Skip to main content
Top
Published in: Breast Cancer Research and Treatment 3/2011

01-10-2011 | Preclinical study

Local anesthetics inhibit kinesin motility and microtentacle protrusions in human epithelial and breast tumor cells

Authors: Jennifer R. Yoon, Rebecca A. Whipple, Eric M. Balzer, Edward H. Cho, Michael A. Matrone, Michelle Peckham, Stuart S. Martin

Published in: Breast Cancer Research and Treatment | Issue 3/2011

Login to get access

Abstract

Detached breast tumor cells produce dynamic microtubule protrusions that promote reattachment of cells and are termed tubulin microtentacles (McTNs) due to their mechanistic distinctions from actin-based filopodia/invadopodia and tubulin-based cilia. McTNs are enriched with vimentin and detyrosinated α-tubulin, (Glu-tubulin). Evidence suggests that vimentin and Glu-tubulin are cross-linked by kinesin motor proteins. Using known kinesin inhibitors, Lidocaine and Tetracaine, the roles of kinesins in McTN formation and function were tested. Live-cell McTN counts, adhesion assays, immunofluorescence, and video microscopy were performed to visualize inhibitor effects on McTNs. Viability and apoptosis assays were used to confirm the non-toxicity of the inhibitors. Treatments of human non-tumorigenic mammary epithelial and breast tumor cells with Lidocaine or Tetracaine caused rapid collapse of vimentin filaments. Live-cell video microscopy demonstrated that Tetracaine reduces motility of intracellular GFP-kinesin and causes centripetal collapse of McTNs. Treatment with Tetracaine inhibited the extension of McTNs and their ability to promote tumor cell aggregation and reattachment. Lidocaine showed similar effects but to a lesser degree. Our current data support a model in which the inhibition of kinesin motor proteins by Tetracaine leads to the reductions in McTNs, and provides a novel mechanism for the ability of this anesthetic to decrease metastatic progression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Nicolson GL, Fidler IJ, Poste G (1986) Effects of tertiary amine local anesthetics on the blood-borne implantation and cell surface properties of metastatic mouse melanoma cells. J Natl Cancer Inst 76:511–519PubMed Nicolson GL, Fidler IJ, Poste G (1986) Effects of tertiary amine local anesthetics on the blood-borne implantation and cell surface properties of metastatic mouse melanoma cells. J Natl Cancer Inst 76:511–519PubMed
2.
go back to reference Whipple RA, Cheung AM, Martin SS (2007) Detyrosinated microtubule protrusions in suspended mammary epithelial cells promote reattachment. Exp Cell Res 313:1326–1336PubMedCrossRef Whipple RA, Cheung AM, Martin SS (2007) Detyrosinated microtubule protrusions in suspended mammary epithelial cells promote reattachment. Exp Cell Res 313:1326–1336PubMedCrossRef
3.
go back to reference Whipple RA, Balzer EM, Cho EH, Matrone MA, Yoon JR, Martin SS (2008) Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cells. Cancer Res 68:5678–5688PubMedCrossRef Whipple RA, Balzer EM, Cho EH, Matrone MA, Yoon JR, Martin SS (2008) Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cells. Cancer Res 68:5678–5688PubMedCrossRef
4.
go back to reference Korb T, Schluter K, Enns A, Spiegel HU, Senninger N, Nicolson GL, Haier J (2004) Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res 299:236–247PubMedCrossRef Korb T, Schluter K, Enns A, Spiegel HU, Senninger N, Nicolson GL, Haier J (2004) Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res 299:236–247PubMedCrossRef
5.
go back to reference Machesky LM (2008) Lamellipodia and filopodia in metastasis and invasion. FEBS Lett 582(14):2102–2111 Machesky LM (2008) Lamellipodia and filopodia in metastasis and invasion. FEBS Lett 582(14):2102–2111
6.
go back to reference Donaldson DJ, Dunlap MK (1981) Epidermal cell migration during attempted closure of skin wounds in the adult newt: observations based on cytochalasin treatment and scanning electron microscopy. J Exp Zool 217:33–43PubMedCrossRef Donaldson DJ, Dunlap MK (1981) Epidermal cell migration during attempted closure of skin wounds in the adult newt: observations based on cytochalasin treatment and scanning electron microscopy. J Exp Zool 217:33–43PubMedCrossRef
7.
go back to reference Frisch SM, Francis H (1994) Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol 124:619–626PubMedCrossRef Frisch SM, Francis H (1994) Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol 124:619–626PubMedCrossRef
8.
go back to reference Reed JC (1998) Dysregulation of apoptosis in cancer. Cancer J Sci Am 4(Suppl 1):S8–S14 Reed JC (1998) Dysregulation of apoptosis in cancer. Cancer J Sci Am 4(Suppl 1):S8–S14
9.
go back to reference Mialhe A, Lafanechere L, Treilleux I, Peloux N, Dumontet C, Bremond A, Panh MH, Payan R, Wehland J, Margolis RL, Job D (2001) Tubulin detyrosination is a frequent occurrence in breast cancers of poor prognosis. Cancer Res 61:5024–5027PubMed Mialhe A, Lafanechere L, Treilleux I, Peloux N, Dumontet C, Bremond A, Panh MH, Payan R, Wehland J, Margolis RL, Job D (2001) Tubulin detyrosination is a frequent occurrence in breast cancers of poor prognosis. Cancer Res 61:5024–5027PubMed
10.
go back to reference Webster DR, Gundersen GG, Bulinski JC, Borisy GG (1987) Assembly and turnover of detyrosinated tubulin in vivo. J Cell Biol 105:265–276PubMedCrossRef Webster DR, Gundersen GG, Bulinski JC, Borisy GG (1987) Assembly and turnover of detyrosinated tubulin in vivo. J Cell Biol 105:265–276PubMedCrossRef
11.
go back to reference Janmey PA, Euteneuer U, Traub P, Schliwa M (1991) Viscoelastic properties of vimentin compared with other filamentous biopolymer networks. J Cell Biol 113:155–160PubMedCrossRef Janmey PA, Euteneuer U, Traub P, Schliwa M (1991) Viscoelastic properties of vimentin compared with other filamentous biopolymer networks. J Cell Biol 113:155–160PubMedCrossRef
12.
go back to reference Gurland G, Gundersen GG (1995) Stable, detyrosinated microtubules function to localize vimentin intermediate filaments in fibroblasts. J Cell Biol 131:1275–1290PubMedCrossRef Gurland G, Gundersen GG (1995) Stable, detyrosinated microtubules function to localize vimentin intermediate filaments in fibroblasts. J Cell Biol 131:1275–1290PubMedCrossRef
13.
14.
go back to reference Vale RD, Reese TS, Sheetz MP (1985) Identification of a novel force-generating protein, kinesin, involved in microtubule-based motility. Cell 42:39–50PubMedCrossRef Vale RD, Reese TS, Sheetz MP (1985) Identification of a novel force-generating protein, kinesin, involved in microtubule-based motility. Cell 42:39–50PubMedCrossRef
15.
go back to reference Coy DL, Hancock WO, Wagenbach M, Howard J (1999) Kinesin’s tail domain is an inhibitory regulator of the motor domain. Nat Cell Biol 1:288–292PubMedCrossRef Coy DL, Hancock WO, Wagenbach M, Howard J (1999) Kinesin’s tail domain is an inhibitory regulator of the motor domain. Nat Cell Biol 1:288–292PubMedCrossRef
16.
17.
go back to reference Kreitzer G, Liao G, Gundersen GG (1999) Detyrosination of tubulin regulates the interaction of intermediate filaments with microtubules in vivo via a kinesin-dependent mechanism. Mol Biol Cell 10:1105–1118PubMed Kreitzer G, Liao G, Gundersen GG (1999) Detyrosination of tubulin regulates the interaction of intermediate filaments with microtubules in vivo via a kinesin-dependent mechanism. Mol Biol Cell 10:1105–1118PubMed
18.
go back to reference Liao G, Gundersen GG (1998) Kinesin is a candidate for cross-bridging microtubules and intermediate filaments. Selective binding of kinesin to detyrosinated tubulin and vimentin. J Biol Chem 273:9797–9803PubMedCrossRef Liao G, Gundersen GG (1998) Kinesin is a candidate for cross-bridging microtubules and intermediate filaments. Selective binding of kinesin to detyrosinated tubulin and vimentin. J Biol Chem 273:9797–9803PubMedCrossRef
19.
go back to reference Fink BR, Kennedy RD, Hendrickson AE, Middaugh ME (1972) Lidocaine inhibition of rapid axonal transport. Anesthesiology 36:422–432PubMedCrossRef Fink BR, Kennedy RD, Hendrickson AE, Middaugh ME (1972) Lidocaine inhibition of rapid axonal transport. Anesthesiology 36:422–432PubMedCrossRef
20.
go back to reference Richards CD (1978) The action of anaesthetics on synaptic transmission. Gen Pharmacol 9:287–293PubMed Richards CD (1978) The action of anaesthetics on synaptic transmission. Gen Pharmacol 9:287–293PubMed
21.
go back to reference Miyamoto Y, Muto E, Mashimo T, Iwane AH, Yoshiya I, Yanagida T (2000) Direct inhibition of microtubule-based kinesin motility by local anesthetics. Biophys J 78:940–949PubMedCrossRef Miyamoto Y, Muto E, Mashimo T, Iwane AH, Yoshiya I, Yanagida T (2000) Direct inhibition of microtubule-based kinesin motility by local anesthetics. Biophys J 78:940–949PubMedCrossRef
22.
go back to reference Spector I, Shochet NR, Kashman Y, Groweiss A (1983) Latrunculins: novel marine toxins that disrupt microfilament organization in cultured cells. Science 219:493–495PubMedCrossRef Spector I, Shochet NR, Kashman Y, Groweiss A (1983) Latrunculins: novel marine toxins that disrupt microfilament organization in cultured cells. Science 219:493–495PubMedCrossRef
23.
go back to reference Casella JF, Flanagan MD, Lin S (1981) Cytochalasin D inhibits actin polymerization and induces depolymerization of actin filaments formed during platelet shape change. Nature 293:302–305PubMedCrossRef Casella JF, Flanagan MD, Lin S (1981) Cytochalasin D inhibits actin polymerization and induces depolymerization of actin filaments formed during platelet shape change. Nature 293:302–305PubMedCrossRef
24.
go back to reference Martin SS, Leder P (2001) Human MCF10A mammary epithelial cells undergo apoptosis following actin depolymerization that is independent of attachment and rescued by Bcl-2. Mol Cell Biol 21:6529–6536PubMedCrossRef Martin SS, Leder P (2001) Human MCF10A mammary epithelial cells undergo apoptosis following actin depolymerization that is independent of attachment and rescued by Bcl-2. Mol Cell Biol 21:6529–6536PubMedCrossRef
25.
go back to reference Gyoeva FK, Gelfand VI (1991) Coalignment of vimentin intermediate filaments with microtubules depends on kinesin. Nature 353:445–448PubMedCrossRef Gyoeva FK, Gelfand VI (1991) Coalignment of vimentin intermediate filaments with microtubules depends on kinesin. Nature 353:445–448PubMedCrossRef
26.
go back to reference Prahlad V, Yoon M, Moir RD, Vale RD, Goldman RD (1998) Rapid movements of vimentin on microtubule tracks: kinesin-dependent assembly of intermediate filament networks. J Cell Biol 143:159–170PubMedCrossRef Prahlad V, Yoon M, Moir RD, Vale RD, Goldman RD (1998) Rapid movements of vimentin on microtubule tracks: kinesin-dependent assembly of intermediate filament networks. J Cell Biol 143:159–170PubMedCrossRef
27.
go back to reference Dunn S, Morrison EE, Liverpool TB, Molina-Paris C, Cross RA, Alonso MC, Peckham M (2008) Differential trafficking of Kif5c on tyrosinated and detyrosinated microtubules in live cells. J Cell Sci 121:1085–1095PubMedCrossRef Dunn S, Morrison EE, Liverpool TB, Molina-Paris C, Cross RA, Alonso MC, Peckham M (2008) Differential trafficking of Kif5c on tyrosinated and detyrosinated microtubules in live cells. J Cell Sci 121:1085–1095PubMedCrossRef
28.
go back to reference Ingber DE (2002) Mechanical signaling and the cellular response to extracellular matrix in angiogenesis and cardiovascular physiology. Circ Res 91:877–887PubMedCrossRef Ingber DE (2002) Mechanical signaling and the cellular response to extracellular matrix in angiogenesis and cardiovascular physiology. Circ Res 91:877–887PubMedCrossRef
29.
go back to reference Ingber DE (2003) Tensegrity II. How structural networks influence cellular information processing networks. J Cell Sci 116:1397–1408PubMedCrossRef Ingber DE (2003) Tensegrity II. How structural networks influence cellular information processing networks. J Cell Sci 116:1397–1408PubMedCrossRef
30.
go back to reference Blick T, Widodo E, Hugo H, Waltham M, Lenburg ME, Neve RM, Thompson EW (2008) Epithelial mesenchymal transition traits in human breast cancer cell lines. Clin Exp Metastasis 25:629–642PubMedCrossRef Blick T, Widodo E, Hugo H, Waltham M, Lenburg ME, Neve RM, Thompson EW (2008) Epithelial mesenchymal transition traits in human breast cancer cell lines. Clin Exp Metastasis 25:629–642PubMedCrossRef
31.
go back to reference Matrone MA, Whipple RA, Thompson K, Cho EH, Vitolo MI, Balzer EM, Yoon JR, Ioffe OB, Tuttle KC, Tan M, Martin SS (2010) Metastatic breast tumors express increased tau, which promotes microtentacle formation and the reattachment of detached breast tumor cells. Oncogene 1–11 Matrone MA, Whipple RA, Thompson K, Cho EH, Vitolo MI, Balzer EM, Yoon JR, Ioffe OB, Tuttle KC, Tan M, Martin SS (2010) Metastatic breast tumors express increased tau, which promotes microtentacle formation and the reattachment of detached breast tumor cells. Oncogene 1–11
32.
go back to reference Hammond JW, Huang CF, Kaech S, Jacobson C, Banker G, Verhey KJ (2010) Posttranslational modifications of tubulin and the polarized transport of kinesin-1 in neurons. Mol Biol Cell 21:572–583 Hammond JW, Huang CF, Kaech S, Jacobson C, Banker G, Verhey KJ (2010) Posttranslational modifications of tubulin and the polarized transport of kinesin-1 in neurons. Mol Biol Cell 21:572–583
33.
go back to reference Krylyshkina O, Kaverina I, Kranewitter W, Steffen W, Alonso MC, Cross RA, Small JV (2002) Modulation of substrate adhesion dynamics via microtubule targeting requires kinesin-1. J Cell Biol 156:349–359PubMedCrossRef Krylyshkina O, Kaverina I, Kranewitter W, Steffen W, Alonso MC, Cross RA, Small JV (2002) Modulation of substrate adhesion dynamics via microtubule targeting requires kinesin-1. J Cell Biol 156:349–359PubMedCrossRef
34.
go back to reference Tsuda Y, Mashimo T, Yoshiya I, Kaseda K, Harada Y, Yanagida T (1996) Direct inhibition of the actomyosin motility by local anesthetics in vitro. Biophys J 71:2733–2741PubMedCrossRef Tsuda Y, Mashimo T, Yoshiya I, Kaseda K, Harada Y, Yanagida T (1996) Direct inhibition of the actomyosin motility by local anesthetics in vitro. Biophys J 71:2733–2741PubMedCrossRef
35.
go back to reference Crevel IM, Lockhart A, Cross RA (1996) Weak and strong states of kinesin and ncd. J Mol Biol 257:66–76PubMedCrossRef Crevel IM, Lockhart A, Cross RA (1996) Weak and strong states of kinesin and ncd. J Mol Biol 257:66–76PubMedCrossRef
36.
go back to reference Hirose K, Lockhart A, Cross RA, Amos LA (1995) Nucleotide-dependent angular change in kinesin motor domain bound to tubulin. Nature 376:277–279PubMedCrossRef Hirose K, Lockhart A, Cross RA, Amos LA (1995) Nucleotide-dependent angular change in kinesin motor domain bound to tubulin. Nature 376:277–279PubMedCrossRef
37.
go back to reference Morris VL, MacDonald IC, Koop S, Schmidt EE, Chambers AF, Groom AC (1993) Early interactions of cancer cells with the microvasculature in mouse liver and muscle during hematogenous metastasis: videomicroscopic analysis. Clin Exp Metastasis 11:377–390PubMedCrossRef Morris VL, MacDonald IC, Koop S, Schmidt EE, Chambers AF, Groom AC (1993) Early interactions of cancer cells with the microvasculature in mouse liver and muscle during hematogenous metastasis: videomicroscopic analysis. Clin Exp Metastasis 11:377–390PubMedCrossRef
38.
go back to reference Tsuji K, Yamauchi K, Yang M, Jiang P, Bouvet M, Endo H, Kanai Y, Yamashita K, Moossa AR, Hoffman RM (2006) Dual-color imaging of nuclear-cytoplasmic dynamics, viability, and proliferation of cancer cells in the portal vein area. Cancer Res 66:303–306PubMedCrossRef Tsuji K, Yamauchi K, Yang M, Jiang P, Bouvet M, Endo H, Kanai Y, Yamashita K, Moossa AR, Hoffman RM (2006) Dual-color imaging of nuclear-cytoplasmic dynamics, viability, and proliferation of cancer cells in the portal vein area. Cancer Res 66:303–306PubMedCrossRef
Metadata
Title
Local anesthetics inhibit kinesin motility and microtentacle protrusions in human epithelial and breast tumor cells
Authors
Jennifer R. Yoon
Rebecca A. Whipple
Eric M. Balzer
Edward H. Cho
Michael A. Matrone
Michelle Peckham
Stuart S. Martin
Publication date
01-10-2011
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 3/2011
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-010-1239-7

Other articles of this Issue 3/2011

Breast Cancer Research and Treatment 3/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine