A high throughput screen identifies Nefopam as targeting cell proliferation in β-catenin driven neoplastic and reactive fibroproliferative disorders

Raymond Poon, Helen Hong, Xin Wei, James Pan, Benjamin A Alman, Raymond Poon, Helen Hong, Xin Wei, James Pan, Benjamin A Alman

Abstract

Fibroproliferative disorders include neoplastic and reactive processes (e.g. desmoid tumor and hypertrophic scars). They are characterized by activation of β-catenin signaling, and effective pharmacologic approaches are lacking. Here we undertook a high throughput screen using human desmoid tumor cell cultures to identify agents that would inhibit cell viability in tumor cells but not normal fibroblasts. Agents were then tested in additional cell cultures for an effect on cell proliferation, apoptosis, and β-catenin protein level. Ultimately they were tested in Apc1638N mice, which develop desmoid tumors, as well as in wild type mice subjected to full thickness skin wounds. The screen identified Neofopam, as an agent that inhibited cell numbers to 42% of baseline in cell cultures from β-catenin driven fibroproliferative disorders. Nefopam decreased cell proliferation and β-catenin protein level to 50% of baseline in these same cell cultures. The half maximal effective concentration in-vitro was 0.5 uM and there was a plateau in the effect after 48 hours of treatment. Nefopam caused a 45% decline in tumor number, 33% decline in tumor volume, and a 40% decline in scar size when tested in mice. There was also a 50% decline in β-catenin level in-vivo. Nefopam targets β-catenin protein level in mesenchymal cells in-vitro and in-vivo, and may be an effective therapy for neoplastic and reactive processes driven by β-catenin mediated signaling.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Nefopam inhibits cell viability in…
Figure 1. Nefopam inhibits cell viability in cultures from aggressive fibromatosis and hypertrophic cutaneous wounds.
Data from the Sulforhodamine B assay. The mean value for the cell culture treated with carrier was arbitrarily defined as 100 for each cell type. A) Data for a dose of 10 µM of Nefopam. There was a significantly lower level of cell viability in the aggressive fibromatosis or the hypertrophic scar cultures compared to that seen in normal tissues. B) Data from different doses of Nefopam in cell cultures from aggressive fibromatosis or from hypertrophic scars, showing a dose dependent decrease in cell viability. C) Time course data for cell viability after days of treatment with 10 µM of Nefopam in cell cultures from aggressive fibromatosis or from hypertrophic scars. Data is given as means and 95% confidence intervals. Control data is generated by treatment with diphenhydramine. Treatment with carrier alone gave identical results as for diphenhydramine. An asterisk over a data point indicates a significant difference form the control cell cultures.
Figure 2. Nefopam inhibits cell proliferation in…
Figure 2. Nefopam inhibits cell proliferation in fibroblast cell cultures from hypertrophic cutaneous wounds and from aggressive fibromatosis tumors.
Percent BrdU incorporation or annexin V staining from the various cell cultures treated with 10 µM of Nefopam. Data is given as means and 95% confidence intervals. An asterisk over a data point indicates a significant difference form the control cell cultures. There was a significant difference in BrdU incorporation in the tumor and hyperplastic scar cultures treated with Nefopam compared to treatment with carrier alone. Treatment with diphenhydramine resulted in the identical findings as for treatment with the carrier.
Figure 3. Nefopam modulates β-catenin protein level.
Figure 3. Nefopam modulates β-catenin protein level.
A) Western blot analysis for β-catenin in aggressive fibromatosis and hypertrophic scar cell cultures treated with Nefopam. A representative Western blot for total β-catenin is shown at the top of the panel. The data is shown in graphical form in the lower part of the panel, as mean and 95% confidence intervals for the relative density compared to the loading control for six pairs of cultures for each condition. An asterisk over a data point indicates a significant difference form the control cell cultures. There is a decrease in β-catenin level with Nefopam treatment in cell cultures from both conditions. B) A representative Western blot for β-catenin and serine-9-phosph-GSK-3-β in normal fibroblasts treated with Wnt3a is shown at the top of the panel. Composite data is shown in graphical form in the lower part of the panel. Actin and total GSK3-β are used as loading controls. There is an increase in serine-9-phosph-GSK-3-β and β-catenin with Wnt3a treatment, which is significantly lowered, close to the baseline level, with Nefopam treatment. C) A representative Western blot for β-catenin in fibroblasts expressing a stabilized form of β-catenin lacking exon three is shown on the top of the panel. Composite data is shown in graphical form on the bottom of the panel. There is a decrease in β-catenin with Nefopam treatment.
Figure 4. Nefopam suppresses the neoplastic phenotype…
Figure 4. Nefopam suppresses the neoplastic phenotype in murine aggressive fibromatosis tumors.
The number of tumors found in male Apc1638N mice. Data is given as the mean and 95% confidence intervals for the number of tumors formed in male mice in no treatment; 0.1% DMSO (carrier); or Nefopam treatment groups. Data is given as means and 95% confidence intervals. An asterisk over a data point indicates a significant difference form the control cell cultures. Mice treated with the carrier showed a comparable number of tumors to mice that received no treatment, while mice treated with Nefopam developed significantly fewer tumors.
Figure 5. Nefopam regulates scar size and…
Figure 5. Nefopam regulates scar size and β-catenin level in cutaneous wound repair.
A to D) Representative histologic sections through the widest margin of scars 14 days after wounding. A) A wound from a mouse treated with TGF-β. B) A wound in a mice treated with carrier only. C) A wound from a mouse treated with TGF-β and Nefopam. D) A wound from a mouse treated with Nefopam. Arrows show the widest width of the scar. The black line is 500 µm in length. E) A graphical representation of the mean and 95% confidence intervals of the widest diameter of the scars in µms. F) A representative Western blot mice showing the β-catenin protein levels in wounds from mice treated with TGF-β, Nefopam, or both, showing that Nefopam decreases β-catenin protein level in the wounds.

References

    1. Alman BA, Li C, Pajerski ME, Diaz-Cano S, Wolfe HJ. Increased beta-catenin protein and somatic APC mutations in sporadic aggressive fibromatoses (desmoid tumors). Am J Pathol. 1997;151:329–334.
    1. Cheon SS, Wei Q, Gurung A, Youn A, Bright T, et al. Beta-catenin regulates wound size and mediates the effect of TGF-beta in cutaneous healing. FASEB J. 2006;20:692–701.
    1. Chen Y, Whetstone HC, Lin AC, Nadesan P, Wei Q, et al. Beta-Catenin Signaling Plays a Disparate Role in Different Phases of Fracture Repair: Implications for Therapy to Improve Bone Healing. PLoS Medicine. 2007;4:e249.
    1. Lam AP, Gottardi CJ. beta-catenin signaling: a novel mediator of fibrosis and potential therapeutic target. Curr Opin Rheumatol. 2011;23:562–567.
    1. Clark RA. Biology of dermal wound repair. Dermatol Clin. 1993;11:647–666.
    1. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–746.
    1. Chen Y, Whetstone HC, Youn A, Nadesan P, Chow EC, et al. Beta-catenin signaling pathway is crucial for bone morphogenetic protein 2 to induce new bone formation. J Biol Chem. 2007;282:526–533.
    1. Cheon S, Poon R, Yu C, Khoury M, Shenker R, et al. Prolonged beta-catenin stabilization and tcf-dependent transcriptional activation in hyperplastic cutaneous wounds. Lab Invest. 2005;85:416–425.
    1. Cheon SS, Cheah AY, Turley S, Nadesan P, Poon R, et al. beta-Catenin stabilization dysregulates mesenchymal cell proliferation, motility, and invasiveness and causes aggressive fibromatosis and hyperplastic cutaneous wounds. Proc Natl Acad Sci U S A. 2002;99:6973–6978.
    1. Cheon SS, Nadesan P, Poon R, Alman BA. Growth factors regulate beta-catenin-mediated TCF-dependent transcriptional activation in fibroblasts during the proliferative phase of wound healing. Exp Cell Res. 2004;293:267–274.
    1. Bock O, Schmid-Ott G, Malewski P, Mrowietz U. Quality of life of patients with keloid and hypertrophic scarring. Arch Dermatol Res. 2006;297:433–438.
    1. Clark RA. U.S. markets for wound management products. In: Clark RA, editor. The molecular and cellular biology of wound repair. new york: plenium press; 1996. pp. 3–20.
    1. Alman BA, Goldberg MJ, Naber SP, Galanopoulous T, Antoniades HN, et al. Aggressive fibromatosis. J Pediatr Orthop. 1992;12:1–10.
    1. Couture J, Mitri A, Lagace R, Smits R, Berk T, et al. A germline mutation at the extreme 3′ end of the APC gene results in a severe desmoid phenotype and is associated with overexpression of beta-catenin in the desmoid tumor. Clin Genet. 2000;57:205–212.
    1. Tejpar S, Nollet F, Li C, Wunder JS, Michils G, et al. Predominance of beta-catenin mutations and beta-catenin dysregulation in sporadic aggressive fibromatosis (desmoid tumor). Oncogene. 1999;18:6615–6620.
    1. Yang JC, Chang AE, Baker AR, Sindelar WF, Danforth DN, et al. Randomized prospective study of the benefit of adjuvant radiation therapy in the treatment of soft tissue sarcomas of the extremity. J Clin Oncol. 1998;16:197–203.
    1. Tejpar S, Li C, Yu C, Poon R, Denys H, et al. Tcf-3 expression and beta-catenin mediated transcriptional activation in aggressive fibromatosis (desmoid tumour). Br J Cancer. 2001;85:98–101.
    1. Bowley E, O'Gorman DB, Gan BS. Beta-catenin signaling in fibroproliferative disease. J Surg Res. 2007;138:141–150.
    1. Weisman JL, Liou AP, Shelat AA, Cohen FE, Guy RK, et al. Searching for new antimalarial therapeutics amongst known drugs. Chem Biol Drug Des. 2006;67:409–416.
    1. Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A. 2007;104:4048–4053.
    1. Heel RC, Brogden RN, Pakes GE, Speight TM, Avery GS. Nefopam: a review of its pharmacological properties and therapeutic efficacy. Drugs. 1980;19:249–267.
    1. Tischler AS, Lee YC, Costopoulos D, Nunnemacher G, DeLellis RA, et al. Establishment of a continuous somatostatin-producing line of medullary thyroid carcinoma cells from BALB/c mice. J Endocrinol. 1986;110:309–313.
    1. Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J Immunol Methods. 1995;184:39–51.
    1. Tobin WE, Gold RH. Nefopam hydrochloride: a novel muscle relaxant. J Clin Pharmacol New Drugs. 1972;12:230–238.
    1. Smits R, van der Houven, van Oordt W, Luz A, Zurcher C, Jagmohan-Changur S, et al. Apc1638N: a mouse model for familial adenomatous polyposis-associated desmoid tumors and cutaneous cysts. Gastroenterology. 1998;114:275–283.
    1. Wang RI, Waite EM. The clinical analgesic efficacy of oral nefopam hydrochloride. J Clin Pharmacol. 1979;19:395–402.
    1. Pillans PI, Woods DJ. Adverse reactions associated with nefopam. N Z Med J. 1995;108:382–384.
    1. Esposito E, Romandini S, Merlo-Pich E, Mennini T, Samanin R. Evidence of the involvement of dopamine in the analgesic effect of nefopam. Eur J Pharmacol. 1986;128:157–164.
    1. Marazziti D, Rotondo A, Ambrogi F, Cassano GB. Analgesia by nefopam: does it act through serotonin? Drugs Exp Clin Res. 1991;17:259–261.
    1. Girard P, Pansart Y, Coppe MC, Verniers D, Gillardin JM. Role of the histamine system in nefopam-induced antinociception in mice. Eur J Pharmacol. 2004;503:63–69.
    1. Verleye M, Andre N, Heulard I, Gillardin JM. Nefopam blocks voltage-sensitive sodium channels and modulates glutamatergic transmission in rodents. Brain Res. 2004;1013:249–255.
    1. Gainetdinov RR, Premont RT, Bohn LM, Lefkowitz RJ, Caron MG. Desensitization of G protein-coupled receptors and neuronal functions. Annu Rev Neurosci. 2004;27:107–144.
    1. Beaulieu JM, Gainetdinov RR, Caron MG. The Akt-GSK-3 signaling cascade in the actions of dopamine. Trends Pharmacol Sci. 2007;28:166–172.
    1. Juarez-Rojop IE, Granados-Soto V, Diaz-Zagoya JC, Flores-Murrieta FJ, Torres-Lopez JE. Involvement of cholecystokinin in peripheral nociceptive sensitization during diabetes in rats as revealed by the formalin response. Pain. 2006;122:118–125.
    1. Vuga LJ, Ben-Yehudah A, Kovkarova-Naumovski E, Oriss T, Gibson KF, et al. WNT5A is a regulator of fibroblast proliferation and resistance to apoptosis. Am J Respir Cell Mol Biol. 2009;41:583–589.
    1. Wunder JS, Nielsen TO, Maki RG, O'Sullivan B, Alman BA. Opportunities for improving the therapeutic ratio for patients with sarcoma. Lancet Oncol. 2007;8:513–524.
    1. Yao H, Ashihara E, Maekawa T. Targeting the Wnt/beta-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 2011;15:873–887.
    1. Sato M. Upregulation of the Wnt/beta-catenin pathway induced by transforming growth factor-beta in hypertrophic scars and keloids. Acta Derm Venereol. 2006;86:300–307.
    1. Hwang I, Seo EY, Ha H. Wnt/beta-catenin signaling: a novel target for therapeutic intervention of fibrotic kidney disease. Arch Pharm Res. 2009;32:1653–1662.
    1. Wei J, Melichian D, Komura K, Hinchcliff M, Lam AP, et al. Canonical Wnt signaling induces skin fibrosis and subcutaneous lipoatrophy: a novel mouse model for scleroderma? Arthritis Rheum. 2011;63:1707–1717.
    1. Varallo VM, Gan BS, Seney S, Ross DC, Roth JH, et al. Beta-catenin expression in Dupuytren's disease: potential role for cell-matrix interactions in modulating beta-catenin levels in vivo and in vitro. Oncogene. 2003;22:3680–3684.
    1. Kim TH, Kim SH, Seo JY, Chung H, Kwak HJ, et al. Blockade of the Wnt/beta-catenin pathway attenuates bleomycin-induced pulmonary fibrosis. Tohoku J Exp Med. 2011;223:45–54.
    1. Henderson WR, Chi EY, Ye X, Nguyen C, Tien YT, et al. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci U S A. 2010;107:14309–14314.
    1. Li C, Bapat B, Alman BA. Adenomatous polyposis coli gene mutation alters proliferation through its beta-catenin-regulatory function in aggressive fibromatosis (desmoid tumor). Am J Pathol. 1998;153:709–714.
    1. Henriksson E, Kjellen E, Wahlberg P, Wennerberg J, Kjellstrom JH. Differences in estimates of cisplatin-induced cell kill in vitro between colorimetric and cell count/colony assays. In Vitro Cell Dev Biol Anim. 2006;42:320–323.
    1. Yokoyama N, Yin D, Malbon CC. Abundance, complexation, and trafficking of Wnt/beta-catenin signaling elements in response to Wnt3a. J Mol Signal. 2007;2:11.
    1. Nakamura T, Sano M, Songyang Z, Schneider MD. A Wnt- and beta-catenin-dependent pathway for mammalian cardiac myogenesis. Proc Natl Acad Sci U S A. 2003;100:5834–5839.
    1. Silkstone D, Hong H, Alman BA. Beta-catenin in the race to fracture repair: in it to Wnt. Nat Clin Pract Rheumatol. 2008;4:413–419.
    1. Case MT, Smith JK, Nelson RA. Reproductive, acute and subacute toxicity studies with nefopam in laboratory animals. Toxicol Appl Pharmacol. 1975;33:46–51.
    1. Poon R, Smits R, Li C, Jagmohan-Changur S, Kong M, et al. Cyclooxygenase-two (COX-2) modulates proliferation in aggressive fibromatosis (desmoid tumor). Oncogene. 2001;20:451–460.
    1. Aymard G, Warot D, Demolis P, Laville I, Diquet B. Sensitive determination of nefopam and its metabolite desmethyl-nefopam in human biological fluids by HPLC. J Pharm Biomed Anal. 2002;30:1013–1021.

Source: PubMed

Подписаться