetic nephropathy
.
J Am Soc Nephrol
2010
;
21
:
763
772
[PubMed]
30.
Hsueh
W
,
Abel
ED
,
Breslow
JL
, et al
.
Recipes for creating animal models of diabetic cardiovascular disease
.
Circ Res
2007
;
100
:
1415
1427
[PubMed]
31.
Yamagishi
N
,
Nakayama
K
,
Wakatsuki
T
,
Hatayama
T
.
Characteristic changes of stress protein expression in streptozotocin-induced diabetic rats
.
Life Sci
2001
;
69
:
2603
2609
[PubMed]
32.
Atalay
M
,
Oksala
N
,
Lappalainen
J
,
Laaksonen
DE
,
Sen
CK
,
Roy
S
.
Heat shock proteins in diabetes and wound healing
.
Curr Protein Pept Sci
2009
;
10
:
85
95
[PubMed]
33.
Galkina
E
,
Ley
K
.
Leukocyte recruitment and vascular injury in diabetic nephropathy
.
J Am Soc Nephrol
2006
;
17
:
368
377
[PubMed]
34.
Chow
FY
,
Nikolic-Paterson
DJ
,
Ozols
E
,
Atkins
RC
,
Rollin
BJ
,
Tesch
GH
.
Monocyte chemoattractant protein-1 promotes the development of diabetic renal injury in streptozotocin-treated mice
.
Kidney Int
2006
;
69
:
73
80
[PubMed]
35.
Navarro-González
JF
,
Mora-Fernández
C
.
The role of inflammatory cytokines in diabetic nephropathy
.
J Am Soc Nephrol
2008
;
19
:
433
442
[PubMed]
36.
Kolosenko
I
,
Grander
D
,
Tamm
KP
.
IL-6 activated JAK/STAT3 pathway and sensitivity to Hsp90 inhibitors in multiple myeloma
.
Curr Med Chem
2014
;
21
:
3042
3047
[PubMed]
37.
Bae
J
,
Munshi
A
,
Li
C
, et al
.
Heat shock protein 90 is critical for regulation of phenotype and functional activity of human T lymphocytes and NK cells
.
J Immunol
2013
;
190
:
1360
1371
[PubMed]
38.
Yun
CH
,
Yoon
SY
,
Nguyen
TT
, et al
.
Geldanamycin inhibits TGF-beta signaling through induction of Hsp70
.
Arch Biochem Biophys
2010
;
495
:
8
13
[PubMed]
39.
Noh
H
,
Kim
HJ
,
Yu
MR
, et al
.
Heat shock protein 90 inhibitor attenuates renal fibrosis through degradation of transforming growth factor-β type II receptor
.
Lab Invest
2012
;
92
:
1583
1596
[PubMed]
40.
Businaro
R
,
Profumo
E
,
Tagliani
A
, et al
.
Heat-shock protein 90: a novel autoantigen in human carotid atherosclerosis
.
Atherosclerosis
2009
;
207
:
74
83
[PubMed]
41.
Kim
J
,
Jang
SW
,
Park
E
,
Oh
M
,
Park
S
,
Ko
J
.
The role of heat shock protein 90 in migration and proliferation of vascular smooth muscle cells in the development of atherosclerosis
.
J Mol Cell Cardiol
2014
;
72
:
157
167
[PubMed]
42.
Salminen
A
,
Paimela
T
,
Suuronen
T
,
Kaarniranta
K
.
Innate immunity meets with cellular stress at the IKK complex: regulation of the IKK complex by HSP70 and HSP90
.
Immunol Lett
2008
;
117
:
9
15
[PubMed]
43.
Marubayashi
S
,
Koppikar
P
,
Taldone
T
, et al
.
HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans
.
J Clin Invest
2010
;
120
:
3578
3593
[PubMed]
44.
Hertlein
E
,
Wagner
AJ
,
Jones
J
, et al
.
17-DMAG targets the nuclear factor-kappaB family of proteins to induce apoptosis in chronic lymphocytic leukemia: clinical implications of HSP90 inhibition
.
Blood
2010
;
116
:
45
53
[PubMed]
45.
Marrero
MB
,
Banes-Berceli
AK
,
Stern
DM
,
Eaton
DC
.
Role of the JAK/STAT signaling pathway in diabetic nephropathy
.
Am J Physiol Renal Physiol
2006
;
290
:
F762
F768
[PubMed]
46.
Mezzano
S
,
Aros
C
,
Droguett
A
, et al
.
NF-kappaB activation and overexpression of regulated genes in human diabetic nephropathy
.
Nephrol Dial Transplant
2004
;
19
:
2505
2512
[PubMed]
47.
Brosius
FC
 3rd
.
New insights into the mechanisms of fibrosis and sclerosis in diabetic nephropathy
.
Rev Endocr Metab Disord
2008
;
9
:
245
254
[PubMed]
48.
Baker
RG
,
Hayden
MS
,
Ghosh
S
.
NF-κB, inflammation, and metabolic disease
.
Cell Metab
2011
;
13
:
11
22
[PubMed]
49.
Chung
J
,
Nguyen
AK
,
Henstridge
DC
, et al
.
HSP72 protects against obesity-induced insulin resistance
.
Proc Natl Acad Sci U S A
2008
;
105
:
1739
1744
[PubMed]
50.
Harrison
EM
,
Sharpe
E
,
Bellamy
CO
, et al
.
Heat shock protein 90-binding agents protect renal cells from oxidative stress and reduce kidney ischemia-reperfusion injury
.
Am J Physiol Renal Physiol
2008
;
295
:
F397
F405
[PubMed]

Supplementary data