16.07.2015 Views

review of literature on clinical pancreatology - The Pancreapedia

review of literature on clinical pancreatology - The Pancreapedia

review of literature on clinical pancreatology - The Pancreapedia

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

immunostain (-60 %). Rosiglitaz<strong>on</strong>e reduced fasting glucose and insulin but induced weightgain. Fibrates impeded weight gain, but <strong>on</strong>ly bezafibrate prevented islet hypertrophy. <strong>The</strong>GLUT2 stain was improved in all treatments, and there were no alterati<strong>on</strong>s in PPAR-alpha.<strong>The</strong>re were morphological signs <str<strong>on</strong>g>of</str<strong>on</strong>g> pancreatitis with fen<str<strong>on</strong>g>of</str<strong>on</strong>g>ibrate, although there were noalterati<strong>on</strong>s in amylase and lipase. Rosiglitaz<strong>on</strong>e exacerbated pancreatic fat infiltrati<strong>on</strong> (+75 %vs HFHS group), and bezafibrate increased PPAR-beta expressi<strong>on</strong> in pancreatic islets. <strong>The</strong>authors c<strong>on</strong>cluded that rosiglitaz<strong>on</strong>e is shown for the first time to exacerbate pancreatic fatinfiltrati<strong>on</strong>; therefore, precauti<strong>on</strong> has to be taken when rosiglitaz<strong>on</strong>e is prescribed to obesepatients [397].Somatostatin receptor subtype 2<strong>The</strong> somatostatin receptor subtype 2 (sst2) behaves as a tumor suppressor when expressedand stimulated by its ligand somatostatin in pancreatic cancer. It was reveal a mechanismunderlying <strong>on</strong>cosuppressive acti<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> sst2, whereby this inhibitory receptor upregulates theexpressi<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> the secreted angioinhibitory factor thrombosp<strong>on</strong>din-1 (TSP-1), asdem<strong>on</strong>strated in exocrine BxPC-3 and endocrine BON pancreatic cancer cells. <strong>The</strong> sst2-dependent upregulati<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> TSP-1 occurs through the inhibiti<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> the PI3K pathway. Itdepends <strong>on</strong> transcripti<strong>on</strong>al and translati<strong>on</strong>al events, involving a previously undescribed IRESin the 5'-UTR <str<strong>on</strong>g>of</str<strong>on</strong>g> TSP-1 mRNA. Chick chorioallantoic membrane was used as an in vivomodel to dem<strong>on</strong>strate that TSP-1 is a critical effector <str<strong>on</strong>g>of</str<strong>on</strong>g> the inhibitory role <str<strong>on</strong>g>of</str<strong>on</strong>g> sst2 <strong>on</strong> theneoangiogenesis and <strong>on</strong>cogenesis induced by pancreatic cancer cells. TSP-1 reduced invitro tubulogenesis <str<strong>on</strong>g>of</str<strong>on</strong>g> endothelial cells when grown in c<strong>on</strong>diti<strong>on</strong>ed medium from pancreaticcancer cells expressing sst2, as compared to those expressing the c<strong>on</strong>trol vector. TSP-1inhibited tumor cell-induced neoangiogenesis by directly sequestering the proangiogenicfactor VEGF, and inactivating the angiogenesis initiated by VEGFR2 phosphorylati<strong>on</strong> inendothelial cells. Using human pancreatic tissue-microarrays, the expressi<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> both sst2and TSP-1 was shown to be correlated during the pancreatic neoplastic program. Bothproteins are nearly undetectable in normal exocrine pancreas and in most invasive cancerlesi<strong>on</strong>s, but their expressi<strong>on</strong> is strikingly upregulated in most preinvasive cancer-adjacentlesi<strong>on</strong>s. <strong>The</strong> upregulati<strong>on</strong> <str<strong>on</strong>g>of</str<strong>on</strong>g> both sst2 and TSP-1 tumor suppressors may functi<strong>on</strong> as anearly negative feedback to restrain pancreatic carcinogenesis [398].SphingolipidsDefeating pancreatic cancer resistance to the chemotherapeutic drug gemcitabine remains achallenge to treat this deadly cancer. Targeting the sphingolipid metabolism for improvingtumor chemosensitivity has recently emerged as a promising strategy. <strong>The</strong> fine balancebetween intracellular levels <str<strong>on</strong>g>of</str<strong>on</strong>g> the prosurvival sphingosine-1-phosphate (S1P) and theproapoptotic ceramide sphingolipids determines cell fate. Am<strong>on</strong>g enzymes that c<strong>on</strong>trol thismetabolism, sphingosine kinase-1 (SphK1), a tumor-associated protein overexpressed inmany cancers, favors survival through S1P producti<strong>on</strong>, and inhibitors <str<strong>on</strong>g>of</str<strong>on</strong>g> SphK1 are used in<strong>on</strong>going <strong>clinical</strong> trials to sensitize epithelial ovarian and prostate cancer cells to variouschemotherapeutic drugs. It was reported that the cellular ceramide/S1P ratio is a criticalbiosensor for predicting pancreatic cancer cell sensitivity to gemcitabine. A low level <str<strong>on</strong>g>of</str<strong>on</strong>g> theceramide/S1P ratio, associated with a high SphK1 activity, correlates with a robust intrinsicpancreatic cancer cell chemoresistance toward gemcitabine. Strikingly, increasing theceramide/S1P ratio, by using pharmacologic (SphK1 inhibitor or ceramide analogue) or smallinterfering RNA-based approaches to up-regulate intracellular ceramide levels or reduceSphK1 activity, sensitized pancreatic cancer cells to gemcitabine. C<strong>on</strong>versely, decreasingthe ceramide/S1P ratio, by up-regulating SphK1 activity, promoted gemcitabine resistance inthese cells [399].

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!