Calcium dobesilate may alleviate diabetes‑induced endothelial dysfunction and inflammation

Yijun Zhou, Jiangzi Yuan, Chaojun Qi, Xinghua Shao, Shan Mou, Zhaohui Ni, Yijun Zhou, Jiangzi Yuan, Chaojun Qi, Xinghua Shao, Shan Mou, Zhaohui Ni

Abstract

Diabetic kidney disease (DKD) is a leading cause of end‑stage renal disease. However, the pathogenesis of DKD remains unclear, and no effective treatments for the disease are available. Thus, there is an urgent need to elucidate the pathogenic mechanisms of DKD and to develop more effective therapies for this disease. Human umbilical vein endothelial cells (HUVECs) were cultured using different D‑glucose concentrations to determine the effect of high glucose (HG) on the cells. Alternatively, HUVECs were incubated with 100 µmol/l calcium dobesilate (CaD) to detect its effects. The authors subsequently measured HUVEC proliferation via cell counting kit‑8 assays. In addition, HUVEC angiogenesis was investigated via migration assays and fluorescein isothiocyanate (FITC)‑labelled bovine serum albumin (BSA) permeability assays. The content or distribution of markers of endothelial dysfunction [vascular endothelial growth factor (VEGF), VEGF receptor (R) and endocan) or inflammation [intercellular adhesion molecule (ICAM)‑1, monocyte chemotactic protein (MCP)‑1 and pentraxin‑related protein (PTX3)] was evaluated via reverse transcription‑quantitative polymerase chain reaction and western blotting. HG treatment induced increased in VEGF, VEGFR, endocan, ICAM‑1, MCP‑1 and PTX3 mRNA and protein expression in HUVECs. HG treatment for 24 to 48 h increased cell proliferation in a time‑dependent manner, but the cell proliferation rate was decreased at 72 h of HG treatment. Conversely, CaD inhibited abnormal cell proliferation. HG treatment also significantly enhanced HVUEC migration compared to the control treatment. In contrast, CaD treatment partially inhibited HUVEC migration compared to HG exposure. HG‑treated HUVECs exhibited increased FITC‑BSA permeability compared to control cells cultured in medium alone; however, CaD application prevented the HG‑induced increase in FITC‑BSA permeability and suppressed HG‑induced overexpression of endothelial markers (VEGF, VEGFR‑2, endocan) and inflammation markers (ICAM‑1, MCP‑1, PTX3) in HUVECs. CaD has angioprotective properties and protects endothelial cells partly by ameliorating HG‑induced inflammation. The current results demonstrated the potential applicability of CaD to the treatment of diabetic nephropathy, particularly during the early stages of this disease.

Figures

Figure 1.
Figure 1.
mRNA expression of VEGF, VEGFR, endocan, ICAM, MCP-1 and PTX3 mRNA in high glucose conditions in human umbilical vein endothelial cells. mRNA expression gradually increased. *P

Figure 2.

Western blot analysis of VEGF,…

Figure 2.

Western blot analysis of VEGF, VEGFR, endocan, ICAM, MCP-1 and PTX3 protein expression.…

Figure 2.
Western blot analysis of VEGF, VEGFR, endocan, ICAM, MCP-1 and PTX3 protein expression. Expression gradually increased under high glucose conditions in human umbilical vein endothelial cells. *P

Figure 3.

Effects of different cell treatments…

Figure 3.

Effects of different cell treatments on HUVEC proliferation. Cell Counting kit-8 assay was…

Figure 3.
Effects of different cell treatments on HUVEC proliferation. Cell Counting kit-8 assay was performed to analysed effect on cell proliferation. HG (35 mmol/l) treatment induced increased cell proliferation at 24 and 48 h. CaD (100 µmol/l) application significantly attenuated abnormal HUVEC proliferation. *P

Figure 4.

CaD prevents migration of HUVECs,…

Figure 4.

CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell…

Figure 4.
CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell assay analysing migration of HUVECs following cell treatment. The stained cells were counted in 10 randomly chosen fields at ×200 magnification. *P&P<0.05 vs. mannitol group. CaD, calcium dobesilate; HG, high glucose; HUVECs, human umbilical vein endothelial cells.

Figure 5.

HG increased the permeability of…

Figure 5.

HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing…

Figure 5.
HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing permeability of HUVECs following cell treatment. *P&P<0.05 vs. mannitol group. HG, high glucose; HUVECs, human umbilical vein endothelial cells; CaD, calcium dobesilate.

Figure 6.

HG (35 mmol/l) increased the…

Figure 6.

HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a…

Figure 6.
HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the mRNA levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P

Figure 7.

HG (35 mmol/l) increased the…

Figure 7.

HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a…

Figure 7.
HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the protein levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P&P<0.05 vs. control group. HG, high glucose; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; ICAM, intercellular adhesion molecule; MCP-1, monocyte chemotactic protein 1; PTX3, pentraxin 3; CaD, calcium dobesilate; HUVECs, human umbilical vein endothelial cells.
All figures (7)
Similar articles
Cited by
References
    1. International Diabetes Federation, corp-author. www.diabetesatlas.org Diabetes Atlas. (7th edition) 2015
    1. Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, et al. Diabetic kidney disease: A report from an ADA Consensus Conference. Am J Kidney Dis. 2014;64:510–533. doi: 10.1053/j.ajkd.2014.08.001. - DOI - PubMed
    1. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015;308:F287–F297. doi: 10.1152/ajprenal.00533.2014. - DOI - PMC - PubMed
    1. Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Haematol Disord Drug Targets. 2014;14:22–33. doi: 10.2174/1871529X14666140401110841. - DOI - PMC - PubMed
    1. Navarro-González JF, Mora-Fernández C, de Fuentes M Muros, García-Pérez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7:327–340. doi: 10.1038/nrneph.2011.51. - DOI - PubMed
Show all 48 references
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2.
Figure 2.
Western blot analysis of VEGF, VEGFR, endocan, ICAM, MCP-1 and PTX3 protein expression. Expression gradually increased under high glucose conditions in human umbilical vein endothelial cells. *P

Figure 3.

Effects of different cell treatments…

Figure 3.

Effects of different cell treatments on HUVEC proliferation. Cell Counting kit-8 assay was…

Figure 3.
Effects of different cell treatments on HUVEC proliferation. Cell Counting kit-8 assay was performed to analysed effect on cell proliferation. HG (35 mmol/l) treatment induced increased cell proliferation at 24 and 48 h. CaD (100 µmol/l) application significantly attenuated abnormal HUVEC proliferation. *P

Figure 4.

CaD prevents migration of HUVECs,…

Figure 4.

CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell…

Figure 4.
CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell assay analysing migration of HUVECs following cell treatment. The stained cells were counted in 10 randomly chosen fields at ×200 magnification. *P&P<0.05 vs. mannitol group. CaD, calcium dobesilate; HG, high glucose; HUVECs, human umbilical vein endothelial cells.

Figure 5.

HG increased the permeability of…

Figure 5.

HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing…

Figure 5.
HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing permeability of HUVECs following cell treatment. *P&P<0.05 vs. mannitol group. HG, high glucose; HUVECs, human umbilical vein endothelial cells; CaD, calcium dobesilate.

Figure 6.

HG (35 mmol/l) increased the…

Figure 6.

HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a…

Figure 6.
HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the mRNA levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P

Figure 7.

HG (35 mmol/l) increased the…

Figure 7.

HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a…

Figure 7.
HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the protein levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P&P<0.05 vs. control group. HG, high glucose; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; ICAM, intercellular adhesion molecule; MCP-1, monocyte chemotactic protein 1; PTX3, pentraxin 3; CaD, calcium dobesilate; HUVECs, human umbilical vein endothelial cells.
All figures (7)
Similar articles
Cited by
References
    1. International Diabetes Federation, corp-author. www.diabetesatlas.org Diabetes Atlas. (7th edition) 2015
    1. Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, et al. Diabetic kidney disease: A report from an ADA Consensus Conference. Am J Kidney Dis. 2014;64:510–533. doi: 10.1053/j.ajkd.2014.08.001. - DOI - PubMed
    1. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015;308:F287–F297. doi: 10.1152/ajprenal.00533.2014. - DOI - PMC - PubMed
    1. Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Haematol Disord Drug Targets. 2014;14:22–33. doi: 10.2174/1871529X14666140401110841. - DOI - PMC - PubMed
    1. Navarro-González JF, Mora-Fernández C, de Fuentes M Muros, García-Pérez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7:327–340. doi: 10.1038/nrneph.2011.51. - DOI - PubMed
Show all 48 references
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3.
Figure 3.
Effects of different cell treatments on HUVEC proliferation. Cell Counting kit-8 assay was performed to analysed effect on cell proliferation. HG (35 mmol/l) treatment induced increased cell proliferation at 24 and 48 h. CaD (100 µmol/l) application significantly attenuated abnormal HUVEC proliferation. *P

Figure 4.

CaD prevents migration of HUVECs,…

Figure 4.

CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell…

Figure 4.
CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell assay analysing migration of HUVECs following cell treatment. The stained cells were counted in 10 randomly chosen fields at ×200 magnification. *P&P<0.05 vs. mannitol group. CaD, calcium dobesilate; HG, high glucose; HUVECs, human umbilical vein endothelial cells.

Figure 5.

HG increased the permeability of…

Figure 5.

HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing…

Figure 5.
HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing permeability of HUVECs following cell treatment. *P&P<0.05 vs. mannitol group. HG, high glucose; HUVECs, human umbilical vein endothelial cells; CaD, calcium dobesilate.

Figure 6.

HG (35 mmol/l) increased the…

Figure 6.

HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a…

Figure 6.
HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the mRNA levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P

Figure 7.

HG (35 mmol/l) increased the…

Figure 7.

HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a…

Figure 7.
HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the protein levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P&P<0.05 vs. control group. HG, high glucose; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; ICAM, intercellular adhesion molecule; MCP-1, monocyte chemotactic protein 1; PTX3, pentraxin 3; CaD, calcium dobesilate; HUVECs, human umbilical vein endothelial cells.
All figures (7)
Similar articles
Cited by
References
    1. International Diabetes Federation, corp-author. www.diabetesatlas.org Diabetes Atlas. (7th edition) 2015
    1. Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, et al. Diabetic kidney disease: A report from an ADA Consensus Conference. Am J Kidney Dis. 2014;64:510–533. doi: 10.1053/j.ajkd.2014.08.001. - DOI - PubMed
    1. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015;308:F287–F297. doi: 10.1152/ajprenal.00533.2014. - DOI - PMC - PubMed
    1. Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Haematol Disord Drug Targets. 2014;14:22–33. doi: 10.2174/1871529X14666140401110841. - DOI - PMC - PubMed
    1. Navarro-González JF, Mora-Fernández C, de Fuentes M Muros, García-Pérez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7:327–340. doi: 10.1038/nrneph.2011.51. - DOI - PubMed
Show all 48 references
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4.
Figure 4.
CaD prevents migration of HUVECs, which partially induced by diabetes or HG. Transwell assay analysing migration of HUVECs following cell treatment. The stained cells were counted in 10 randomly chosen fields at ×200 magnification. *P&P<0.05 vs. mannitol group. CaD, calcium dobesilate; HG, high glucose; HUVECs, human umbilical vein endothelial cells.
Figure 5.
Figure 5.
HG increased the permeability of HUVECs, which inhibited by CaD. Permeability assay analysing permeability of HUVECs following cell treatment. *P&P<0.05 vs. mannitol group. HG, high glucose; HUVECs, human umbilical vein endothelial cells; CaD, calcium dobesilate.
Figure 6.
Figure 6.
HG (35 mmol/l) increased the mRNA levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the mRNA levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P

Figure 7.

HG (35 mmol/l) increased the…

Figure 7.

HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a…

Figure 7.
HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the protein levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P&P<0.05 vs. control group. HG, high glucose; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; ICAM, intercellular adhesion molecule; MCP-1, monocyte chemotactic protein 1; PTX3, pentraxin 3; CaD, calcium dobesilate; HUVECs, human umbilical vein endothelial cells.
All figures (7)
Figure 7.
Figure 7.
HG (35 mmol/l) increased the protein levels of VEGF, VEGFR-2, endocan in a time-dependent manner from 12 to 48 h. However, the growth trend began to slow down in 72 h. 100 µmol/l CaD prevented the increase of VEGF, VEGFR-2 and endocan levels induced by HG. HG also increased the protein levels of ICAM, MCP-1 and PTX3 in a time-dependent manner from 12 to 72 h. 100 µmol/l CaD prevented the increase the expression of these inflammatory factors induced by HG. (A) HUVECs were incubated with control or experimental medium for 12 h. (B) HUVECs were incubated with control or experimental medium for 24 h. (C) HUVECs were incubated with control or experimental medium for 48 h. (D) HUVECs were incubated with control or experimental medium for 72 h. *P&P<0.05 vs. control group. HG, high glucose; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; ICAM, intercellular adhesion molecule; MCP-1, monocyte chemotactic protein 1; PTX3, pentraxin 3; CaD, calcium dobesilate; HUVECs, human umbilical vein endothelial cells.

References

    1. International Diabetes Federation, corp-author. Diabetes Atlas. (7th edition) 2015
    1. Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD, et al. Diabetic kidney disease: A report from an ADA Consensus Conference. Am J Kidney Dis. 2014;64:510–533. doi: 10.1053/j.ajkd.2014.08.001.
    1. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015;308:F287–F297. doi: 10.1152/ajprenal.00533.2014.
    1. Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Haematol Disord Drug Targets. 2014;14:22–33. doi: 10.2174/1871529X14666140401110841.
    1. Navarro-González JF, Mora-Fernández C, de Fuentes M Muros, García-Pérez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7:327–340. doi: 10.1038/nrneph.2011.51.
    1. Galkina E, Ley K. Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am Soc Nephrol. 2006;17:368–377. doi: 10.1681/ASN.2005080859.
    1. Sugimoto H, Shikata K, Hirata K, Akiyama K, Matsuda M, Kushiro M, Shikata Y, Miyatake N, Miyasaka M, Makino H. Increased expression of intercellular adhesion molecule-1 (ICAM-1) in diabetic rat glomeruli: Glomerular hyperfiltration is a potential mechanism of ICAM-1 upregulation. Diabetes. 1997;46:2075–2081. doi: 10.2337/diab.46.12.2075.
    1. Seron D, Cameron JS, Haskard DO. Expression of VCAM-1 in the normal and diseased kidney. Nephrol Dial Transplant. 1991;6:917–922. doi: 10.1093/ndt/6.12.917.
    1. Tejerina T, Ruiz E. Calcium dobesilate: Pharmacology and future approaches. Gen Pharmacol. 1998;31:357–360. doi: 10.1016/S0306-3623(98)00040-8.
    1. Javadzadeh A, Ghorbanihaghjo A, Adl FH, Andalib D, Khojasteh-Jafari H, Ghabili K. Calcium dobesilate reduces endothelin-1 and high-sensitivity C-reactive protein serum levels in patients with diabetic retinopathy. Mol Vis. 2013;19:62–68.
    1. Rabe E, Jaeger KA, Bulitta M, Pannier F. Calcium dobesilate in patients suffering from chronic venous insufficiency: A double-blind, placebo-controlled, clinical trial. Phlebology. 2011;26:162–168. doi: 10.1258/phleb.2010.010051.
    1. Hall JF. Modern management of hemorrhoidal disease. Gastroenterol Clin North Am. 2013;42:759–772. doi: 10.1016/j.gtc.2013.09.001.
    1. Zhang X. Therapeutic effects of calcium dobesilate on diabetic nephropathy mediated through reduction of expression of PAI-1. Exp Ther Med. 2013;5:295–299. doi: 10.3892/etm.2012.755.
    1. Jafarey M, Ashtiyani S Changizi, Najafi H. Calcium dobesilate for prevention of gentamicin-induced nephrotoxicity in rats. Iran J Kidney Dis. 2014;8:46–52.
    1. Pan SL, Guh JH, Huang YW, Chern JW, Chou JY, Teng CM. Identification of apoptotic and antiangiogenic activities of terazosin in human prostate cancer and endothelial cells. J Urol. 2003;169:724–729. doi: 10.1097/00005392-200302000-00074.
    1. Irwin DC, van Patot MC Tissot, Tucker A, Bowen R. Direct ANP inhibition of hypoxia-induced inflammatory pathways in pulmonary microvascular and macrovascular endothelial monolayers. Am J Physiol Lung Cell Mol Physiol. 2005;288:L849–L859. doi: 10.1152/ajplung.00294.2004.
    1. Demirtas S, Caliskan A, Guclu O, Yazici S, Karahan O, Yavuz C, Mavitas B. Can calcium dobesilate be used safely for peripheral microvasculopathies that require neoangiogenesis? Med Sci Monit Basic Res. 2013;19:253–257. doi: 10.12659/MSMBR.889427.
    1. Zhang X, Liu W, Wu S, Jin J, Li W, Wang N. Calcium dobesilate for diabetic retinopathy: A systematic review and meta-analysis. Sci China Life Sci. 2015;58:101–107. doi: 10.1007/s11427-014-4792-1.
    1. Krakoff J, Lindsay RS, Looker HC, Nelson RG, Hanson RL, Knowler WC. Incidence of retinopathy and nephropathy in youth-onset compared with adult-onset type 2 diabetes. Diabetes Care. 2003;26:76–81. doi: 10.2337/diacare.26.1.76.
    1. Seyer-Hansen K. Renal hypertrophy in experimental diabetes mellitus. Kidney Int. 1983;23:643–646. doi: 10.1038/ki.1983.71.
    1. Seyer-Hansen K. Renal hypertrophy in streptozotocin-diabetic rats. Clin Sci Mol Med. 1976;51:551–555.
    1. Nyengaard JR, Rasch R. The impact of experimental diabetes mellitus in rats on glomerular capillary number and sizes. Diabetologia. 1993;36:189–194. doi: 10.1007/BF00399948.
    1. Osterby R, Nyberg G. New vessel formation in the renal corpuscles in advanced diabetic glomerulopathy. J Diabet Complications. 1987;1:122–127. doi: 10.1016/S0891-6632(87)80069-7.
    1. Wehner H, Nelischer G. Morphometric investigations on intrarenal vessels of streptozotocin-diabetic rats. Virchows Arch A Pathol Anat Histopathol. 1991;419:231–235. doi: 10.1007/BF01626353.
    1. Nakagawa T, Kosugi T, Haneda M, Rivard CJ, Long DA. Abnormal angiogenesis in diabetic nephropathy. Diabetes. 2009;58:1471–1478. doi: 10.2337/db09-0119.
    1. Kanesaki Y, Suzuki D, Uehara G, Toyoda M, Katoh T, Sakai H, Watanabe T. Vascular endothelial growth factor gene expression is correlated with glomerular neovascularization in human diabetic nephropathy. Am J Kidney Dis. 2005;45:288–294. doi: 10.1053/j.ajkd.2004.09.020.
    1. Min W, Yamanaka N. Three-dimensional analysis of increased vasculature around the glomerular vascular pole in diabetic nephropathy. Virchows Arch A Pathol Anat Histopathol. 1993;423:201–207. doi: 10.1007/BF01614771.
    1. Yuan J, Fang W, Lin A, Ni Z, Qian J. Angiopoietin-2/Tie2 signaling involved in TNF-α induced peritoneal angiogenesis. Int J Artif Organs. 2012;35:655–662.
    1. Cooper ME, Vranes D, Youssef S, Stacker SA, Cox AJ, Rizkalla B, Casley DJ, Bach LA, Kelly DJ, Gilbert RE. Increased renal expression of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 in experimental diabetes. Diabetes. 1999;48:2229–2239. doi: 10.2337/diabetes.48.11.2229.
    1. Hohenstein B, Hausknecht B, Boehmer K, Riess R, Brekken RA, Hugo CP. Local VEGF activity but not VEGF expression is tightly regulated during diabetic nephropathy in man. Kidney Int. 2006;69:1654–1661. doi: 10.1038/sj.ki.5000294.
    1. Baelde HJ, Eikmans M, Doran PP, Lappin DW, de Heer E, Bruijn JA. Gene expression profiling in glomeruli from human kidneys with diabetic nephropathy. Am J Kidney Dis. 2004;43:636–650. doi: 10.1053/j.ajkd.2003.12.028.
    1. Baelde HJ, Eikmans M, Lappin DW, Doran PP, Hohenadel D, Brinkkoetter PT, Van der Woude FJ, Waldherr R, Rabelink TJ, de Heer E, Bruijn JA. Reduction of VEGF-A and CTGF expression in diabetic nephropathy is associated with podocyte loss. Kidney Int. 2007;71:637–645. doi: 10.1038/sj.ki.5002101.
    1. Lindenmeyer MT, Kretzler M, Boucherot A, Berra S, Yasuda Y, Henger A, Eichinger F, Gaiser S, Schmid H, Rastaldi MP, et al. Interstitial vascular rarefaction and reduced VEGF-A expression in human diabetic nephropathy. J Am Soc Nephrol. 2007;18:1765–1776. doi: 10.1681/ASN.2006121304.
    1. García-García PM, Getino-Melián MA, Domínguez-Pimentel V, Navarro-González JF. Inflammation in diabetic kidney disease. World J Diabetes. 2014;5:431–443. doi: 10.4239/wjd.v5.i4.431.
    1. Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 2004;27:813–823. doi: 10.2337/diacare.27.3.813.
    1. Navarro JF, Milena FJ, Mora C, León C, García J. Renal pro-inflammatory cytokine gene expression in diabetic nephropathy: Effect of angiotensin-converting enzyme inhibition and pentoxifylline administration. Am J Nephrol. 2006;26:562–570. doi: 10.1159/000098004.
    1. Sekizuka K, Tomino Y, Sei C, Kurusu A, Tashiro K, Yamaguchi Y, Kodera S, Hishiki T, Shirato I, Koide H. Detection of serum IL-6 in patients with diabetic nephropathy. Nephron. 1994;68:284–285. doi: 10.1159/000188281.
    1. Moriwaki Y, Yamamoto T, Shibutani Y, Aoki E, Tsutsumi Z, Takahashi S, Okamura H, Koga M, Fukuchi M, Hada T. Elevated levels of interleukin-18 and tumor necrosis factor-alpha in serum of patients with type 2 diabetes mellitus: Relationship with diabetic nephropathy. Metabolism. 2003;52:605–608. doi: 10.1053/meta.2003.50096.
    1. Navarro JF, Mora C, Muros M, García J. Urinary tumour necrosis factor-alpha excretion independently correlates with clinical markers of glomerular and tubulointerstitial injury in type 2 diabetic patients. Nephrol Dial Transplant. 2006;21:3428–3434. doi: 10.1093/ndt/gfl469.
    1. Royall JA, Berkow RL, Beckman JS, Cunningham MK, Matalon S, Freeman BA. Tumor necrosis factor and interleukin 1 alpha increase vascular endothelial permeability. Am J Physiol. 1989;257:L399–L410.
    1. Navarro-González JF, Mora-Fernández C. The role of inflammatory cytokines in diabetic nephropathy. J Am Soc Nephrol. 2008;19:433–442. doi: 10.1681/ASN.2007091048.
    1. Vestra M Dalla, Mussap M, Gallina P, Bruseghin M, Cernigoi AM, Saller A, Plebani M, Fioretto P. Acute-phase markers of inflammation and glomerular structure in patients with type 2 diabetes. J Am Soc Nephrol. 2005;16(Suppl 1):S78–S82. doi: 10.1681/ASN.2004110961.
    1. Ruef C, Budde K, Lacy J, Northemann W, Baumann M, Sterzel RB, Coleman DL. Interleukin 6 is an autocrine growth factor for mesangial cells. Kidney Int. 1990;38:249–257. doi: 10.1038/ki.1990.193.
    1. Mariño E, Cardier JE. Differential effect of IL-18 on endothelial cell apoptosis mediated by TNF-alpha and Fas (CD95) Cytokine. 2003;22:142–148. doi: 10.1016/S1043-4666(03)00150-9.
    1. Stuyt RJ, Netea MG, Geijtenbeek TB, Kullberg BJ, Dinarello CA, van der Meer JW. Selective regulation of intercellular adhesion molecule-1 expression by interleukin-18 and interleukin-12 on human monocytes. Immunology. 2003;110:329–334. doi: 10.1046/j.1365-2567.2003.01747.x.
    1. Bertani T, Abbate M, Zoja C, Corna D, Perico N, Ghezzi P, Remuzzi G. Tumor necrosis factor induces glomerular damage in the rabbit. Am J Pathol. 1989;134:419–430.
    1. DiPetrillo K, Coutermarsh B, Gesek FA. Urinary tumor necrosis factor contributes to sodium retention and renal hypertrophy during diabetes. Am J Physiol Renal Physiol. 2003;284:F113–F121. doi: 10.1152/ajprenal.00026.2002.
    1. Dipetrillo K, Gesek FA. Pentoxifylline ameliorates renal tumor necrosis factor expression, sodium retention, and renal hypertrophy in diabetic rats. Am J Nephrol. 2004;24:352–359. doi: 10.1159/000079121.

Source: PubMed

3
Iratkozz fel