325
Views
0
CrossRef citations to date
0
Altmetric
Commentary

Pancreatic cancer with Nest-in tendencies

&
Pages 559-561 | Received 14 Jan 2010, Accepted 15 Jan 2010, Published online: 15 Mar 2011

Abstract

Commentary to:

Nestin is a novel target for suppressing pancreatic cancer cell migration, invasion and metastasis

Yoko Matsuda, Zenya Naito, Kiyoko Kawahara, Nando Nakazawa, Murray Korc and Toshiyuki Ishiwata

In the previous issue of Cancer Biology & Therapy, Matsuda et al. present a role for Nestin in pancreatic ductal adenocarcinoma (PDAC) migration, invasion and metastasis.Citation1 Nestin, a class VI intermediate filament, has been implicated in cancer progression in a variety of tissues including the pancreas.Citation2Citation8 Nestin was originally identified as a neural stem cell marker and it is implicated in stem cell self-renewal.Citation9,Citation10 However, the exact role of Nestin in cancer progression and the possible contribution of the stem cell in this context is unknown.

Although Nestin is an intermediate filament protein, its cellular functions are diversified beyond the structural (summarized in ). Nestin is expressed during embryogenesis in actively proliferating precursor cells of the central nervous system and muscle.Citation9,Citation11,Citation12 Nestin is downregulated in differentiated cells but remains present in small populations of cells in adult tissues, and it is upregulated during tissue regeneration. These observations highlight a role for Nestin in cellular proliferation, and they also led to the hypothesis that Nestin is a progenitor/stem cell-specific protein. Nestin knockout mice are embryonic-lethal and have fewer neural stem cells in the developing neural tube.Citation10 Neural stem cells derived from Nestin knock-out mice have decreased self-renewal capacity but do not show any differences in cellular proliferation or, interestingly, any effects on cytoskeleton integrity.Citation10 Nestin has been identified in other tissues undergoing differentiation and/or regeneration including the testes, teeth, pancreas and kidney,Citation13Citation17 and it is also implicated in neo-vascularization in the hair follicle, skin and colon.Citation18Citation20 The wide variety of tissues in which Nestin is important suggests a conserved function in stemness or regeneration. However, Nestin can also contribute to cancer progression.

Cancer cell growth mirrors the proliferative aspects of both tissue regeneration and stem cell self-renewal. The cancer stem cell hypothesis proposes that a transformed progenitor/stem cell, with its infinite regenerative capacity, can give rise to a tumor. Furthermore, the cancer stem cell must be targeted therapeutically in order for the tumor to ultimately disappear. Nestin expression has been correlated with tumor severity in neuroblastomas, gliomas, melanomas and prostate cancers.Citation2,Citation4,Citation7 Given its expression patterns in stem cells, regenerating tissues and neovascularization, its correlation with tumor severity aligns with the cancer stem cell hypothesis. However, its direct roles in tumor progression are unknown.

There are other hypotheses for how Nestin may contribute to tumor progression in addition to its potential role in maintaining (cancer) stem cell self-renewal. Tumors require blood vessel infiltration or angiogenesis in order to survive, and Nestin is expressed in the endothelial cells that generate the vasculature.Citation20,Citation21 However, this data is correlative, and the necessity for and mechanism of action of Nestin in tumor angiogenesis has yet to be tested. The most-studied hypothesis for how Nestin contributes to tumor progression is that it impacts cell motility. Nestin correlates with tumor severity, and aggressive tumors are characterized by higher levels of invasion, migration and metastasis.

Kleeberger et al. reported that while Nestin did not affect prostate cancer cell viability or proliferation, it was necessary for cell motility and invasion in vitro.Citation4 Furthermore, Nestin appeared to mediate prostate cancer cell migration and metastasis in xenograft experiments in vivo. These experiments emphasized a role for Nestin in cell motility rather than proliferation. Nestin is also implicated in melanoma cell invasion, although its contribution has not been challenged directly.Citation22 It is also a potential marker for circulating melanoma cells, which may serve a prognostic purpose. 23 Kawamoto et al. previously showed that Nestin was expressed in ∼30% of pancreatic cancer patient samples and did not correlate with prognosis or survival.Citation24 However, they observed Nestin expression at sites of peripheral nerve fibers and adjacent stromal tissue, suggesting that Nestin may contribute to a tumor micro-environment that promotes invasion. As in the examples above, much of the data surrounding the role of Nestin in cancer progression is correlative, and mechanistic studies are needed.

The study by Matsuda et al. focuses on the role of Nestin in pancreatic ductal adenocarcinoma (PDAC) invasion.Citation1 Pancreatic cancer is the fourth-deadliest cancer in American men and it has a 5-year survival rate of only 5–6%.Citation25 PDAC is the most common presentation of pancreatic cancer, accounting for ~85% of cases.Citation26 It is a highly invasive cancer, and it rapidly metastasizes to the lymphatic system, spleen, liver and lungs at very early stages in tumor development.Citation26 It is commonly treated with surgery and/or gemcitabine, sometimes in combination with oxaliplatin, but response to chemotherapy is very poor and new regimens need to be developed that impede the rapid tumor cell dissemination that is characteristic of this tumor.

Because Nestin is expressed in ∼30% of PDAC cases and is implicated in cellular invasion and motility, Matsuda et al. hypothesized that Nestin contributes to PDAC invasion, migration and metastasis.Citation1 They transfected two PDAC cell lines (PANC-1 and PK-45H) with scrambled control or Nestin shRNA. Nestin knockdown led to increased cell-cell contact and elevated levels of F-actin at the cell periphery. Nestin knockdown had no effect on PDAC growth potential in vitro or subcutaneously in vivo. However, Nestin contributed to PDAC cell migration and invasion in vitro according to scratch assays, Boyden chamber assays and modified Boyden chamber assays with matrigel.

To investigate a possible mechanism behind Nestin-mediated migration and invasion, the authors performed a PCR array for genes associated with metastasis. The most highly upregulated gene in Nestin knockdown cells relative to control cells was E-cadherin. Immunofluorescence experiments revealed that E-cadherin was localized to cell-cell junctions at the cell membrane of Nestin knockdown PDAC cells. E-cadherin is best characterized as a mediator of epithelial-to-mesenchymal transition (EMT), which is often a prerequisite for cellular invasion and migration. During EMT initiation, the transcription factors Twist, Slug and Snail suppress E-cadherin to allow decreased cell-cell adhesion. Twist was undetectable in the PDAC cells, but Slug mRNA was increased in Nestin knockdown PANC-1 cells relative to control cells and Snail mRNA was decreased in PK-45H cells. Slug levels did not change in the Nestin knockdown PK-45H cells compared to the control, while Snail levels did not change in the Nestin knockdown PANC-1 cells. The disparity in expression of these three transcription factors suggests that E-cadherin is regulated in myriad ways, and the mechanistic connection between Nestin and E-cadherin remains unknown. Finally, the authors observed decreased liver metastases of Nestin knockdown PANC-1 cells compared to control six weeks after injecting the cells into the spleens of mice. Of note, the proliferative capacities of the metastatic outgrowths were the same in the Nestin knockdown cells and the control cells, according to Ki-67 immunostaining. Together, this report shows that Nestin contributes to PDAC cell invasion, migration and metastatic capability, but not to cellular proliferation. Furthermore, Nestin expression negatively correlates with E-cadherin expression, and an absence of E-cadherin may promote EMT and the invasive phenotype.

This report corroborates those that present a role for Nestin in cell invasion and migration but not proliferation, which is of particular interest for studying a highly metastatic cancer such as PDAC. It proposes a relationship between Nestin, an intermediate filament and E-cadherin, an adhesion protein implicated in EMT. Nestin expression was also found to be inversely correlated with E-cadherin expression in aggressive anaplastic thyroid carcinoma cells.Citation27 Interestingly, E-cadherin was recently implicated as a moderator of neural stem cell self-renewal, but in that setting E-cadherin expression was positively-correlated with Nestin-positive neural stem cells.Citation28 The mechanistic functions of Nestin are likely complex, and they are sure to differ with cellular context. For all of the hypotheses and initial evidence for the multiple ways that Nestin may impact cancer progression, mechanistic proof is still needed and Matsuda et al. are up for the challenge.

Figures and Tables

Table 1 Supporting evidence for nestin involvement in various biofunctions

Commentary to:

References

  • Matsuda Y, Naito Z, Kawahara K, Nakazawa N, Korc M, Ishiwata T. Nestin is a novel target for suppressing pancratic cancer cell migration, invasion and metastasis. Cancer Biol Ther 2011; 11:512 - 523
  • Yang XH, Wu QL, Yu XB, Xu CX, Ma BF, Zhang XM, et al. Nestin expression in different tumours and its relevance to malignant grade. J Clin Pathol 2008; 61:467 - 473
  • Chen Z, Wang T, Luo H, Lai Y, Yang X, Li F, et al. Expression of Nestin in lymph node metastasis and lymphangiogenesis in non-small cell lung cancer patients. Hum Pathol 2010; 41:737 - 744
  • Kleeberger W, Bova GS, Nielsen ME, Herawi M, Chuang AY, Epstein JI, et al. Roles for the stem cell associated intermediate filament Nestin in prostate cancer migration and metastasis. Cancer Res 2007; 67:9199 - 9206
  • Li H, Cherukuri P, Li N, Cowling V, Spinella M, Cole M, et al. Nestin is expressed in the basal/myoepithelial layer of the mammary gland and is a selective marker of basal epithelial breast tumors. Cancer Res 2007; 67:501 - 510
  • Lim YC, Oh SY, Cha YY, Kim SH, Jin X, Kim H. Cancer stem cell traits in squamospheres derived from primary head and neck squamous cell carcinomas. Oral Oncol 2011; 47:83 - 91
  • Piras F, Perra MT, Murtas D, Minerba L, Floris C, Maxia C, et al. The stem cell marker Nestin predicts poor prognosis in human melanoma. Oncol Rep 2010; 23:17 - 24
  • Krupkova O Jr, Loja T, Zambo I, Veselska R. Nestin expression in human tumors and tumor cell lines. Neoplasma 2010; 57:291 - 298
  • Lendahl U, Zimmerman LB, McKay RD. CNS stem cells express a new class of intermediate filament protein. Cell 1990; 60:585 - 595
  • Park D, Xiang AP, Mao FF, Zhang L, Di CG, Liu XM, et al. Nestin is required for the proper self-renewal of neural stem cells. Stem Cells 2010; 28:2162 - 2171
  • Kachinsky AM, Dominov JA, Miller JB. Myogenesis and the intermediate filament protein, Nestin. Dev Biol 1994; 165:216 - 228
  • Sejersen T, Lendahl U. Transient expression of the intermediate filament Nestin during skeletal muscle development. J Cell Sci 1993; 106:1291 - 1300
  • Frojdman K, Pelliniemi LJ, Lendahl U, Virtanen I, Eriksson JE. The intermediate filament protein Nestin occurs transiently in differentiating testis of rat and mouse. Differentiation 1997; 61:243 - 249
  • Terling C, Rass A, Mitsiadis TA, Fried K, Lendahl U, Wroblewski J. Expression of the intermediate filament Nestin during rodent tooth development. Int J Dev Biol 1995; 39:947 - 956
  • Kim SY, Lee SH, Kim BM, Kim EH, Min BH, Bendayan M, et al. Activation of Nestin-positive duct stem (NPDS) cells in pancreas upon neogenic motivation and possible cytodifferentiation into insulin-secreting cells from NPDS cells. Dev Dyn 2004; 230:1 - 11
  • Ishiwata T, Kudo M, Onda M, Fujii T, Teduka K, Suzuki T, et al. Defined localization of Nestin-expressing cells in L-arginine-induced acute pancreatitis. Pancreas 2006; 32:360 - 368
  • Vansthertem D, Gossiaux A, Decleves AE, Caron N, Nonclercq D, Legrand A, et al. Expression of Nestin, vimentin and NCAM by renal interstitial cells after ischemic tubular injury. J Biomed Biotechnol 2010; 2010:193 - 259
  • Amoh Y, Li L, Yang M, Moossa AR, Katsuoka K, Penman S, et al. Nascent blood vessels in the skin arise from Nestin-expressing hair-follicle cells. Proc Natl Acad Sci USA 2004; 101:13291 - 13295
  • Aki R, Amoh Y, Li L, Katsuoka K, Hoffman RM. Nestin-expressing interfollicular blood vessel network contributes to skin transplant survival and wound healing. J Cell Biochem 2010; 110:80 - 86
  • Teranishi N, Naito Z, Ishiwata T, Tanaka N, Furukawa K, Seya T, et al. Identification of neovasculature using Nestin in colorectal cancer. Int J Oncol 2007; 30:593 - 603
  • Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA. Proliferation of immature tumor vessels is a novel marker of clinical progression in prostate cancer. Cancer Res 2009; 69:4708 - 4715
  • Florenes VA, Holm R, Myklebost O, Lendahl U, Fodstad O. Expression of the neuroectodermal intermediate filament Nestin in human melanomas. Cancer Res 1994; 54:354 - 356
  • Fusi A, Ochsenreither S, Busse A, Rietz A, Keilholz U. Expression of the stem cell marker Nestin in peripheral blood of patients with melanoma. Br J Dermatol 2010; 163:107 - 114
  • Kawamoto M, Ishiwata T, Cho K, Uchida E, Korc M, Naito Z, et al. Nestin expression correlates with nerve and retroperitoneal tissue invasion in pancreatic cancer. Hum Pathol 2009; 40:189 - 198
  • Jemal A, Siegel R, Xu J, Ward E. Cancer statistics. CA Cancer J Clin 2010; 60:277 - 300
  • Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, Depinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2006; 20:1218 - 1249
  • Liu J, Brown RE. Immunohistochemical detection of epithelialmesenchymal transition associated with stemness phenotype in anaplastic thyroid carcinoma. Int J Clin Exp Pathol 2010; 3:755 - 762
  • Karpowicz P, Willaime-Morawek S, Balenci L, DeVeale B, Inoue T, van der Kooy D. E-Cadherin regulates neural stem cell self-renewal. J Neurosci 2009; 29:3885 - 3896

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.