GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Endocrinology, The Endocrine Society, Vol. 148, No. 9 ( 2007-09-01), p. 4371-4384
    Abstract: Diabetes is associated with increased generation of cytokines and tissue inflammation, but it is unclear how increased cytokine synthesis is causally related to the development of diabetic complications. Here, we report that exposure to high (25 mm) glucose, but not iso-osmotic concentrations of mannitol or 3-methyl glucose, increased TNF-α secretion by rat and human aortic smooth muscle cells in culture. The increase in TNF-α production was prevented by actinomycin D and cycloheximide, indicating transcriptional activation of TNF-α gene. High glucose (HG)-induced TNF-α release was specifically inhibited by protein kinase C (PKC)-δ inhibitor (Rottlerin; EMD Biosciences, San Diego, CA), but not PKC-β2 inhibitor (CGP53353; Tocris Cookson Inc., Ellisville, MO), indicating the possible involvement of PKC-δ in HG signaling. TNF-α secretion was also prevented by pretreating cells with aldose reductase (AR) inhibitors, sorbinil or tolrestat and in cells treated with antisense AR mRNA. Inhibition of AR also prevented the increase in TNF-α mRNA. Addition of anti-TNF-α antibodies or soluble TNF-α receptors 1 and 2 to the medium or RNA interference ablation of TNF-α attenuated nuclear factor-κB activation and prevented HG-stimulated cell growth. These data indicate that AR is required for HG-induced TNF-α synthesis and release. In vivo, the release of TNF-α by HG leading to autocrine stimulation of TNF-α synthesis may be a critical step in the development of the cardiovascular complications of diabetes. Interruption of the autocrine effects of TNF-α may be a useful strategy for treating diabetic vasculopathies.
    Type of Medium: Online Resource
    ISSN: 0013-7227 , 1945-7170
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2007
    detail.hit.zdb_id: 2011695-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2011
    In:  Angiogenesis Vol. 14, No. 2 ( 2011-5), p. 209-221
    In: Angiogenesis, Springer Science and Business Media LLC, Vol. 14, No. 2 ( 2011-5), p. 209-221
    Type of Medium: Online Resource
    ISSN: 0969-6970 , 1573-7209
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 2003393-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 1384-1384
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 1384-1384
    Abstract: We have recently shown that Aldose Reductase (AR) known to be involved in oxidative stress-signaling, initiated by inflammatory markers, prevents human colon cancer cell growth in culture as well as in nude mice xenografts. Inhibition of AR also prevents azoxymethane-induced aberrant crypt foci formation in mice. In order to understand the chemopreventive mechanism of AR inhibition in colon cancer, we have investigated the role of AR in the mediation of angiogenic signals in vitro and in vivo models. Using human umbilical vein endothelial cells (HUVEC), we have investigated the effect of AR inhibition on VEGF- and bFGF- induced tube as well as spheroid formation in in vitro angiogenesis. We have also determined the invasion and migration of HUVEC by inhibition of AR. Further, to determine the in vivo efficacy of AR inhibition on angiogenesis, matrigel plug assay was performed using a rat model. Our results show that inhibition of AR significantly prevented the VEGF- and bFGF -induced proliferation and generation of reactive oxygen species in HUVEC. Further, AR inhibition also prevented the VEGF- and bFGF- induced secretion/expression of IL-6, MMP1, MMP9, ICAM, and VCAM. Matrigel plug model of angiogenesis in rats showed that inhibition of AR prevented the infiltration of blood cells, invasion, migration and formation of capillary like structures, and expression of blood vessels markers CD31 and vWF. Thus, our results demonstrate that AR inhibitors could be novel drugs for cancer chemoprevention by inhibiting angiogenesis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1384.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3233-3233
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 3233-3233
    Abstract: Lung cancer is the second most common cancer and the leading cause of cancer death among both men and women. Although, tobacco smoking is the dominant etiologic factor responsible for the vast majority of lung cancers, other factors such as asbestos, pollutants, polycyclic aromatic hydrocarbons, nutritional and dietary factors which can cause pulmonary inflammation can also drive carcinogenesis by altering cellular and molecular targets and pathways that are crucial to normal tissue homeostasis. However, the molecular signaling events specific to the pulmonary inflammation and their effects on pulmonary cells leading to carcinogenesis are poorly understood. Hence, we investigated the role of polyol pathway enzyme aldose reductase (AR), an oxidative stress response protein that catalyzes the reduction of lipid peroxidation -derived lipid aldehydes and their glutathione conjugates, in the mediation of carcinogenic signals leading to lung cancer. We examined the effect of pharmacological and siRNA -mediated inhibition of AR on in vitro human lung carcinoma cell line A549 growth as well as tumor growth in nude mice xenografts. We have also examined the molecular mechanisms by which AR inhibition prevents carcinogenic signaling events that cause lung cancer cell growth. Treatment of A549 cells with AR inhibitors prevented the activation of NF-kB and expression of NF-kB- dependent inflammatory markers such as iNOS and Cox-2. AR inhibition also prevented the proliferation of cultured human lung cancer cells. Further, AR inhibition significantly arrested the tumor growth in nude mice bearing lung adenocarcinoma xenografts. Similar results were observed in AR-siRNA treated nude mice xenografts. Immunohistochemical analysis of nude mice tumor cross sections indicated that AR inhibition prevents activation of iNOS, Cox-2, AR and NF-kB. Our studies thus strongly suggest that AR may be a critical regulator of signaling pathway(s) that cause lung cancer. Hence, disrupting these signals by AR inhibition could be a novel preventive approach in lung cancer progression. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3233.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Elsevier BV ; 2007
    In:  Cancer Letters Vol. 252, No. 2 ( 2007-7), p. 299-306
    In: Cancer Letters, Elsevier BV, Vol. 252, No. 2 ( 2007-7), p. 299-306
    Type of Medium: Online Resource
    ISSN: 0304-3835
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2007
    detail.hit.zdb_id: 195674-7
    detail.hit.zdb_id: 2004212-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 294, No. 5 ( 2019-02), p. 1634-
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Diabetes Association ; 2006
    In:  Diabetes Vol. 55, No. 4 ( 2006-04-01), p. 901-910
    In: Diabetes, American Diabetes Association, Vol. 55, No. 4 ( 2006-04-01), p. 901-910
    Abstract: The objective of this study was to determine whether the polyol pathway enzyme aldose reductase mediates diabetes abnormalities in vascular smooth muscle cell (SMC) growth. Aldose reductase inhibitors (tolrestat or sorbinil) or antisense aldose reductase mRNA prevented hyperproliferation of cultured rat aortic SMCs induced by high glucose. Cell cycle progression in the presence of high glucose was blocked by tolrestat, which induced a G0-G1 phase growth arrest. In situ, diabetes increased SMC growth and intimal hyperplasia in balloon-injured carotid arteries of streptozotocin-treated rats, when examined 7 or 14 days after injury. Treatment with tolrestat (15 mg · kg−1 · day−1) diminished intimal hyperplasia and decreased SMC content of the lesion by 25%. Although tolrestat treatment increased immunoreactivity of the lesion with antibodies raised against protein adducts of the lipid peroxidation product 4-hydroxy trans-2-nonenal, no compensatory increase in lesion fibrosis was observed. Collectively, these results suggest that inhibition of aldose reductase prevents glucose-induced stimulation of SMC growth in culture and in situ. Even though inhibition of aldose reductase increases vascular oxidative stress, this approach may be useful in preventing abnormal SMC growth in vessels of diabetic patients.
    Type of Medium: Online Resource
    ISSN: 0012-1797 , 1939-327X
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2006
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Letters, Elsevier BV, Vol. 355, No. 1 ( 2014-12), p. 141-147
    Type of Medium: Online Resource
    ISSN: 0304-3835
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 195674-7
    detail.hit.zdb_id: 2004212-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Elsevier BV ; 2011
    In:  Chemico-Biological Interactions Vol. 191, No. 1-3 ( 2011-5), p. 346-350
    In: Chemico-Biological Interactions, Elsevier BV, Vol. 191, No. 1-3 ( 2011-5), p. 346-350
    Type of Medium: Online Resource
    ISSN: 0009-2797
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2011
    detail.hit.zdb_id: 1496834-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 424-424
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 424-424
    Abstract: Worldwide colorectal cancer (CRC) is the third most common cause of cancer and is the second leading cause of cancer deaths in the USA. Although therapeutic options for patients with CRC have increased substantially, including earlier diagnosis, improved surgery as well as chemo- and radiation therapies, ∼61% of patients have metastatic disease and of these, 90% die within 5 years of diagnosis. Hence better therapeutic interventions to manage the metastatic process in CRC patients are mandatory. We have recently shown that inhibition of aldose reductase (AR), an enzyme that catalyzes reduction of lipid aldehydes and their glutathione conjugates, prevents human colon cancer cell growth in culture as well as in nude mice xenografts by inhibiting the NF-kB-dependent activation of inflammatory and carcinogenic markers. We have now investigated the effect of AR inhibition in metastatic spread of human CRC cells in nude mice and the molecular mechanisms by which AR inhibition prevents colon cancer cell invasion, migration, adhesion and metastasis. Our results indicate that AR inhibition prevents growth factors-induced migration and invasion of HT-29 CRC cells in a dose-dependent manner. AR inhibition also significantly inhibited adhesion of CRC cells to endothelial cells and inhibited the expression of ICAM-1, VCAM-1 and VE-cadherin. Further, we injected KM20 cells, transfected with a plasmid containing green fluorescent protein (GFP), into the spleen of athymic nude mice to establish liver metastases. The mice were treated with AR inhibitor or siRNA for 30 days. Our results indicate that AR inhibition or ablation prevents in vivo invasion and metastasis of CRC cells by reducing levels of AR, MMP2, cyclin D1, and CD34 and suppresses the activation of NF-kB. Since AR inhibitor fidarestat aggressively prevents human tumor growth and metastasis in nude mice and cellular models, and has already undergone phase-iii clinical studies for diabetic neuropathy, it is an excellent candidate for clinical application in preventing colon cancer tumorigenesis and metastasis. Funding Support: NIH/NCI CA129383 (SKS). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 424.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...