1,921
Views
34
CrossRef citations to date
0
Altmetric
Editorial

Stathmin 1: a protein with many tasks. New biomarker and potential target in cancer

, MD
Pages 631-634 | Published online: 12 Jun 2012

Abstract

Stathmin 1 (STMN1) is a critical protein involved in microtubule polymerization and is necessary for survival of cancer cells. This editorial describes the role of targeted therapeutics which disrupt STMN1 modulation and such effect on cancer survival.

1. Introduction

The ‘many tasks' of Stathmin 1 (STMN1) were recently described by Belletti and Baldassarre Citation[1]. The name stathmin is derived from the term ‘stathmos', the Greek word for ‘relay'. It represents stathmin's role as a critical intermediate during signal transduction in modulation and control of microtubule polymerization. STMN1 is a protein composed of 149 amino acids organized into four domains (I – IV) as defined by limited proteolysis. The core region (amino acids 42 – 126) is the minimum fragment required for tubulin interaction with the additional requirement of either an N- or C-terminal extension Citation[2]. There are four phosphorylation domains, designated as Ser 16, 25, 38, and 63. Of the four phosphorylation sites, only Ser 16 is conserved throughout the STMN1 family.

2. STMN1 function

Modulation of STMN1 Citation[3] can result in mitotic arrest, thereby exhibiting a critical role in microtubule dynamics. Microtubules are protein polymers comprising α/β tubulin heterodimers, which contribute to and are essential for the structure and function of the cell. These functions include intracellular transport, cell motility, and polarity. Dynamics of microtubule function can best be described as an alternating pattern of stabilization and destabilization. Stathmin-mediated destabilization can result from either tubulin sequestration or ‘catastrophe'. The latter results from microtubule depolymerization and is counterbalanced by ‘rescue', which is effected by polymerization. The transition between the two phases during the various portions of the cell cycle is regulated by microtubule-stabilizing and microtubule-destabilizing proteins Citation[4]. The process of mitotic spindle formation is a coordinated, balanced interaction between the stabilizing activities of microtubule-associated proteins (XMAP215, EB1), motor proteins (predominantly kinesin, e.g., Eg-5), and plus-end depolymerases, including XKCM1, MCAK and STMN1 Citation[5]. A tightly regulated sequenced pattern of STMN1 phosphorylation and de-phosphorylation is necessary for entry into prophase and, terminally, into cytokinesis, respectively Citation[6,7]. As recently pointed out by Belletti and Baldassarre Citation[1], these functions are critical for malignant cell survival.

It is also postulated that during the metaphase to anaphase transition, stathmin effects poleward kinetochore spindle movement by increasing minus-end catastrophe frequency Citation[8]. Moreover, it has been shown that exogenous STMN1 affects metaphase-to-anaphase transition by its role in kinetochore-associated microtubule detachment during anaphase, thereby resulting in chromosomal instability with over a 100-fold increase in micronucleus formation Citation[9]. To exit mitosis and allow for cytokinesis, the microtubules undergo depolymerization, which requires the dephosphorylation of stathmin reactivating its tubulin-binding property.

3. Relevance of STMN1 to cancer

Decreased expression of p27 has been linked to prognosis in a number of tumor types. Besides being a CDK inhibitor of Cyclin D1, p27 has been shown to play a role in cell motility Citation[10]. Baldassare recently showed that stathmin is a p27-binding partner, and on the basis of his data postulates that p27 interferes with stathmin binding and sequestration of tubulin, consequently inhibiting cell motility and microtubule depolymerization. Furthermore, p27 is downregulated in transformed cells. Low p27 and high STMN1 correlate with metastatic behavior of sarcoma cells in vivo Citation[10]. Similar results have been shown in other cancers Citation[11,12].

Based on differential proteogenomic signaling assessments of patients from our program Citation[13,14], as well as published literature by others, there is a sound rationale for the therapeutic targeting of STMN1 in cancer patients Citation[15]. Stathmin is highly expressed in a variety of human malignancies Citation[16,17] and has been shown to be upregulated in multiple cancers. Moreover, upregulated stathmin has been shown to be extensively correlated with poor survival. In one particular analysis involving 1,076 endometrial cancer specimens, stathmin overexpression was significantly associated with poor survival of the patients and with nodal metastasis Citation[18].

The E2F sites in the stathmin promoter also appear involved in the regulation of stathmin via activity of c-JUN and FoxM1 Citation[19]. Negative regulators of stathmin involved in cancer have also been identified. For instance, induction of WT p53 results in decreased stathmin expression via direct transcriptional repression Citation[20], possibly involving p21WAF1 and EGR-1 Citation[21].

Stathmin expression and function have also been shown to be controlled at the post-transcriptional level with involvement of cancer-related proteins. In colorectal cancer cells, overexpression of PUMA, a p53-induced effector of apoptosis Citation[22], results in stathmin degradation via a proteasome-dependent pathway. This finding is further supported by the fact that in some tumor samples the expression levels of stathmin mRNA and protein are uncoupled Citation[23], suggesting that this mechanism may be important in some pathological conditions. More widely accepted, the modulation of stathmin activity can be achieved by protein sequestration. The CDK inhibitor p27Kip1 Citation[24] and the transcription factor STAT3 Citation[25] are both able to bind stathmin, therefore preventing its ability to sequester free αβ-tubulin heterodimers.

Several studies have demonstrated that stathmin depletion results in cell cycle arrest and induction of apoptosis, thereby playing a significant role in malignant cell death. This is further supported by the observation that stathmin is a target of ASK1-p38 and JNK kinases, signals involved in response to cellular stress. Stathmin has also shown to be protective of arsenic- Citation[26] and paclitaxel-induced apoptosis Citation[19,27]. It is also interesting to note that in human cancer stathmin expression may influence sensitivity/resistance to treatment with selected cytotoxic drugs. Several studies in vitro indicate that tumor-derived cell lines with high stathmin expression negatively influence the response to microtubule targeting drugs Citation[19,28].

4. Potential STMN1-targeting therapeutics

A variety of target-specific anti-stathmin effectors, including ribozymes Citation[29] and si-RNA Citation[16,30,] have been used to silence stathmin in vitro as singlets Citation[16,29,30] and in combination with chemotherapeutic agents where additive synergistic interactions have been demonstrated (i.e., taxanes) Citation[31-33]. Both ribozyme and siRNA inhibition of stathmin mRNA result in an increase in G2M phase cell population, an inhibition of clonogenicity, and a marked increase in apoptosis Citation[16,30,34]. The latter may be due to the effect of modulation of microtubule network mobility on the proportion of Bax/Bcl-2 and Bax/Bcl-xL heterodimers Citation[35,36]. More recently, we have demonstrated marked (93%) knockdown of STMN1 using a novel bifunctional RNA interference technology Citation[37]. We have shown a 5-log dosing improvement in growth inhibition involving CCL-247 colon cancer cells comparing bi-shRNAi STMN1 to siRNAi STMN1 targeting the identical mRNA sequence and in coordination with cleavage product quantitation. These results justified efficacy testing in murine models, which demonstrated response and survival advantage in multiple tumor models following systemic treatment with bi-shRNAi STMN1 LP Citation[38]. These results, combined with safety demonstrated by toxicity and biodistribution studies in STMN1 biorelevant pigs, enabled Phase I clinical investigation to be opened with a novel bi-shRNAi STMN1 LP therapeutic.

5. Conclusion

STMN1 is extensively involved in signal transduction of malignant cells. Experimental therapeutic modalities, such as bi-shRNAi STMN1 LP, may one day contribute to cancer management as a novel STMN1 down-modulator. A Phase I clinical trial has recently been initiated to test the effect of STMN1 knockdown on cancer response. Chemotherapy modalities, such as taxanes, have broad clinical reach, commonly involving breast cancer, lung cancer, ovarian cancer, prostate cancer, sarcoma, and gastric cancer to name a few. The mechanism of action by which taxanes mediate antitumor activity involves microtubule polymerization. It is likely that initial clinical opportunities with STMN1 knockdown will involve combination with taxanes.

Declaration of interest

The author cofounded Gradalis, Inc. and is a shareholder. Gradalis has in development a novel bifunctional RNA interference based nanoparticle technology targeting STMN1.

Bibliography

  • Belletti B, Baldassarre G. Stathmin: a protein with many tasks. New biomarker and potential target in cancer. Expert Opin Ther Targets 2011;15(11):1249-66
  • Cassimeris L. The oncoprotein 18/stathmin family of microtubule destabilizers. Curr Opin Cell Biol 2002;14(1):18-24
  • Wang F, Wang LX, Li SL, Downregulation of stathmin is involved in malignant phenotype reversion and cell apoptosis in esophageal squamous cell carcinoma. J Surg Oncol 2011;103(7):704-15
  • Mistry SJ, Bank A, Atweh GF. Targeting stathmin in prostate cancer. Mol Cancer Ther 2005;4(12):1821-9
  • Niethammer P, Kronja I, Kandels-Lewis S, Discrete states of a protein interaction network govern interphase and mitotic microtubule dynamics. PLoS Biol 2007;5(2):e29
  • Iancu-Rubin C, Nasrallah CA, Atweh GF. Stathmin prevents the transition from a normal to an endomitotic cell cycle during megakaryocytic differentiation. Cell Cycle (Georgetown, Tex) 2005;4(12):1774-82
  • Gavet O, Ozon S, Manceau V, The stathmin phosphoprotein family: intracellular localization and effects on the microtubule network. J Cell Sci 1998;111(Pt 22):3333-46
  • Manna T, Thrower D, Miller HP, Stathmin strongly increases the minus end catastrophe frequency and induces rapid treadmilling of bovine brain microtubules at steady state in vitro. J Biol Chem 2006;281(4):2071-8
  • Holmfeldt P, Sellin ME, Gullberg M. Upregulated Op18/stathmin activity causes chromosomal instability through a mechanism that evades the spindle assembly checkpoint. Exp Cell Res 2010;316(12):2017-26
  • Baldassarre G, Belletti B, Nicoloso MS, p27(Kip1)-stathmin interaction influences sarcoma cell migration and invasion. Cancer Cell 2005;7(1):51-63
  • Jeon TY, Han ME, Lee YW, Overexpression of stathmin1 in the diffuse type of gastric cancer and its roles in proliferation and migration of gastric cancer cells. Br J Cancer 2010;102(4):710-18
  • Zheng P, Liu YX, Chen L, Stathmin, a new target of PRL-3 identified by proteomic methods, plays a key role in progression and metastasis of colorectal cancer. J Proteome Res 2010;9(10):4897-905
  • Nemunaitis J, Senzer N, Khalil I, Proof concept for clinical justification of network mapping for personalized cancer therapeutics. Cancer Gene Ther 2007;14(8):686-95
  • Shen Y SN, Nemunaitis J. Use of proteomics analysis for molecular precision approaches in cancer therapy. Drug Target Insights 2008;3:12
  • Rana S, Maples PB, Senzer N, Nemunaitis J. Stathmin 1: a novel therapeutic target for anticancer activity. Expert Rev Anticancer Ther 2008;8(9):1461-70
  • Alli E, Yang JM, Hait WN. Silencing of stathmin induces tumor-suppressor function in breast cancer cell lines harboring mutant p53. Oncogene 2007;26(7):1003-12
  • Nishio K, Nakamura T, Koh Y, Oncoprotein 18 overexpression increases the sensitivity to vindesine in the human lung carcinoma cells. Cancer 2001;91(8):1494-9
  • Trovik J, Wik E, Stefansson IM, Stathmin overexpression identifies high-risk patients and lymph node metastasis in endometrial cancer. Clin Cancer Res 2011;17(10):3368-77
  • Carr JR, Park HJ, Wang Z, FoxM1 mediates resistance to herceptin and paclitaxel. Cancer Res 2010;70(12):5054-63
  • Ahn J, Murphy M, Kratowicz S, Down-regulation of the stathmin/Op18 and FKBP25 genes following p53 induction. Oncogene 1999;18(43):5954-8
  • Fang L, Min L, Lin Y, Downregulation of stathmin expression is mediated directly by Egr1 and associated with p53 activity in lung cancer cell line A549. Cell Signal 2010;22(1):166-73
  • Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 2001;7(3):683-94
  • Belletti B, Nicoloso MS, Schiappacassi M, Stathmin activity influences sarcoma cell shape, motility, and metastatic potential. Mol Biol Cell 2008;19(5):2003-13
  • Schiappacassi M, Lovisa S, Lovat F, Role of T198 modification in the regulation of p27(Kip1) protein stability and function. PLoS One 2011;6(3):e17673
  • Verma NK, Dourlat J, Davies AM, STAT3-stathmin interactions control microtubule dynamics in migrating T-cells. J Biol Chem 2009;284(18):12349-62
  • Wang X, Ren JH, Lin F, Stathmin is involved in arsenic trioxide-induced apoptosis in human cervical cancer cell lines via PI3K linked signal pathway. Cancer Biol Ther 2010;10(6):632-43
  • Jiang L, Chen Y, Chan CY, Down-regulation of stathmin is required for TGF-beta inducible early gene 1 induced growth inhibition of pancreatic cancer cells. Cancer Lett 2009;274(1):101-8
  • Su D, Smith SM, Preti M, Stathmin and tubulin expression and survival of ovarian cancer patients receiving platinum treatment with and without paclitaxel. Cancer 2009;115(11):2453-63
  • Mistry SJ, Benham CJ, Atweh GF. Development of ribozymes that target stathmin, a major regulator of the mitotic spindle. Antisense Nucleic Acid Drug Dev 2001;11(1):41-9
  • Zhang HZ, Wang Y, Gao P, Silencing stathmin gene expression by survivin promoter-driven siRNA vector to reverse malignant phenotype of tumor cells. Cancer Biol Ther 2006;5(11):1457-61
  • Wang R, Dong K, Lin F, Inhibiting proliferation and enhancing chemosensitivity to taxanes in osteosarcoma cells by RNA interference-mediated downregulation of stathmin expression. Mol Med (Cambridge, Mass) 2007;13(11-12):567-75
  • Mistry SJ, Atweh GF. Therapeutic interactions between stathmin inhibition and chemotherapeutic agents in prostate cancer. Mol Cancer Ther 2006;5(12):3248-57
  • Ngo TT, Peng T, Liang XJ, The 1p-encoded protein stathmin and resistance of malignant gliomas to nitrosoureas. J Natl Cancer Inst 2007;99(8):639-52
  • Iancu C, Mistry SJ, Arkin S, Effects of stathmin inhibition on the mitotic spindle. J Cell Sci 2001;114(Pt 5):909-16
  • Longuet M, Serduc R, Riva C. Implication of bax in apoptosis depends on microtubule network mobility. Int J Oncol 2004;25(2):309-17
  • Singer S, Ehemann V, Brauckhoff A, Protumorigenic overexpression of stathmin/Op18 by gain-of-function mutation in p53 in human hepatocarcinogenesis. Hepatology (Baltimore, Md) 2007;46(3):759-68
  • Rao DD, Maples PB, Senzer N, Enhanced target gene knockdown by a bifunctional shRNA: a novel approach of RNA interference. Cancer Gene Ther 2010;17(11):780-91
  • Phadke AP, Jay CM, Wang Z, In vivo safety and antitumor efficacy of bifunctional small hairpin RNAs specific for the human stathmin 1 oncoprotein. DNA Cell Biol 2011;30(9):715-26

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.