Skip to main content

Advertisement

Log in

Role of Dopamine and D2 Dopamine Receptor in the Pathogenesis of Inflammatory Bowel Disease

  • Original Article
  • Published:
Digestive Diseases and Sciences Aims and scope Submit manuscript

Abstract

Background

VEGF-induced vascular permeability and blood vessels remodeling are key features of inflammatory bowel disease (IBD) pathogenesis. Dopamine through D2 receptor (D2R) inhibits VEGF/VPF-mediated vascular permeability and angiogenesis in tumor models. In this study, we tested the hypothesis that pathogenesis of IBD is characterized by the disturbance of dopaminergic system and D2R activity.

Methods

IL-10 knockout (KO) mice and rats with iodoacetamide-induced ulcerative colitis (UC) were treated intragastrically with D2R agonists quinpirole (1 mg/100 g) or cabergoline (1 or 5 µg/100 g). Macroscopic, histologic, and clinical features of IBD, colonic vascular permeability, and angiogenesis were examined.

Results

Although colonic D2R protein increased, levels of tyrosine hydroxylase and dopamine transporter DAT decreased in both models of IBD. Treatment with quinpirole decreased the size of colonic lesions in rats with iodoacetamide-induced UC (p < 0.01) and reduced colon wet weight in IL-10 KO mice (p < 0.05). Quinpirole decreased colonic vascular permeability (p < 0.001) via downregulation of c-Src and Akt phosphorylation. Cabergoline (5 µg/100 g) reduced vascular permeability but did not affect angiogenesis and improved signs of iodoacetamide-induced UC in rats (p < 0.05).

Conclusions

Treatment with D2R agonists decreased the severity of UC in two animal models, in part, by attenuation of enhanced vascular permeability and prevention of excessive vascular leakage. Hence, the impairment dopaminergic system seems to be a feature of IBD pathogenesis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Burisch J, Jess T, Martinato M, et al. The burden of inflammatory bowel disease in Europe. J Crohns Colitis. 2013;7:322–337.

    Article  PubMed  Google Scholar 

  2. Rocchi A, Benchimol EI, Bernstein CN, et al. Inflammatory bowel disease: a Canadian burden of illness review. Can J Gastroenterol. 2012;26:811–817.

    PubMed  PubMed Central  Google Scholar 

  3. Ording AG, Horváth-Puhó E, Erichsen R, et al. Five-year mortality in colorectal cancer patients with ulcerative colitis or Crohn’s disease: a nationwide population-based cohort study. Inflamm Bowel Dis. 2013;19:800–805.

    Article  PubMed  Google Scholar 

  4. Brahme F, Lindstrom C. A comparative radiographic and pathological study of intestinal vaso-architecture in Crohn’s disease and in ulcerative colitis. Gut. 1970;11:928–940.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chidlow JH Jr, Langston W, Greer JJM, et al. Differential angiogenic regulation of experimental colitis. Am J Pathol. 2006;169:2014–2030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Danese S, Sans M, Motte CDL, et al. Angiogenesis as a novel component of inflammatory bowel disease pathogenesis. Gastroeneterology. 2006;130:2060–2073.

    Article  CAS  Google Scholar 

  7. Foitzik T, Kruschewski M, Kroesen A, et al. Does microcirculation play a role in the pathogenesis of inflammatory bowel diseases? Answers from intravital microscopic studies in animal models. Int J Colorectal Dis. 1999;14:29–34.

    Article  CAS  PubMed  Google Scholar 

  8. Hatoum OA, Miura H, Binion DG. The vascular contribution in the pathogenesis of inflammatory bowel disease. Am J Physiol Heart Circ Physiol. 2003;285:1791–1796.

    Article  Google Scholar 

  9. Spalinger J, Patriquin H, Miron MC, et al. Doppler US in patients with Crohn disease: vessel density in the diseased bowel reflects disease activity. Radiology. 2000;217:787–791.

    Article  CAS  PubMed  Google Scholar 

  10. Schinzari F, Armuzzi A, De Pascalis B, et al. Tumor necrosis factor-alpha antagonism improves endothelial dysfunction in patients with Crohn’s disease. Clin Pharmacol Ther. 2008;83:70–76.

    Article  CAS  PubMed  Google Scholar 

  11. Tolstanova G, Khomenko T, Deng X, et al. Neutralizing anti-vascular endothelial growth factor (VEGF) antibody reduces severity of experimental ulcerative colitis in rats: direct evidence for the pathogenic role of VEGF. J Pharmacol Exp Ther. 2009;328:749–757.

    Article  CAS  PubMed  Google Scholar 

  12. Chidlow JH Jr, Glawe JD, Pattilllo CB, et al. VEGF164 isoform specific regulation of T-cell-dependent experimental colitis in mice. Inflamm Bowel Dis. 2011;17:1501–1512.

    Article  PubMed  Google Scholar 

  13. Jerkic M, Peter M, Ardelean D, et al. Dextran sulfate sodium leads to chronic colitis and pathological angiogenesis in Endoglin heterozygous mice. Inflamm Bowel Dis. 2010;16:1859–1870.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Scaldaferri F, Vetrano S, Sans M, et al. VEGF-A links angiogenesis and inflammation in inflammatory bowel disease pathogenesis. Gastroenterology. 2009;136:585–595.

    Article  CAS  PubMed  Google Scholar 

  15. Plevy S, Silverberg MS, Lockton S, et al. Combined Serological, Genetic, and Inflammatory Markers Differentiate Non-IBD, Crohn’s Disease, and Ulcerative Colitis Patients. Inflamm Bowel Dis. 2013;19:1139–1148.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Coriat R, Mir O, Leblanc S, et al. Feasibility of anti-VEGF agent bevacizumab in patients with Crohn’s disease. Inflamm Bowel Dis. 2011;17:1632.

    Article  PubMed  Google Scholar 

  17. Shaughnessy AF. Monoclonal antibodies: magic bullets with a hefty price tag. BMJ. 2012;345:8346.

    Article  Google Scholar 

  18. Loriot Y, Boudou-Rouquette P, Billemont B, et al. Acute exacerbation of hemorrhagic rectocolitis during antiangiogenic therapy with sunitinib and sorafenib. Ann Oncol. 2008;19:1975.

    Article  CAS  PubMed  Google Scholar 

  19. Basu S, Nagy JA, Pal S, et al. The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat Med. 2001;7:569–574.

    Article  CAS  PubMed  Google Scholar 

  20. Sarkar C, Chakroborty D, Chowdhury UR, et al. Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin Cancer Res. 2008;14:2502–2510.

    Article  CAS  PubMed  Google Scholar 

  21. Basu S, Sarkar C, Chakroborty D, et al. Ablation of peripheral dopaminergic nerves stimulates malignant tumor growth by inducing vascular permeability factor/vascular endothelial growth factor-mediated angiogenesis. Cancer Res. 2004;64:5551–5555.

    Article  CAS  PubMed  Google Scholar 

  22. Bhattacharya R, Sinha S, Yang SP, et al. The neurotransmitter dopamine modulates vascular permeability in the endothelium. J Mol Signal. 2008;3:14.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Chakroborty D, Sarkar C, Yu H, et al. Dopamine stabilizes tumor blood vessels by up-regulating angiopoietin 1 expression in pericytes and Kruppel-like factor-2 expression in tumor endothelial cells. Proc Natl Acad Sci USA. 2011;108:20730–20735.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Magro F, Vieira-Coelho MA, Fraga S, et al. Impaired synthesis or cellular storage of norepinephrine, dopamine, and 5-hydroxytryptamine in human inflammatory bowel disease. Dig Dis Sci. 2002;47:216–224.

    Article  CAS  PubMed  Google Scholar 

  25. Magro F, Fraga S, Ribeiro T, et al. Decreased availability of intestinal dopamine in transmural colitis may relate to inhibitory effects of interferon-gamma upon l-DOPA uptake. Acta Physiol Scand. 2004;180:379–386.

    Article  CAS  PubMed  Google Scholar 

  26. Magro F, Cunha E, Araujo F, et al. Dopamine D2 receptor polymorphisms in inflammatory bowel disease and the refractory response to treatment. Dig Dis Sci. 2006;51:2039–2044.

    Article  CAS  PubMed  Google Scholar 

  27. Satoh H, Sato F, Takami K, et al. New ulcerative colitis model induced by SH blockers in rats and the effects of antiinflammatory drugs on the colitis. Jpn J Pharmacol. 1997;73:299–309.

    Article  CAS  PubMed  Google Scholar 

  28. Castaneda FE, Walia B, Vijay-Kumar M, et al. Targeted deletion of metalloproteinase 9 attenuates experimental colitis in mice: central role of epithelial-derived MMP. Gastroenterology. 2005;129:1991–2008.

    Article  CAS  PubMed  Google Scholar 

  29. Patterson CE, Rhoades RA, Garcia JG. Evans blue dye as a marker of albumin clearance in cultured endothelial monolayer and isolated lung. J Appl Physiol. 1992;72:865–873.

    Article  CAS  PubMed  Google Scholar 

  30. Tolstanova G, Deng X, French SW, et al. Early endothelial damage and increased colonic vascular permeability in the development of experimental ulcerative colitis in rats and mice. Lab Invest. 2012;92:9–21.

    Article  CAS  PubMed  Google Scholar 

  31. Khomenko T, Szabo S, Deng X, et al. Suppression of early growth response factor-1 with egr-1 antisense oligodeoxynucleotide aggravates experimental duodenal ulcers. Am J Physiol Gastrointest Liver Physiol. 2006;290:G1211–G1218.

    Article  CAS  PubMed  Google Scholar 

  32. Webster J. A comparative review of the tolerability profiles of dopamine agonists in the treatment of hyperprolactinaemia and inhibition of lactation. Drug Saf. 1996;14:228–238.

    Article  CAS  PubMed  Google Scholar 

  33. Benedetti MS, Dostert P, Barone D, et al. In vivo interaction of cabergoline with rat brain dopamine receptors labelled with [3H]N-n-propylnorapomorphine. Eur J Pharmacol. 1990;187:399–408.

    Article  CAS  PubMed  Google Scholar 

  34. Lahlou S. Involvement of spinal dopamine receptors in mediation of the hypotensive and bradycardic effects of systemic quinpirole in anaesthetised rats. Eur J Pharmacol. 1998;353:227–237.

    Article  CAS  PubMed  Google Scholar 

  35. Gomez R, Gonzalez-Izquierdo M, Zimmermann RC, et al. Low-dose dopamine agonist administration blocks vascular endothelial growth factor (VEGF)-mediated vascular hyperpermeability without altering VEGF receptor 2-dependent luteal angiogenesis in a rat ovarian hyperstimulation model. Endocrinology. 2006;147:5400–5411.

    Article  CAS  PubMed  Google Scholar 

  36. Berg DJ, Davidson N, Kuhn R, et al. Enterocolitis and colon cancer in interleukin-10-deficient mice are associated with aberrant cytokine production and CD4+ TH1-like responses. J Clin Invest. 1996;98:1010–1020.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gavard J, Gutkind JS. VEGF controls endothelial-cell permeability by promoting the β-arrestin-dependent endocytosis of VE-cadherin. Nat Cell Biol. 2006;8:1223–1234.

    Article  CAS  PubMed  Google Scholar 

  38. Kilic E, Kilic U, Wang Y, et al. The phosphatidylinositol-3 kinase/Akt pathway mediates VEGF’s neuroprotective activity and induces blood brain barrier permeability after focal cerebral ischemia. FASEB J. 2006;20:1185–1187.

    Article  CAS  PubMed  Google Scholar 

  39. Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63:182–217.

    Article  CAS  PubMed  Google Scholar 

  40. Eaker EY, Bixler GB, Dunn AJ, et al. Dopamine and norepinephrine in the gastrointestinal tract of mice and the effects of neurotoxins. J Pharmacol Exp Ther. 1988;244:438–442.

    CAS  PubMed  Google Scholar 

  41. Eisenhofer G, Aneman A, Friberg P, et al. Substantial production of dopamine in the human gastrointestinal tract. J Clin Endocrinol Metab. 1997;82:3864–3871.

    Article  CAS  PubMed  Google Scholar 

  42. Eldrup E, Richter EA, Christensen NJ. DOPA, norepinephrine, and dopamine in rat tissues: no effect of sympathectomy on muscle DOPA. Am J Physiol. 1989;256:284–287.

    Google Scholar 

  43. Mezey E, Eisenhofer G, Hansson S, et al. Non-neuronal dopamine in the gastrointestinal system. Clin Exp Pharmacol Physiol Suppl. 1999;26:14–22.

    Google Scholar 

  44. Kim HJ, Koh PO, Kang SS, et al. The localization of dopamine D2 receptor mRNA in the human placenta and the anti-angiogenic effect of apomorphine in the chorioallantoic membrane. Life Sci. 2001;68:1031–1040.

    Article  CAS  PubMed  Google Scholar 

  45. Sinha S, Vohra PK, Bhattacharya R, et al. Dopamine regulates phosphorylation of VEGF receptor 2 by engaging Src-homology-2-domain-containing protein tyrosine phosphatase 2. J Cell Sci. 2009;122:3385–3392.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li ZS, Schmauss C, Cuenca A, et al. Physiological modulation of intestinal motility by enteric dopaminergic neurons and the D2 receptor: analysis of dopamine receptor expression, location, development, and function in wild-type and knock-out mice. J Neurosci. 2006;26:2798–2807.

    Article  CAS  PubMed  Google Scholar 

  47. Szabo S, Sandrock AW, Nafradi J, et al. Dopamine and dopamine receptors in the gut: their possible role in duodenal ulceration. In: Kawasaki M, et al., eds. Advances in dopamine research. Oxford: Pergamon Press; 1982:165–170.

    Chapter  Google Scholar 

  48. Tian YM, Chen X, Luo DZ, et al. Alteration of dopaminergic markers in gastrointestinal tract of different rodent models of Parkinson’s disease. Neuroscience. 2008;153:634–644.

    Article  CAS  PubMed  Google Scholar 

  49. Eldrup E, Richter EA. DOPA, dopamine, and DOPAC concentrations in the rat gastrointestinal tract decrease during fasting. Am J Physiol Endocrinol Metab. 2000;279:815–822.

    Google Scholar 

  50. Asano Y, Hiramoto T, Nishino R, et al. Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol. 2012;303:1288–1295.

    Article  Google Scholar 

  51. Bryan-Lluka LJ, O’Donnell SR. Dopamine and adrenaline, but not isoprenaline, are substrates for uptake and metabolism in isolated perfused lungs of rats. Naunyn Schmiedebergs Arch Pharmacol. 1992;346:20–26.

    Article  CAS  PubMed  Google Scholar 

  52. Tarnawski A, Coron E, Mosnier JF, et al. In-vivo detection by confocal endomicroscopy of two distinct structural abnormalities in angioarchitecture and increased VP in colonic mucosa of patients with IBD in remission: mechanistic implications. Gastroenterology. 2009;136:112.

    Google Scholar 

  53. Colucci M, Cervio M, Faniglione M, et al. Intestinal dysmotility and enteric neurochemical changes in a Parkinson’s disease rat model. Auton Neurosci. 2012;169:77–86.

    Article  CAS  PubMed  Google Scholar 

  54. Tolstanova G, Khomenko T, Deng X, et al. New molecular mechanisms of the unexpectedly complex role of VEGF in ulcerative colitis. Biochem Biophys Res Commun. 2010;399:613–616.

    Article  CAS  PubMed  Google Scholar 

  55. Alvarez C, Martí-Bonmatí L, Novella-Maestre E, et al. Dopamine agonist cabergoline reduces hemoconcentration and ascites in hyperstimulated women undergoing assisted reproduction. J Clin Endocrinol Metab. 2007;92:2931–2937.

    Article  CAS  PubMed  Google Scholar 

  56. Szabo S, Horner HC, Maull H, et al. Biochemical changes in tissue catecholamines and serotonin in duodenal ulceration caused by cysteamine or propionitrile in the rat. J Pharmacol Exp Ther. 1987;240:871–878.

    CAS  PubMed  Google Scholar 

  57. Horner HC, Szabo S. Differential effect of changing central and peripheral catecholamine levels in cysteamine-induced duodenal ulcer in the rat. Life Sci. 1981;29:2437–2443.

    Article  CAS  PubMed  Google Scholar 

  58. Devos D, Lebouvier T, Lardeux B, et al. Colonic inflammation in Parkinson’s disease. Neurobiol Dis. 2013;50:42–48.

    Article  CAS  PubMed  Google Scholar 

  59. Forsyth Christopher B, Kathleen M, et al. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS One. 2011;6:28032.

    Article  Google Scholar 

  60. Salat-Foix D, Tran K, Ranawaya R, et al. Increased intestinal permeability and Parkinson disease patients: chicken or egg? Can J Neurol Sci. 2012;39:185–188.

    Article  CAS  PubMed  Google Scholar 

  61. Ray A, Henke PG, Sullivan RM. Effects of intra-amygdalar dopamine agonists and antagonists on gastric stress lesions in rats. Neurosci Lett. 1988;84:302–306.

    Article  CAS  PubMed  Google Scholar 

  62. Ray A, Henke PG. The basolateral amygdala, dopamine and gastric stress ulcer formation in rats. Brain Res. 1991;558:335–338.

    Article  CAS  PubMed  Google Scholar 

  63. McKenna F, McLaughlin PJ, Lewis BJ, et al. Dopamine receptor expression on human T- and B-lymphocytes, monocytes, neutrophils, eosinophils and NK cells: a flow cytometric study. J Neuroimmunol. 2002;132:34–40.

    Article  CAS  PubMed  Google Scholar 

  64. Besser MJ, Ganor Y, Levite M. Dopamine by itself activates either D2, D3 or D1/D5 dopaminergic receptors in normal human T-cells and triggers the selective secretion of either IL-10, TNFalpha or both. J Neuroimmunol. 2005;169:161–171.

    Article  CAS  PubMed  Google Scholar 

  65. Check JH, Katsoff B, Cohen R. Novel highly effective medical treatment of severe treatment refractory Crohn’s disease using sympathomimetic amines: case report. Inflamm Bowel Dis. 2010;16:1999–2000.

    Article  PubMed  Google Scholar 

  66. Check JH, Katsoff B, Cohen R. Case report showing that a woman with ulcerative colitis refractory to standard therapy responded well to the sympathomimetic amine dextroamphetamine sulfate. Inflamm Bowel Dis. 2011;17:870–871.

    Article  PubMed  Google Scholar 

  67. Check JH, Amadi C, Kaplan H, Katsoff D. The treatment of idiopathic edema, a cause of chronic pelvic pain in women: effectively controlled chronic refractory urticaria–case reports. Clin Exp Obstet Gynecol. 2006;33:183–184.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The present study was supported by a Department of Veterans Affairs, Veterans Health Administration Merit Review Grant VAMR0710-580 and VAMR0810-877 to Zs. Sandor and S. Szabo and by U.S. Civilian Research & Development Foundation (CRDF) CREST II Junior Scientist Research Collaboration Program 09DP036-05 and the Ministry of Education and Science of Ukraine Grant 15BF036-01 to G. Tolstanova

Conflict of interest

None.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sandor Szabo.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tolstanova, G., Deng, X., Ahluwalia, A. et al. Role of Dopamine and D2 Dopamine Receptor in the Pathogenesis of Inflammatory Bowel Disease. Dig Dis Sci 60, 2963–2975 (2015). https://doi.org/10.1007/s10620-015-3698-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10620-015-3698-5

Keywords

Navigation