Next Article in Journal
Treatment of Parkinson’s Disease through Personalized Medicine and Induced Pluripotent Stem Cells
Next Article in Special Issue
Tubulin βII and βIII Isoforms as the Regulators of VDAC Channel Permeability in Health and Disease
Previous Article in Journal
Mechanism of Siponimod: Anti-Inflammatory and Neuroprotective Mode of Action
Previous Article in Special Issue
Synthesis and Biological Evaluation of Novel Triple-Modified Colchicine Derivatives as Potent Tubulin-Targeting Anticancer Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Over-Expression of βII-Tubulin and Especially Its Localization in Cell Nuclei Correlates with Poorer Outcomes in Colorectal Cancer

1
N.N. Alexandrov National Cancer Centre of Belarus, 223040 Minsk, Belarus
2
Department of Pathology, Belarusian State Medical University, 220116 Minsk, Belarus
3
Minsk City Clinical Oncologic Dispensary, 220013 Minsk, Belarus
4
Department of Gastroenterology and Nutrition, Belarusian Medical Academy of Post-Graduate Education, 220013 Minsk, Belarus
5
Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
*
Author to whom correspondence should be addressed.
Cells 2019, 8(1), 25; https://doi.org/10.3390/cells8010025
Submission received: 17 December 2018 / Accepted: 2 January 2019 / Published: 7 January 2019
(This article belongs to the Special Issue Tubulin: Structure, Functions and Roles in Disease)

Abstract

:
Tubulin is a heterodimer of α and β subunits, both existing as isotypes differing in amino acid sequence encoded by different genes. Specific isotypes of tubulin have associations with cancer that are not well understood. Previous studies found that βII-tubulin is expressed in a number of transformed cells and that this isotype is found in cell nuclei in non-microtubule form. The association of βII expression and its nuclear localization with cancer progression has not previously been addressed. We here used a monoclonal antibody to βII to examine patients with colorectal cancer and found that patients whose tumors over-express βII have a greatly decreased life expectancy which is even shorter in those patients with nuclear βII. Our results suggest that βII-tubulin may facilitate cancer growth and metastasis and, to accomplish this, may not need to be in microtubule form. Furthermore, βII expression and localization could be a useful prognostic marker. We also found that βII appears in the nuclei of otherwise normal cells adjacent to the tumor. It is possible therefore that cancer cells expressing βII influence nearby cells to do the same and to localize βII in their nuclei by an as yet uncharacterized regulatory pathway.

1. Introduction

Colorectal cancer (CRC) is the third most common cancer in men and the second in women in the United States [1]. According to the World Health Organization statistics, CRC is the fourth most common cause of death from cancer after female breast cancer, prostate cancer in men, and lung cancer, with more than 1.4 million expected cases of incidence every year [2]. Expected morbidity and mortality in 2035 are 2.4 and 1.3 million new cases, respectively [3]. Tubulin, the subunit protein of microtubules [4,5], is an α/β heterodimer [6]. Both α and β exist as isotypes differing in amino acid sequence and encoded by different genes [7,8]. Considerable evidence has accumulated that tubulin can exist in cells in non-microtubule forms [9,10,11,12,13,14]. This is especially the case for the βII isotype, which often occurs in cell nuclei, possibly in the form of a reticulum, but not as a microtubule [9]. Specific nuclear localization of βII-tubulin was demonstrated not only by immunohistochemistry using a monoclonal antibody to βII [15], but also by immunoblotting of a purified nuclear fraction, and by the fact that fluorescently labeled αβII-tubulin when micro-injected into these cells, went into the nuclei whereas fluorescently labeled, micro-injected αβIII and αβIV did not [9]. Nuclear βII is particularly common in cancer cells, less common in cultured cells, and much less common in normal cells in situ [16,17,18]. βII is commonly over-expressed in tumors where the normal cells express little or no βII [18]. This is especially significant because βII is also the tubulin isotype with which some of the most successful anti-tumor drugs, namely, paclitaxel and vinblastine, interact the best [19,20].
We previously showed that βII is expressed in cells excised from the tumors of patients with CRC, both metastatic and non-metastatic [18]. Normal colon expresses little or no βII [21,22]. We also showed that βII occurs in the nuclei of cells excised from the tumors of patients with CRC, as well as many other cancers [18]. Although the previous study only examined samples from 15 patients with CRC, no difference was observed in βII localization in metastatic and non-metastatic disease [18]. However, in some other cancers, it appeared that nuclear βII was most likely to occur in metastatic tumors [18].
In this study, we used a monoclonal antibody to βII-tubulin [15] to examine surgical material from patients with CRC and measured the life expectancies of the patients. We found that over-expression of βII was correlated with a shorter life expectancy of patients with CRC. The life expectancy was even shorter for patients in whose tumors βII was localized to the cell nuclei. We also found that otherwise normal cells close to the tumor also expressed βII and localized it to their nuclei.
Our results have both cell biological and clinical implications. They suggest that there exists an as yet uncharacterized pathway whereby βII is synthesized and localized to nuclei in both cancer cells and in nearby normal cells, somehow influenced by the cancer, and that this pathway may be correlated with increased aggressiveness of the cancer. Our findings also raise questions about the role of βII tubulin in both normal and cancerous cells. From a clinical perspective, these results also have implications about the possible prognostic utility for patients with CRC of βII expression and nuclear localization. Furthermore, the current understanding of how anti-tubulin drugs operate in cancer is that they freeze microtubule dynamics [23,24,25]. If tubulin, in non-microtubule form, is affecting cancer progression then that understanding needs to be expanded. In other words, the fact that βII apparently exists in advanced CRC in non-microtubule form raises the possibility that tubulin does not need to be in a microtubule to promote cancer cell growth and proliferation and that non-microtubule tubulin may constitute a novel and hitherto unexplored target for cancer chemotherapy, and may even have a function in normal cells.

2. Materials and Methods

2.1. Source of Patients

Investigations were carried out following the rules of the Declaration of Helsinki of 1975, revised in 2013. The research was approved by the Ethics Committee of the Belarusian State Medical University prior to commencing the study. The study included 124 patients (55 male, 69 female, median age 65.0 years old, q1–q3 57.0–73.0 years old) with CRC (See Table S1 in Supplementary Material). All patients had had a bowel resection performed by the same surgeon in Minsk City Clinical Oncological Dispensary in 2009–2011. Patients’ follow up was carried out according to the national protocols. Progression was defined as tumor growth after radical resection of the bowel segment. The median time of dynamic follow-up was 3.56 years (q1–q2 1.2–4.4 years, maximum 5.5 years). Medical examinations were conducted once every six months during the first two years after operation and once a year after two years after operation. Only patients who signed the informed consent were included. Results were not available to clinicians at the time of patients’ treatment and follow-up.

2.2. Tissue Samples

The tissue specimens were dissected from the edge of the tumors. Pathological analysis was performed on resected specimens and staged according to the American Joint Committee on Cancer [26]. For the purpose of the research the blocks containing the deepest invasive margin were selected. Non-tumor colonic mucosa or mucosa at a distance of no less than 1 cm from the tumor were used as a control group (60 cases). Features of the tumors are presented in Table S2 in the Supplementary Material.

2.3. Immunohistochemistry

Paraffin-embedded tissues (4-μm-thick) were cut from tissue blocks and mounted on slides coated with 3-aminopropylenetriethoxy-silane, then deparaffinized in xylene and rehydrated in alcohol. Endogenous peroxidase activity was inhibited by 3% hydrogen peroxide for 20 min. After washing twice in 0.05 M Tris buffered saline (TBS, Sigma Aldrich, Darmstadt, Germany) non-specific binding was blocked using 1% bovine serum albumin in TBS for 30 min. Antigen retrieval was carried out in a Pascal Pressure Cooker (Dako, Carpinteria, CA, USA) at 125 °C and 25 psi for 30 s with 0.01 M citrate buffer (pH 6.0). At the next step slides were incubated with primary anti-mouse monoclonal antibodies to βII-tubulin (clone JDR3B8, IgG2b-isotype, 1:40, BioGenex, Fremont, CA, USA) at 4 °C overnight. This antibody does not distinguish between the different forms of βII-tubulin (βIIA, βIIB and βIIC). Subsequently they were washed twice with TBS and then staining was detected by Super SensitiveTM Polymer-HRP IHC Detection System (BioGenex, Fremont, CA, USA). Diaminobenzidine (DAB, DAKO, Glostrup, Denmark) was used as a chromogen. Then slides were counter-stained with hematoxylin and mounted in Cytoseal (ThermoScientific, Waltham, MA, USA). Nerve fibers and nerve ganglia were used as a positive inner control due to their intense reactivity.

2.4. Immunohistochemistry Evaluation

Tissue samples were analyzed using a Leica DM5000 B microscope at ×200 magnification. βII-tubulin expression was evaluated separately in the central regions of the tumor and in the deepest invasive margin. The invasive front was defined as the deepest invasive margin of the tumor within one field of vision (×200). Moreover, expression was assessed both in the nuclei and in the cytoplasm. The nerve trunks served as an internal positive control. Positive immunoperoxidase staining of nerves for βII has been previously reported for a variety of tissues [18,21]. Slides stained by immunohistochemistry without using primary antibody served as a negative control. Immunohistochemical staining was interpreted as either positive or negative and all cases were classified in groups according to the absence or presence of βII-tubulin expression. Those cases in which any number of cells showed positive nuclear or cytoplasmic staining were classified as positive while those cases with absence of brown staining were classified as negative. Most of the cases (76%) were negative for βII-tubulin. We did not observe any signs of non-specific staining as a “side effect”, for example. Usually positive staining was clearly localized to the cytoplasm or nucleus or both. During acquisition of the images the pathologist was blinded to any clinical or staging data. We previously used the same immunohistochmical approach to examine the distribution of βIII-tubulin in CRC [27].

2.5. Statistical Analysis

Image analysis was performed using RStudio, v. 0.98.1103 (RStudio, Inc., Boston, MA, USA). Groups were compared using the Wilcoxon test (pw). Survival curves for different groups were obtained using the Kaplan-Meier estimator and then compared by a log-rank test (plr). Null hypothesis was rejected at р < 0.05.

2.6. Data Sharing

All the figures, original data, and protocols are available, although patient identifying information is not. Please contact Dr. Portyanko for such information.

3. Results

βII-tubulin expression was detected in 30 cases of CRC (28.0%). Cell cytoplasm showed positive staining in all the tumors (30 cases—28.0%). Furthermore, in 14 of these 30 cases βII-tubulin was present in the nuclei (14 cases—11.2%). (Figure 1A). To highlight the difference in cytoplasmic staining and nuclear staining for βII-tubulin, the samples are shown at higher magnification in Figure 2 to illustrate both cytoplasmic (Figure 2A) and nuclear (Figure 2B) staining of βII-tubulin.
In most cases fewer than 5% of the tumor cells demonstrated unambiguous expression of βII-tubulin; the intensity of this staining was variable but its presence was unambiguous. Positively stained cancer cells were either concentrated as small βII-positive foci or were diffusely scattered across the tumor (Figure 1B). No mitotic spindles containing βII-tubulin were observed. The averaged number of mitoses for every case varied from 0 to 11.3. The number of mitoses in the groups with or without βII-tubulin expression did not show a statistically significant difference either in the center (p = 0.64) or in the invasive front (p = 0.85). It was not clear if βII was forming microtubules. Both the area of the tumor center (30 tumors—24.0%) and the area of the tumor invasive front (19 tumors—15.2%) exhibited positively stained regions. (Table 1).
Malignant cells expressed βII-tubulin exclusively in the tumor center in 56.7% (17/30 tumors) of all cases while in 20.0% (6/30 tumors) expression was detected only in the invasive front. In the other cases both the center and the invasive front showed positive staining. The Wilcoxon matched pairs test did not reveal a statistically significant difference between the expression of βII-tubulin in the center and in the invasive front (pw = 0.1075). It should be noted that the expression of this isotype in the invasive front was associated with higher probability of disease progression. The results showed that the presence of βII was associated with decreased survival, a pattern that was even more striking when βII was in the nuclei. This difference was revealed both for cytoplasmic (p = 0.0168) and nuclear (p = 0.0000) patterns of immunostaining (Figure 3). The log rank test showed that there was no difference in survival time between the groups with and without the expression of βII-tubulin either in the cytoplasm (p = 0.452) or in the nuclei (p = 0.245) of cancer cells in the tumor center.
One of the observations in our study was that 24 of 58 tumors containing adjacent normal mucosa also had positive nuclear βII-staining in this otherwise normal area. However, the resection margins, which are parts of apparently non-tumorous bowel mucosa after surgical resection of bowel segment with a tumor, did not reveal positive nuclear or cytoplasmic βII-staining (Figure 1C).

4. Discussion

The findings reported here raise questions of both clinical and biological significance. The data clearly show that over-expression of βII in CRC is associated with shortened survival and that this is even more pronounced for patients with nuclear βII. It is interesting that the shapes of the two curves are different (Figure 3). Survival for the first 400 days is not very different between βII over-expression and localization of βII in the nucleus, but after 400 days, survival drops off very steeply for the latter group, to the point where very few survive after 1200 days. Granted that there are other mechanisms for prognosticating survival in patients with CRC [28,29], then, at the very least, observation of βII may provide, if the test is further developed, another predictive tool, one requiring only a biopsy. Furthermore, it is conceivable that the presence of nuclear βII may indicate somewhat poorer survival overall with even poorer survival after 500 days; this kind of high-resolution prognostication may be useful for patients.
At this point, it is worth mentioning that some studies have found that mRNA levels for the two forms of βII-tubulin (βIIa and βIIb) are very low in certain colon cancer cell lines [30] and in some cases of CRC [31,32]. However, there is also evidence for over-expression of βII-tubulin in other cases of CRC and other cancers, sometimes associated with increased drug resistance [33,34,35]. In either case, we must recall that mRNA expression and protein expression are not always correlated and that, if we extrapolate from studies suggesting that expression of βII mRNA may be low in the cancers we have studied, this putative discrepancy raises the possibility that the issue in these tumors is one of decreased degradation of βII. Future studies may resolve this issue.
It is not yet clear what functions βII and nuclear βII serve for cancer cells. Some evidence in the literature raises the possibility that βII-tubulin may be involved in membrane rearrangements [11,36,37], some of which may involve microtubules [38]. Since cancer cells grow, divide, and migrate, it is reasonable to speculate that new membrane is often being made or rearranged [39] and hence that βII could be useful in this regard, perhaps mediating microtubule-membrane connections. This could account for the over-expression of βII that has been observed in CRC and a large number of other cancers [18]. The function of nuclear βII in cancer cells, however, remains a mystery. There is evidence, however, that nuclear βII can interact with antitumor drugs, such as paclitaxel and vinblastine [17,40,41], indicating that, if nuclear βII is indeed present in CRC and other tumors, then chemotherapeutic strategies may need to take this into account.
The observation that otherwise normal cells adjacent to the tumor express βII, including nuclear βII (Figure 1C), has been made before for a variety of tumors [18], but not in any quantitative manner, as we have done here. There are three implications of this finding. First, if a tumor is searched for in a biopsy, then if the probe misses the actual tumor, observation of cells containing βII and, even more strikingly, nuclear βII, would imply the nearby presence of a tumor, which could powerfully augment the utility of a biopsy. Second, the mechanism by which βII is made in otherwise normal cells adjacent to the tumor and localized to the nuclei is unknown. Our observations imply the existence of a hitherto unknown signaling pathway that affects tubulin biosynthesis and subcellular localization. Perhaps the pathway involves production of a substance by the tumor that influences the nearby cells to behave in this way. Third, the mechanism by which such a substance might enter the normal cells is also unknown. It may require a nanotubule of some kind. Further exploration of these mechanisms could not only add to our knowledge of basic cellular regulatory pathways, but also reveal hitherto unsuspected targets for novel chemotherapies.

Supplementary Materials

The following supplementary materials are available online.

Author Contributions

Author contributions were as follows: Conceptualization, A.P.; Data curation, A.M., G.T., and J.G.; Formal analysis, K.R.; Funding acquisition, A.P.; Investigation, A.M., A.N., T.B., and A.P.; Methodology, A.P.; Project administration, A.P.; Resources, G.T. and J.G.; Software, K.R.; Supervision, A.P.; Validation, A.P.; Visualization, K.R.; Writing—original draft, K.R.; Writing—review and editing, R.L.

Funding

This work was financially supported by the International Science and Technology Center (B-1636) and State Program of Scientific Investigation of Belarus (No. 1.2.42).

Acknowledgments

We would like to thank Consuelo Walss-Bass for her suggestions and insights.

Conflicts of Interest

Ludueña owns 500,000 shares in OncoVista Innovative Technology, a company that is attempting to develop novel anti-tumor drugs; he is also on their Scientific Advisory Board. In addition he has licensed isotype-specific monoclonal antibodies to tubulin to various companies and receives royalties from the sales. The other authors declare no conflict of interest. The founding sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.

References

  1. U.S. Cancer Statistics Working Group. United States Cancer Statistics, 1999–2014; Incidence and Mortality Web-based Report; Department of Health and Human Services, Centers for Disease Control and Prevention and National Cancer Institute: Atlanta, GA, USA, 2017. Available online: www.cdc.gov/uscs (accessed on 1 January 2019).
  2. Stewart, B.W.; Wild, C.P. World Cancer Report 2014; International Agency for Research on Cancer: Lyon, France, 2014; ISBN 978-92-832-0429-9. [Google Scholar]
  3. Ferlay, J.; Soerjomataram, I.; Ervik, M.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11, 2013; International Agency for Research on Cancer: Lyon, France, 2013; Available online: http://globocan.iarc.fr (accessed on 6 February 2015).
  4. Dustin, P. Microtubules, 2nd ed.; Springer: Berlin, Germany, 1984; ISBN 3-540-13283-X. [Google Scholar]
  5. Kreis, T.; Vale, R. Guidebook to the Cytoskeletal and Motor Proteins, 2nd ed.; Oxford University Press: Oxford, UK, 1999; ISBN-13 978-0198599562. [Google Scholar]
  6. Ludueña, R.F.; Shooter, E.M.; Wilson, L. The structure of the tubulin dimer. J. Biol. Chem. 1977, 252, 7006–7014. [Google Scholar] [PubMed]
  7. Ludueña, R.F.; Banerjee, A. The isotypes of tubulin: Distribution and functional significance. In Cancer Drug Discovery and Development: The Role of Microtubules in Cell Biology, Neurobiology, and Oncology; Fojo, T., Ed.; Humana Press: Totowa, NJ, USA, 2008; pp. 123–175. ISBN 978-1-59745-336-3. [Google Scholar]
  8. Ludueña, R.F.; Yeh, I.T.; Joe, P.; Guo, J. Functional significance of vertebrate β-tubulin isotypes. Meditsina 2011, 72, 50–57. (In Russian) [Google Scholar]
  9. Walss, C.; Kreisberg, J.I.; Luduena, R.F. Presence of the βII-isotype of tubulin in the nuclei of cultured rat kidney mesangial cells. Cell Motil. Cytoskeleton 1999, 42, 274–284. [Google Scholar] [CrossRef]
  10. Walss-Bass, C.; Prasad, V.; Kreisberg, J.I.; Ludueña, R.F. Interaction of the βIV-tubulin isotype with actin stress fibers in cultured rat kidney mesangial cells. Cell Motil. Cytoskeleton 2001, 49, 200–207. [Google Scholar] [CrossRef] [PubMed]
  11. Kourmouli, N.; Dialynas, G.; Petraki, C.; Pyrpasopoulou, A.; Singh, P.M.; Georgatos, S.D.; Theodoropoulos, P.A. Binding of heterochromatin protein 1 to the nuclear envelope is regulated by a soluble form of tubulin. J. Biol. Chem. 2001, 276, 13007–13014. [Google Scholar] [CrossRef] [PubMed]
  12. Carré, M.; André, N.; Carles, G.; Borghi, H.; Brichese, L.; Briand, C.; Braguer, D. Tubulin is an inherent component of mitochondrial membranes that interacts with the voltage-dependent anion channel. J. Biol. Chem. 2002, 277, 33664–33669. [Google Scholar] [CrossRef] [PubMed]
  13. Yanagida, M.; Hayano, T.; Yamauchi, Y.; Shinkawa, T.; Natsume, T.; Isobe, T.; Takahashi, N. Human fibrillarin forms a sub-complex with splicing factor 2-associated p32, protein arginine methyltransferases, and tubulins α3 and β1 that is independent of its association with preribosomal ribonucleoprotein complexes. J. Biol. Chem. 2004, 279, 1607–1614. [Google Scholar] [CrossRef]
  14. McIntosh, J.R.; Grishchuk, E.; Morphew, M.K.; Efremov, A.K.; Zhudenkov, K.; Volkov, V.A.; Cheeseman, I.M.; Desai, A.; Mastronarde, D.N.; Ataullakhanov, A.I. Fibrils connect microtubule tips with kinetochores: A mechanism to couple tubulin dynamics to chromosome motion. Cell 2008, 135, 322–333. [Google Scholar] [CrossRef]
  15. Banerjee, A.; Roach, M.C.; Wall, K.A.; Lopata, M.A.; Cleveland, D.W.; Ludueña, R.F. A monoclonal antibody against the type II isotype of β-tubulin. Preparation of isotypically altered tubulin. J. Biol. Chem. 1988, 263, 3029–3034. [Google Scholar]
  16. Walss-Bass, C.; Xu, K.; David, S.; Fellous, A.; Ludueña, R.F. Occurrence of nuclear βII-tubulin in cultured cells. Cell Tissue Res. 2002, 308, 215–223. [Google Scholar] [CrossRef]
  17. Xu, K.; Ludueña, R.F. Characterization of nuclear βII-tubulin in tumor cells: A possible novel target for taxol. Cell Motil. Cytoskeleton 2002, 53, 39–52. [Google Scholar] [CrossRef] [PubMed]
  18. Yeh, I.-T.; Ludueña, R.F. The βII isotype of tubulin is present in the cell nuclei of a variety of cancers. Cell Motil. Cytoskeleton 2004, 57, 96–106. [Google Scholar] [CrossRef] [PubMed]
  19. Derry, W.B.; Wilson, L.; Khan, I.A.; Ludueña, R.F.; Jordan, M.A. Taxol differentially modulates the dynamics of microtubules assembled from unfractionated and purified β-tubulin isotypes. Biochemistry 1997, 36, 3554–3562. [Google Scholar] [CrossRef] [PubMed]
  20. Khan, I.A.; Ludueña, R.F. Different effects of vinblastine on the polymerization of isotypically purified tubulins from bovine brain. Investig. New Drugs 2003, 21, 3–13. [Google Scholar] [CrossRef] [PubMed]
  21. Roach, M.C.; Boucher, V.L.; Walss, C.; Ravdin, P.M.; Ludueña, R.F. Preparation of a monoclonal antibody specific for the class I isotype of β-tubulin: The β isotypes of tubulin differ in their cellular distributions within human tissues. Cell Motil. Cytoskeleton 1998, 39, 273–285. [Google Scholar] [CrossRef]
  22. Oda, E.; Nakamura, Y.; Yamamoto, M.; Kojiro, M. Immunohistochemical distribution of tubulin beta II in human normal and neoplastic tissues. Kurume Med. J. 2005, 52, 117–125. [Google Scholar] [CrossRef] [PubMed]
  23. Dhamodharan, R.; Jordan, M.A.; Thrower, D.; Wilson, L.; Wadsworth, P. Vinblastine suppresses dynamics of individual microtubules in living interphase cells. Mol. Biol. Cell 1995, 6, 1215–1229. [Google Scholar] [CrossRef]
  24. Yvon, A.C.; Wadsworth, P.; Jordan, M.A. Taxol suppresses dynamics of individual microtubules in living human tumor cells. Mol. Biol. Cell 1999, 10, 947–959. [Google Scholar] [CrossRef]
  25. Rathinasamy, K.; Jindal, B.; Asthana, J.; Singh, P.; Balaji, P.V.; Panda, D. Griseofulvin stabilizes microtubule dynamics, activates p53 and inhibits the proliferation of MCF-7 cells synergistically with vinblastine. BMC Cancer 2010, 10, 213. [Google Scholar] [CrossRef]
  26. Edge, S.B.; Byrd, D.R.; Compton, C.C.; Fritz, A.G.; Greene, F.L.; Trotti, A. AJCC Cancer Staging Manual, 7th ed.; Springer: New York, NY, USA, 2009; ISBN-13 978-1845974749. [Google Scholar]
  27. Portyanko, A.; Kovalev, P.; Gorgun, J.; Cherstvoy, E. Beta(III)-tubulin at the invasive margin of colorectal cancer: Possible link to invasion. Virchows Archiv. 2009, 454, 541–548. [Google Scholar] [CrossRef]
  28. Compton, C.C.; Fielding, L.P.; Burgart, L.J.; Conley, B.; Cooper, H.S.; Hamilton, S.R.; Hammond, M.E.H.; Henson, D.E.; Hutter, R.V.P.; Nagle, R.B.; et al. Prognostic factors in colorectal cancer: College of American Pathologists Consensus Statement 1999. Arch. Pathol. Lab. Med. 2000, 124, 979–994. [Google Scholar] [CrossRef] [PubMed]
  29. De Divitiis, C.; Nasti, G.; Montano, M.; Fisichella, R.; Iaffaioli, R.V.; Berretta, M. Prognostic and predictive response factors in colorectal cancer patients: Between hope and reality. World J. Gastroenterol. 2014, 20, 15049–15059. [Google Scholar] [CrossRef] [PubMed]
  30. Hiser, L.; Aggarwal, A.; Young, R.; Frankfurter, A.; Spano, A.; Correia, J.J.; Lobert, S. Comparison of β-tubulin mRNA and protein levels in 12 human cancer cell lines. Cell Motil. Cytoskeleton 2006, 63, 41–52. [Google Scholar] [CrossRef] [PubMed]
  31. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.U.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio Cancer Genomics Portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed]
  32. Gao, J.; Aksov, B.A.; Dogrusog, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 2, 6. [Google Scholar] [CrossRef] [PubMed]
  33. LaBonte, M.J.; Wilson, P.M.; Fazzone, W.; Groshen, S.; Lenz, H.J.; Ladner, R.D. DNA microarray profiling of genes differentially regulated by the histone deacetylase inhibitors vorinostat and LBH589 in colon cancer cell lines. BMC Med. Genom. 2009, 2, 67. [Google Scholar] [CrossRef] [PubMed]
  34. Dessein, A.F.; Stechly, L.; Jonckheere, N.; Dumont, P.; Monté, D.; Leteurtre, E.; Truant, S.; Pruvot, F.R.; Figeac, M.; Hebbar, M.; et al. Autocrine induction of invasive and metastatic phenotypes by the MIF-CRCR4 axis in drug-resistant human colon cancer cells. Cancer Res. 2010, 70, 4644–4654. [Google Scholar] [CrossRef]
  35. Cirillo, L.; Gotta, M.; Meraldi, P. The elephant in the room: The role of microtubules in cancer. In Cell Division Machinery and Disease; Gotta, M., Meraldi, P., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 93–124. ISBN 978-3-319-57125-6. [Google Scholar]
  36. Guo, J.; Walss-Bass, C.; Ludueña, R.F. The beta isotypes of tubulin in neuronal differentiation. Cytoskeleton 2010, 67, 431–441. [Google Scholar] [CrossRef]
  37. Guo, J.; Qiang, M.; Ludueña, R.F. The distribution of β-tubulin isotypes in cultured neurons from embryonic, newborn, and adult mouse brains. Brain Res. 2011, 1420, 8–18. [Google Scholar] [CrossRef]
  38. Georgatos, S.D.; Theodoropoulos, P.A. Rules to remodel by: What drives nuclear envelope disassembly and reassembly during mitosis? Crit. Rev. Eukaryot. Gene Expr. 1999, 9, 373–381. [Google Scholar] [CrossRef]
  39. Rashid, A.; Pizer, E.S.; Moga, M.; Milgraum, L.Z.; Zahurak, M.; Pasternack, G.R.; Kuhajda, F.P.; Hamilton, S.R. Elevated expression of fatty acid synthase and fatty acid synthetic activity in colorectal neoplasia. Am. J. Pathol. 1997, 150, 201–208. [Google Scholar] [PubMed]
  40. Rowinsky, E.K.; Donehower, R.C. Drug therapy: Paclitaxel (Taxol). N. Engl. J. Med. 1995, 332, 1004–1014. [Google Scholar] [CrossRef] [PubMed]
  41. Walss-Bass, C.; Kreisberg, J.I.; Ludueña, R.F. Effect of the anti-tumor drug vinblastine on nuclear βII-tubulin in cultured rat kidney mesangial cells. Investig. New Drugs 2003, 21, 15–26. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immunohistochemical staining of βII-tubulin in colorectal cancer specimen detected by peroxidase-mediated diaminobenzidine (DAB)-staining (brown) with arrows indicating epithelial compartments of colorectal cancer (CRC) (1) and stroma (2). (A) Epithelial compartment of tumor cells showing moderate cytoplasmic and more intense nuclear βII-tubulin staining. There are single positive stromal cells (original magnification ×200). (B) Adenocarcinoma at lower magnification (original magnification ×100). There are single positive stromal cells. (C) Normal colonic mucosa (left) adjacent to tumor complexes (right) showing appearance of positive nuclear βII staining (original magnification ×200).
Figure 1. Immunohistochemical staining of βII-tubulin in colorectal cancer specimen detected by peroxidase-mediated diaminobenzidine (DAB)-staining (brown) with arrows indicating epithelial compartments of colorectal cancer (CRC) (1) and stroma (2). (A) Epithelial compartment of tumor cells showing moderate cytoplasmic and more intense nuclear βII-tubulin staining. There are single positive stromal cells (original magnification ×200). (B) Adenocarcinoma at lower magnification (original magnification ×100). There are single positive stromal cells. (C) Normal colonic mucosa (left) adjacent to tumor complexes (right) showing appearance of positive nuclear βII staining (original magnification ×200).
Cells 08 00025 g001
Figure 2. Immunohistochemical staining of βII-tubulin in CRC specimen detected by peroxidase-mediated DAB-staining (brown) with arrows indicating epithelial compartment of CRC (1) and stroma (2). (A) Epithelial compartment of tumor cells showing moderate cytoplasmic βII-staining, (original magnification ×400). (B) Epithelial compartment of tumor cells showing strong focal nuclear βII-staining, (original magnification ×400).
Figure 2. Immunohistochemical staining of βII-tubulin in CRC specimen detected by peroxidase-mediated DAB-staining (brown) with arrows indicating epithelial compartment of CRC (1) and stroma (2). (A) Epithelial compartment of tumor cells showing moderate cytoplasmic βII-staining, (original magnification ×400). (B) Epithelial compartment of tumor cells showing strong focal nuclear βII-staining, (original magnification ×400).
Cells 08 00025 g002
Figure 3. Progression-free survival in patients as a function of the presence of βII-positive staining in the invasive front. (A) The progression-free survival is decreased in patients with the presence of cytoplasmic βII-tubulin in the invasive front (plr = 0.0168). (B) Patients with the presence of nuclear βII tubulin staining in the invasive front demonstrate worse prognosis in comparison with patients without positive staining in the nuclei (plr < 0.001).
Figure 3. Progression-free survival in patients as a function of the presence of βII-positive staining in the invasive front. (A) The progression-free survival is decreased in patients with the presence of cytoplasmic βII-tubulin in the invasive front (plr = 0.0168). (B) Patients with the presence of nuclear βII tubulin staining in the invasive front demonstrate worse prognosis in comparison with patients without positive staining in the nuclei (plr < 0.001).
Cells 08 00025 g003
Table 1. βII-tubulin expression in colorectal cancer (CRC).
Table 1. βII-tubulin expression in colorectal cancer (CRC).
Intensity of StainingβII-Tubulin Expression
Tumor CenterInvasive FrontNormal Mucosa
N%N%N%
Negative9576.010684.83458.6
Positive3024.01915.22441.4
All12510012510058100

Share and Cite

MDPI and ACS Style

Ruksha, K.; Mezheyeuski, A.; Nerovnya, A.; Bich, T.; Tur, G.; Gorgun, J.; Luduena, R.; Portyanko, A. Over-Expression of βII-Tubulin and Especially Its Localization in Cell Nuclei Correlates with Poorer Outcomes in Colorectal Cancer. Cells 2019, 8, 25. https://doi.org/10.3390/cells8010025

AMA Style

Ruksha K, Mezheyeuski A, Nerovnya A, Bich T, Tur G, Gorgun J, Luduena R, Portyanko A. Over-Expression of βII-Tubulin and Especially Its Localization in Cell Nuclei Correlates with Poorer Outcomes in Colorectal Cancer. Cells. 2019; 8(1):25. https://doi.org/10.3390/cells8010025

Chicago/Turabian Style

Ruksha, Kseniya, Artur Mezheyeuski, Alexander Nerovnya, Tatyana Bich, Gennady Tur, Julia Gorgun, Richard Luduena, and Anna Portyanko. 2019. "Over-Expression of βII-Tubulin and Especially Its Localization in Cell Nuclei Correlates with Poorer Outcomes in Colorectal Cancer" Cells 8, no. 1: 25. https://doi.org/10.3390/cells8010025

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop