Skip to main content
Erschienen in: Annals of Surgical Oncology 4/2009

01.04.2009 | Hepatobiliary and Pancreatic Tumors

Low-Level Expression of Smad7 Correlates with Lymph Node Metastasis and Poor Prognosis in Patients with Pancreatic Cancer

verfasst von: Peng Wang, MD, Jie Fan, MD, Zhen Chen, MD, Zhi-Qiang Meng, MD, Jian-Min Luo, PhD, Jun-Hua Lin, MD, Zhen-Hua Zhou, MD, Hao Chen, MD, Kun Wang, MD, Zu-De Xu, MD, Lu-Ming Liu, MD

Erschienen in: Annals of Surgical Oncology | Ausgabe 4/2009

Einloggen, um Zugang zu erhalten

Abstract

Background

Whether Smad7 acts as a tumor proliferation promoting factor or as a metastatic suppressor in human pancreatic cancer remains unclear. This study aims to determine the prognostic value of Smad7 in patients with pancreatic adenocarcinoma.

Methods

Surgical specimens obtained from 71 patients with pancreatic adenocarcinoma were immunohistochemically assessed for Smad7, Ki-67, MMP2, CD34, and Smad4 expression. The relationship between Smad7 expression and the clinicopathological characteristics of patients with pancreatic adenocarcinoma were also evaluated.

Results

Fifty-one of 71 specimens (71.8%) were Smad7 positive and 20 specimens were Smad7 negative. Negative expression of Smad7 correlated with lymph node metastasis, liver metastasis after surgery, and a poor survival rate (P = 0.0004, 0.0044, and 0.0003, respectively). We also found an inverse correlation between the expression of Smad7 and MMP2 (P = 0.0189). Multivariate analysis revealed that Smad7 expression was an independent prognostic factor [hazard ratio (HR) 0.3902; 95% confidence interval (CI) 0.1839–0.8277; P = 0.0142]. Furthermore, in both Smad4-negative and Smad4-positive groups, survival of patients with Smad7-positive tumors was significantly better than those with Smad7-negative tumors (both P < 0.0001).

Conclusions

We conclude that low-level expression of Smad7 in pancreatic cancer is significantly associated with lymph node metastasis, high MMP2 expression, and poor prognosis.
Literatur
1.
Zurück zum Zitat Goggins M, Shekher M, Turnacioglu K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res. 1998;58:5329–32.PubMed Goggins M, Shekher M, Turnacioglu K, et al. Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res. 1998;58:5329–32.PubMed
2.
Zurück zum Zitat Jonson T, Gorunova L, Dawiskiba S, et al. Molecular analyses of the 15q and 18q SMAD genes in pancreatic cancer. Genes Chromosomes Cancer. 1999;24:62–71.PubMedCrossRef Jonson T, Gorunova L, Dawiskiba S, et al. Molecular analyses of the 15q and 18q SMAD genes in pancreatic cancer. Genes Chromosomes Cancer. 1999;24:62–71.PubMedCrossRef
3.
Zurück zum Zitat Villanueva A, Garcia C, Paules AB, et al. Disruption of the antiproliferative TGF-beta signaling pathways in human pancreatic cancer cells. Oncogene. 1998;17:1969–78.PubMedCrossRef Villanueva A, Garcia C, Paules AB, et al. Disruption of the antiproliferative TGF-beta signaling pathways in human pancreatic cancer cells. Oncogene. 1998;17:1969–78.PubMedCrossRef
4.
Zurück zum Zitat Markowitz S, Wang J, Myeroff L, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995;268:1336–8.PubMedCrossRef Markowitz S, Wang J, Myeroff L, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science. 1995;268:1336–8.PubMedCrossRef
5.
Zurück zum Zitat Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–3.PubMedCrossRef Hahn SA, Schutte M, Hoque AT, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350–3.PubMedCrossRef
6.
Zurück zum Zitat Schutte M, Hruban RH, Hedrick L, et al. DPC4 gene in various tumor types. Cancer Res. 1996;56:2527–30.PubMed Schutte M, Hruban RH, Hedrick L, et al. DPC4 gene in various tumor types. Cancer Res. 1996;56:2527–30.PubMed
7.
Zurück zum Zitat Togo G, Toda N, Kanai F, et al. A transforming growth factor beta type II receptor gene mutation common in sporadic cecum cancer with microsatellite instability. Cancer Res. 1996;56:5620–3.PubMed Togo G, Toda N, Kanai F, et al. A transforming growth factor beta type II receptor gene mutation common in sporadic cecum cancer with microsatellite instability. Cancer Res. 1996;56:5620–3.PubMed
8.
Zurück zum Zitat Miyaki M, Iijima T, Konishi M, et al. Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene. 1999;18:3098–103.PubMedCrossRef Miyaki M, Iijima T, Konishi M, et al. Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis. Oncogene. 1999;18:3098–103.PubMedCrossRef
9.
Zurück zum Zitat Wang D, Kanuma T, Mizunuma H, et al. Analysis of specific gene mutations in the transforming growth factor-beta signal transduction pathway in human ovarian cancer. Cancer Res. 2000;60:4507–12.PubMed Wang D, Kanuma T, Mizunuma H, et al. Analysis of specific gene mutations in the transforming growth factor-beta signal transduction pathway in human ovarian cancer. Cancer Res. 2000;60:4507–12.PubMed
10.
Zurück zum Zitat Boulay JL, Mild G, Lowy A, et al. SMAD7 is a prognostic marker in patients with colorectal cancer. Int J Cancer. 2003;104:446–9.PubMedCrossRef Boulay JL, Mild G, Lowy A, et al. SMAD7 is a prognostic marker in patients with colorectal cancer. Int J Cancer. 2003;104:446–9.PubMedCrossRef
11.
Zurück zum Zitat Cerutti JM, Ebina KN, Matsuo SE, et al. Expression of Smad4 and Smad7 in human thyroid follicular carcinoma cell lines. J Endocrinol Invest. 2003;26:516–21.PubMed Cerutti JM, Ebina KN, Matsuo SE, et al. Expression of Smad4 and Smad7 in human thyroid follicular carcinoma cell lines. J Endocrinol Invest. 2003;26:516–21.PubMed
12.
Zurück zum Zitat Kim YH, Lee HS, Lee HJ, et al. Prognostic significance of the expression of Smad4 and Smad7 in human gastric carcinomas. Ann Oncol. 2004;15:574–80.PubMedCrossRef Kim YH, Lee HS, Lee HJ, et al. Prognostic significance of the expression of Smad4 and Smad7 in human gastric carcinomas. Ann Oncol. 2004;15:574–80.PubMedCrossRef
13.
Zurück zum Zitat Monteleone G, Del Vecchio Blanco G, Palmieri G, et al. Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection. Gastroenterology. 2004;126:674–82.PubMedCrossRef Monteleone G, Del Vecchio Blanco G, Palmieri G, et al. Induction and regulation of Smad7 in the gastric mucosa of patients with Helicobacter pylori infection. Gastroenterology. 2004;126:674–82.PubMedCrossRef
14.
Zurück zum Zitat Dowdy SC, Mariani A, Reinholz MM, et al. Overexpression of the TGF-beta antagonist Smad7 in endometrial cancer. Gynecol Oncol. 2005;96:368–73.PubMedCrossRef Dowdy SC, Mariani A, Reinholz MM, et al. Overexpression of the TGF-beta antagonist Smad7 in endometrial cancer. Gynecol Oncol. 2005;96:368–73.PubMedCrossRef
15.
Zurück zum Zitat Kuang C, Xiao Y, Liu X, et al. In vivo disruption of TGF-beta signaling by Smad7 leads to premalignant ductal lesions in the pancreas. Proc Natl Acad Sci USA. 2006;103:1858–63.PubMedCrossRef Kuang C, Xiao Y, Liu X, et al. In vivo disruption of TGF-beta signaling by Smad7 leads to premalignant ductal lesions in the pancreas. Proc Natl Acad Sci USA. 2006;103:1858–63.PubMedCrossRef
16.
Zurück zum Zitat Kleeff J, Maruyama H, Friess H, et al. Smad6 suppresses TGF-beta-induced growth inhibition in COLO–357 pancreatic cancer cells and is overexpressed in pancreatic cancer. Biochem Biophys Res Commun. 1999;255:268–73.PubMedCrossRef Kleeff J, Maruyama H, Friess H, et al. Smad6 suppresses TGF-beta-induced growth inhibition in COLO–357 pancreatic cancer cells and is overexpressed in pancreatic cancer. Biochem Biophys Res Commun. 1999;255:268–73.PubMedCrossRef
17.
Zurück zum Zitat Halder SK, Beauchamp RD, Datta PK. Smad7 induces tumorigenicity by blocking TGF-beta-induced growth inhibition and apoptosis. Exp Cell Res. 2005;307:231–46.PubMedCrossRef Halder SK, Beauchamp RD, Datta PK. Smad7 induces tumorigenicity by blocking TGF-beta-induced growth inhibition and apoptosis. Exp Cell Res. 2005;307:231–46.PubMedCrossRef
18.
Zurück zum Zitat Akhurst RJ, Derynck R. TGF-beta signaling in cancer—a double-edged sword. Trends Cell Biol. 2001;11:S44–51.PubMed Akhurst RJ, Derynck R. TGF-beta signaling in cancer—a double-edged sword. Trends Cell Biol. 2001;11:S44–51.PubMed
19.
Zurück zum Zitat Wakefield LM, Roberts AB. TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev. 2002;12:22–9.PubMedCrossRef Wakefield LM, Roberts AB. TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev. 2002;12:22–9.PubMedCrossRef
20.
Zurück zum Zitat Azuma H, Ehata S, Miyazaki H, et al. Effect of Smad7 expression on metastasis of mouse mammary carcinoma JygMC(A) cells. J Natl Cancer Inst. 2005;97:1734–46.PubMedCrossRef Azuma H, Ehata S, Miyazaki H, et al. Effect of Smad7 expression on metastasis of mouse mammary carcinoma JygMC(A) cells. J Natl Cancer Inst. 2005;97:1734–46.PubMedCrossRef
21.
Zurück zum Zitat Javelaud D, Mohammad KS, McKenna CR, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res. 2007;67:2317–24.PubMedCrossRef Javelaud D, Mohammad KS, McKenna CR, et al. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis. Cancer Res. 2007;67:2317–24.PubMedCrossRef
22.
Zurück zum Zitat Sobin LH, Wittekind CH. International Union Against Cancer. TNM classification of malignant tumours (6th ed.). New York: Wiley-Liss; 2002. Sobin LH, Wittekind CH. International Union Against Cancer. TNM classification of malignant tumours (6th ed.). New York: Wiley-Liss; 2002.
23.
Zurück zum Zitat Fukuchi M, Fukai Y, Masuda N, et al. High-level expression of the Smad ubiquitin ligase Smurf2 correlates with poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Res. 2002;62:7162–5.PubMed Fukuchi M, Fukai Y, Masuda N, et al. High-level expression of the Smad ubiquitin ligase Smurf2 correlates with poor prognosis in patients with esophageal squamous cell carcinoma. Cancer Res. 2002;62:7162–5.PubMed
24.
Zurück zum Zitat Takahashi H, Oda T, Hasebe T, et al. Biologically different subgroups of invasive ductal carcinoma of the pancreas: Dpc4 status according to the ratio of intraductal carcinoma components. Clin Cancer Res. 2004;10:3772–9.PubMedCrossRef Takahashi H, Oda T, Hasebe T, et al. Biologically different subgroups of invasive ductal carcinoma of the pancreas: Dpc4 status according to the ratio of intraductal carcinoma components. Clin Cancer Res. 2004;10:3772–9.PubMedCrossRef
25.
Zurück zum Zitat Ebisawa T, Fukuchi M, Murakami G, et al. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 2001;276:12477–80.PubMedCrossRef Ebisawa T, Fukuchi M, Murakami G, et al. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 2001;276:12477–80.PubMedCrossRef
26.
Zurück zum Zitat Shi W, Sun C, He B, et al. GADD34-PP1c recruited by Smad7 dephosphorylates TGFbeta type I receptor. J Cell Biol. 2004;164:291–300.PubMedCrossRef Shi W, Sun C, He B, et al. GADD34-PP1c recruited by Smad7 dephosphorylates TGFbeta type I receptor. J Cell Biol. 2004;164:291–300.PubMedCrossRef
27.
Zurück zum Zitat Hata A, Lagna G, Massague J, et al. Smad6 inhibits BMP/Smad1 signaling by specifically competing with the Smad4 tumor suppressor. Genes Dev. 1998;12:186–97.PubMedCrossRef Hata A, Lagna G, Massague J, et al. Smad6 inhibits BMP/Smad1 signaling by specifically competing with the Smad4 tumor suppressor. Genes Dev. 1998;12:186–97.PubMedCrossRef
28.
Zurück zum Zitat Bai S, Cao X. A nuclear antagonistic mechanism of inhibitory Smads in transforming growth factor-beta signaling. J Biol Chem. 2002;277:4176–82.PubMedCrossRef Bai S, Cao X. A nuclear antagonistic mechanism of inhibitory Smads in transforming growth factor-beta signaling. J Biol Chem. 2002;277:4176–82.PubMedCrossRef
29.
Zurück zum Zitat Osawa H, Nakajima M, Kato H, et al. Prognostic value of the expression of Smad6 and Smad7, as inhibitory Smads of the TGF-beta superfamily, in esophageal squamous cell carcinoma. Anticancer Res. 2004;24:3703–9.PubMed Osawa H, Nakajima M, Kato H, et al. Prognostic value of the expression of Smad6 and Smad7, as inhibitory Smads of the TGF-beta superfamily, in esophageal squamous cell carcinoma. Anticancer Res. 2004;24:3703–9.PubMed
30.
Zurück zum Zitat Kleeff J, Ishiwata T, Maruyama H, et al. The TGF-beta signaling inhibitor Smad7 enhances tumorigenicity in pancreatic cancer. Oncogene. 1999;18:5363–72.PubMedCrossRef Kleeff J, Ishiwata T, Maruyama H, et al. The TGF-beta signaling inhibitor Smad7 enhances tumorigenicity in pancreatic cancer. Oncogene. 1999;18:5363–72.PubMedCrossRef
31.
Zurück zum Zitat Zhu HJ, Iaria J, Sizeland AM. Smad7 differentially regulates transforming growth factor beta-mediated signaling pathways. J Biol Chem. 1999;274:32258–64.PubMedCrossRef Zhu HJ, Iaria J, Sizeland AM. Smad7 differentially regulates transforming growth factor beta-mediated signaling pathways. J Biol Chem. 1999;274:32258–64.PubMedCrossRef
32.
Zurück zum Zitat Dahler AL, Cavanagh LL, Saunders NA. Suppression of keratinocyte growth and differentiation by transforming growth factor beta1 involves multiple signaling pathways. J Invest Dermatol. 2001;116:266–74.PubMedCrossRef Dahler AL, Cavanagh LL, Saunders NA. Suppression of keratinocyte growth and differentiation by transforming growth factor beta1 involves multiple signaling pathways. J Invest Dermatol. 2001;116:266–74.PubMedCrossRef
33.
Zurück zum Zitat Blanchette F, Rivard N, Rudd P, et al. Cross-talk between the p42/p44 MAP kinase and Smad pathways in transforming growth factor beta 1-induced furin gene transactivation. J Biol Chem. 2001;276:33986–94.PubMedCrossRef Blanchette F, Rivard N, Rudd P, et al. Cross-talk between the p42/p44 MAP kinase and Smad pathways in transforming growth factor beta 1-induced furin gene transactivation. J Biol Chem. 2001;276:33986–94.PubMedCrossRef
34.
Zurück zum Zitat Javelaud D, Delmas V, Moller M, et al. Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo. Oncogene. 2005;24:7624–9.PubMedCrossRef Javelaud D, Delmas V, Moller M, et al. Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo. Oncogene. 2005;24:7624–9.PubMedCrossRef
35.
Zurück zum Zitat Boyer Arnold N, Korc M. Smad7 abrogates transforming growth factor-beta1-mediated growth inhibition in COLO–357 cells through functional inactivation of the retinoblastoma protein. J Biol Chem. 2005;280:21858–66.PubMedCrossRef Boyer Arnold N, Korc M. Smad7 abrogates transforming growth factor-beta1-mediated growth inhibition in COLO–357 cells through functional inactivation of the retinoblastoma protein. J Biol Chem. 2005;280:21858–66.PubMedCrossRef
36.
Zurück zum Zitat Kleeff J, Korc M. Up-regulation of transforming growth factor (TGF)-beta receptors by TGF-beta1 in COLO-357 cells. J Biol Chem. 1998;273:7495–500.PubMedCrossRef Kleeff J, Korc M. Up-regulation of transforming growth factor (TGF)-beta receptors by TGF-beta1 in COLO-357 cells. J Biol Chem. 1998;273:7495–500.PubMedCrossRef
37.
Zurück zum Zitat Nicolas FJ, Hill CS. Attenuation of the TGF-beta-Smad signaling pathway in pancreatic tumor cells confers resistance to TGF-beta-induced growth arrest. Oncogene. 2003;22:3698–711.PubMedCrossRef Nicolas FJ, Hill CS. Attenuation of the TGF-beta-Smad signaling pathway in pancreatic tumor cells confers resistance to TGF-beta-induced growth arrest. Oncogene. 2003;22:3698–711.PubMedCrossRef
38.
Zurück zum Zitat Subramanian G, Schwarz RE, Higgins L, et al. Targeting endogenous transforming growth factor beta receptor signaling in SMAD4-deficient human pancreatic carcinoma cells inhibits their invasive phenotype1. Cancer Res. 2004;64:5200–11.PubMedCrossRef Subramanian G, Schwarz RE, Higgins L, et al. Targeting endogenous transforming growth factor beta receptor signaling in SMAD4-deficient human pancreatic carcinoma cells inhibits their invasive phenotype1. Cancer Res. 2004;64:5200–11.PubMedCrossRef
39.
Zurück zum Zitat Pardali K, Moustakas A. Actions of TGF-beta as tumor suppressor and pro-metastatic factor in human cancer. Biochim Biophys Acta. 2007;1775:21–62.PubMed Pardali K, Moustakas A. Actions of TGF-beta as tumor suppressor and pro-metastatic factor in human cancer. Biochim Biophys Acta. 2007;1775:21–62.PubMed
40.
Zurück zum Zitat Ijichi H, Ikenoue T, Kato N, et al. Systematic analysis of the TGF-beta-Smad signaling pathway in gastrointestinal cancer cells. Biochem Biophys Res Commun. 2001;289:350–7.PubMedCrossRef Ijichi H, Ikenoue T, Kato N, et al. Systematic analysis of the TGF-beta-Smad signaling pathway in gastrointestinal cancer cells. Biochem Biophys Res Commun. 2001;289:350–7.PubMedCrossRef
41.
Zurück zum Zitat Ijichi H, Otsuka M, Tateishi K, et al. Smad4-independent regulation of p21/WAF1 by transforming growth factor-beta. Oncogene. 2004;23:1043–51.PubMedCrossRef Ijichi H, Otsuka M, Tateishi K, et al. Smad4-independent regulation of p21/WAF1 by transforming growth factor-beta. Oncogene. 2004;23:1043–51.PubMedCrossRef
42.
Zurück zum Zitat Leivonen SK, Ala-Aho R, Koli K, et al. Activation of Smad signaling enhances collagenase-3 (MMP-13) expression and invasion of head and neck squamous carcinoma cells. Oncogene. 2006;25:2588–600.PubMedCrossRef Leivonen SK, Ala-Aho R, Koli K, et al. Activation of Smad signaling enhances collagenase-3 (MMP-13) expression and invasion of head and neck squamous carcinoma cells. Oncogene. 2006;25:2588–600.PubMedCrossRef
43.
Zurück zum Zitat Delcore R, Rodriguez FJ, Forster J, et al. Significance of lymph node metastases in patients with pancreatic cancer undergoing curative resection. Am J Surg. 1996;172:463–8; discussion 8–9PubMedCrossRef Delcore R, Rodriguez FJ, Forster J, et al. Significance of lymph node metastases in patients with pancreatic cancer undergoing curative resection. Am J Surg. 1996;172:463–8; discussion 8–9PubMedCrossRef
44.
Zurück zum Zitat Pedrazzoli S, DiCarlo V, Dionigi R, et al. Standard versus extended lymphadenectomy associated with pancreatoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas: a multicenter, prospective, randomized study. Lymphadenectomy Study Group. Ann Surg. 1998;228:508–17.PubMedCrossRef Pedrazzoli S, DiCarlo V, Dionigi R, et al. Standard versus extended lymphadenectomy associated with pancreatoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas: a multicenter, prospective, randomized study. Lymphadenectomy Study Group. Ann Surg. 1998;228:508–17.PubMedCrossRef
45.
Zurück zum Zitat Koshiba T, Hosotani R, Wada M, et al. Involvement of matrix metalloproteinase-2 activity in invasion and metastasis of pancreatic carcinoma. Cancer. 1998;82:642–50.PubMedCrossRef Koshiba T, Hosotani R, Wada M, et al. Involvement of matrix metalloproteinase-2 activity in invasion and metastasis of pancreatic carcinoma. Cancer. 1998;82:642–50.PubMedCrossRef
46.
Zurück zum Zitat Juuti A, Lundin J, Nordling S, et al. Epithelial MMP-2 expression correlates with worse prognosis in pancreatic cancer. Oncology. 2006;71:61–8.PubMedCrossRef Juuti A, Lundin J, Nordling S, et al. Epithelial MMP-2 expression correlates with worse prognosis in pancreatic cancer. Oncology. 2006;71:61–8.PubMedCrossRef
47.
48.
Zurück zum Zitat Enholm B, Paavonen K, Ristimaki A, et al. Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia. Oncogene. 1997;14:2475–83.PubMedCrossRef Enholm B, Paavonen K, Ristimaki A, et al. Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia. Oncogene. 1997;14:2475–83.PubMedCrossRef
49.
Zurück zum Zitat Benckert C, Jonas S, Cramer T, et al. Transforming growth factor beta 1 stimulates vascular endothelial growth factor gene transcription in human cholangiocellular carcinoma cells. Cancer Res. 2003;63:1083–92.PubMed Benckert C, Jonas S, Cramer T, et al. Transforming growth factor beta 1 stimulates vascular endothelial growth factor gene transcription in human cholangiocellular carcinoma cells. Cancer Res. 2003;63:1083–92.PubMed
50.
Zurück zum Zitat Dickson MC, Martin JS, Cousins FM, et al. Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. Development. 1995;121:1845–54.PubMed Dickson MC, Martin JS, Cousins FM, et al. Defective haematopoiesis and vasculogenesis in transforming growth factor-beta 1 knock out mice. Development. 1995;121:1845–54.PubMed
51.
Zurück zum Zitat Oshima M, Oshima H, Taketo MM. TGF-beta receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis. Dev Biol. 1996;179:297–302.PubMedCrossRef Oshima M, Oshima H, Taketo MM. TGF-beta receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis. Dev Biol. 1996;179:297–302.PubMedCrossRef
52.
Zurück zum Zitat Li DY, Sorensen LK, Brooke BS, et al. Defective angiogenesis in mice lacking endoglin. Science. 1999;284:1534–7.PubMedCrossRef Li DY, Sorensen LK, Brooke BS, et al. Defective angiogenesis in mice lacking endoglin. Science. 1999;284:1534–7.PubMedCrossRef
53.
Zurück zum Zitat Larsson J, Goumans MJ, Sjostrand LJ, et al. Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptor-deficient mice. EMBO J. 2001;20:1663–73.PubMedCrossRef Larsson J, Goumans MJ, Sjostrand LJ, et al. Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptor-deficient mice. EMBO J. 2001;20:1663–73.PubMedCrossRef
Metadaten
Titel
Low-Level Expression of Smad7 Correlates with Lymph Node Metastasis and Poor Prognosis in Patients with Pancreatic Cancer
verfasst von
Peng Wang, MD
Jie Fan, MD
Zhen Chen, MD
Zhi-Qiang Meng, MD
Jian-Min Luo, PhD
Jun-Hua Lin, MD
Zhen-Hua Zhou, MD
Hao Chen, MD
Kun Wang, MD
Zu-De Xu, MD
Lu-Ming Liu, MD
Publikationsdatum
01.04.2009
Verlag
Springer-Verlag
Erschienen in
Annals of Surgical Oncology / Ausgabe 4/2009
Print ISSN: 1068-9265
Elektronische ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-008-0284-5

Weitere Artikel der Ausgabe 4/2009

Annals of Surgical Oncology 4/2009 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.