Skip to main content

Psychosoziale Aspekte der Nahrungsaufnahme, Verdauung und Ausscheidung

  • Chapter
  • First Online:
Funktionelle Magen-Darm-Störungen im Kindes- und Jugendalter
  • 526 Accesses

Zusammenfassung

Der Gastrointestinaltrakt (GIT) ist in seinen Grundfunktionen autonom. Der GIT ist aber sensorisch und motorisch mit dem zentralen Nervensystem (ZNS) verbunden, sodass die gastrointestinalen Grundfunktionen in den Verhaltenskontext eingebunden werden. Besondere Bedeutung haben hier die Warnsysteme, die der Abwehr von Körperschädigungen dienen. Dazu gehören sowohl Spinalnerven, die nozizeptive Reflexe und Schmerzempfindungen auslösen, als auch Vagusafferenzen, die eine Krankheitsreaktion induzieren.

Aus Studien wissen wir, dass psychische und körperliche Vorgänge im GIT eng miteinander verbunden sind, unter anderem über hormonelle Systeme oder das vegetative Nervensystem mit Sympathikus und Parasympathikus. Solche Zusammenhänge werden sichtbar, wenn wir in angstauslösenden Situationen wiederholt zur Toilette müssen oder Bauchschmerzen bekommen. Im Umkehrschluss können körperliche Beschwerden aber auch Ängste auslösen und bei länger anhaltenden Symptomen zu depressiven Verstimmungen führen. Hierbei spielt die Hirn-Darm-Achse eine wichtige Rolle.

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

Access this chapter

Subscribe and save

Springer+ Basic
$34.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 34.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

Literatur

  1. Furness, J.B., The enteric nervous system and neurogastroenterology. Nat Rev Gastroenterol Hepatol, 2012. 9(5): p. 286–94.

    Google Scholar 

  2. Sasselli, V., V. Pachnis, and A.J. Burns, The enteric nervous system. Dev Biol, 2012. 366(1): p. 64–73.

    Google Scholar 

  3. J.B., F., The Enteric Nervous System. 2006, Oxford: Wiley-Blackwell.

    Google Scholar 

  4. Lazarus, R.S. and S. Folkman, Stress, appraisal, and coping. 1984, New York: Springer.

    Google Scholar 

  5. Mirsky, I.A., et al., Etiology of duodenal ulcer. I. Relation of specific psychological characteristics to rate of gastric secretion (serum pepsinogen). Psychosom Med, 1957. 19(1): p. 1–10.

    Google Scholar 

  6. Engel, G.L., Studies of ulcerative colitis I. Clinical data bearing on the nature of the somatic process. Psychosom Med, 1954. 16(6): p. 496–501.

    Google Scholar 

  7. Whitehead, W.E., et al., Learned illness behavior in patients with irritable bowel syndrome and peptic ulcer. Dig Dis Sci, 1982. 27(3): p. 202–8.

    Google Scholar 

  8. Tache, Y., et al., Brain and Gut CRF Signaling: Biological Actions and Role in the Gastrointestinal Tract. Curr Mol Pharmacol, 2018. 11(1): p. 51–71.

    Google Scholar 

  9. Kozakai, Y., et al., The role of peripheral corticotropin-releasing factor signaling in a rat model of stress-induced gastric hyperalgesia. Biochem Biophys Res Commun, 2019. 519(4): p. 797–802.

    Google Scholar 

  10. Nozu, T. and T. Okumura, Corticotropin-releasing factor receptor type 1 and type 2 interaction in irritable bowel syndrome. J Gastroenterol, 2015. 50(8): p. 819–30.

    Google Scholar 

  11. Almy, T.P., F. Kern, Jr., and F.K. Abbot, Constipation and diarrhea, as reactions to life stress. Res Publ Assoc Res Nerv Ment Dis, 1949. 29: p. 724–31.

    Google Scholar 

  12. Almy, T.P., F. Kern, Jr., and M. Tulin, Alterations in colonic function in man under stress; experimental production of sigmoid spasm in healthy persons. Gastroenterology, 1949. 12(3): p. 425–36.

    Google Scholar 

  13. Steadman, C.J., et al., Control of muscle tone in the human colon. Gut, 1992. 33(4): p. 541–6.

    Google Scholar 

  14. Gorard, D.A., et al., Intestinal transit in anxiety and depression. Gut, 1996. 39(4): p. 551–5.

    Google Scholar 

  15. Cao, S.G., et al., Effects of psychological stress on small intestinal motility and expression of cholecystokinin and vasoactive intestinal polypeptide in plasma and small intestine in mice. World J Gastroenterol, 2005. 11(5): p. 737–40.

    Google Scholar 

  16. Welgan, P., H. Meshkinpour, and M. Beeler, Effect of anger on colon motor and myoelectric activity in irritable bowel syndrome. Gastroenterology, 1988. 94(5 Pt 1): p. 1150–6.

    Google Scholar 

  17. Welgan, P., H. Meshkinpour, and L. Ma, Role of anger in antral motor activity in irritable bowel syndrome. Dig Dis Sci, 2000. 45(2): p. 248–51.

    Google Scholar 

  18. Rao, S.S., et al., Psychological and physical stress induce differential effects on human colonic motility. Am J Gastroenterol, 1998. 93(6): p. 985–90.

    Google Scholar 

  19. Tache, Y. and M. Million, Role of corticotropin-releasing factor signaling in stress-related alterations of colonic motility and hyperalgesia. J Neurogastroenterol Motil, 2015. 21(1): p. 8–24.

    Google Scholar 

  20. Tache, Y., Corticotrophin-releasing factor 1 activation in the central amygdale and visceral hyperalgesia. Neurogastroenterol Motil, 2015. 27(1): p. 1–6.

    Google Scholar 

  21. Trzeciak, P. and M. Herbet, Role of the Intestinal Microbiome, Intestinal Barrier and Psychobiotics in Depression. Nutrients, 2021. 13(3).

    Google Scholar 

  22. Koren, O., et al., A guide to enterotypes across the human body: meta-analysis of microbial community structures in human microbiome datasets. PLoS Comput Biol, 2013. 9(1): p. e1002863.

    Google Scholar 

  23. Dusko Ehrlich, S. and MetaH.I.T.consortium. Metagenomics of the intestinal microbiota: potential applications. Gastroenterol Clin Biol, 2010. 34 Suppl 1: p. S23–8.

    Google Scholar 

  24. Sudo, N., et al., Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol, 2004. 558(Pt 1): p. 263–75.

    Google Scholar 

  25. Bailey, M.T., et al., Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain Behav Immun, 2011. 25(3): p. 397–407.

    Google Scholar 

  26. Greenwood-Van Meerveld, B., A.C. Johnson, and D. Grundy, Gastrointestinal Physiology and Function. Handb Exp Pharmacol, 2017. 239: p. 1–16.

    Google Scholar 

  27. Tache, Y. and M.H. Perdue, Role of peripheral CRF signalling pathways in stress-related alterations of gut motility and mucosal function. Neurogastroenterol Motil, 2004. 16 Suppl 1: p. 137–42.

    Google Scholar 

  28. Pavlov, V.A., S.S. Chavan, and K.J. Tracey, Molecular and Functional Neuroscience in Immunity. Annu Rev Immunol, 2018. 36: p. 783–812.

    Google Scholar 

  29. Overman, E.L., J.E. Rivier, and A.J. Moeser, CRF induces intestinal epithelial barrier injury via the release of mast cell proteases and TNF-alpha. PLoS One, 2012. 7(6): p. e39935.

    Google Scholar 

  30. Yuan, P.Q., et al., Urocortins and CRF receptor type 2 variants in the male rat colon: gene expression and regulation by endotoxin and anti-inflammatory effect. Am J Physiol Gastrointest Liver Physiol, 2016. 310(6): p. G387–98.

    Google Scholar 

  31. Tetel, M.J., et al., Steroids, stress and the gut microbiome-brain axis. J Neuroendocrinol, 2018. 30(2).

    Google Scholar 

  32. Kao, A.C., S. Harty, and P.W. Burnet, The Influence of Prebiotics on Neurobiology and Behavior. Int Rev Neurobiol, 2016. 131: p. 21–48.

    Google Scholar 

  33. Barbosa, R.S.D. and M.A. Vieira-Coelho, Probiotics and prebiotics: focus on psychiatric disorders – a systematic review. Nutr Rev, 2020. 78(6): p. 437–450.

    Google Scholar 

  34. Ligezka, A.N., et al., A systematic review of microbiome changes and impact of probiotic supplementation in children and adolescents with neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry, 2021. 108: p. 110187.

    Google Scholar 

  35. Rincel, M. and M. Darnaudery, Maternal separation in rodents: a journey from gut to brain and nutritional perspectives. Proc Nutr Soc, 2020. 79(1): p. 113–132.

    Google Scholar 

  36. Liu, R.T., R.F.L. Walsh, and A.E. Sheehan, Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci Biobehav Rev, 2019. 102: p. 13–23.

    Google Scholar 

  37. Ansari, F., et al., The Effects of Probiotics and Prebiotics on Mental Disorders: A Review on Depression, Anxiety, Alzheimer, and Autism Spectrum Disorders. Curr Pharm Biotechnol, 2020. 21(7): p. 555–565.

    Google Scholar 

  38. Ritchie, J., Pain from distension of the pelvic colon by inflating a balloon in the irritable colon syndrome. Gut, 1973. 14(2): p. 125–32.

    Google Scholar 

  39. Elsenbruch, S. & Labrenz, F. Nocebo effects and experimental models in visceral pain. International Review of Neurobiology, 2018. 138, 285–306.

    Google Scholar 

  40. Ford AC, Sperber AD, Corsetti M, Camilleri M. Irritable bowel syndrome. Lancet. 2020. 21;396(10263):1675–1688. https://doi.org/10.1016/S0140-6736(20)31548-8. Epub 2020 Oct 10. PMID: 33049223.

  41. Schmulson MJ. Brain-gut interaction in irritable bowel syndrome: new findings of a multicomponent disease model. Isr Med Assoc J. 2001. 3(2):104–10. PMID: 11347592.

    Google Scholar 

  42. Zar S, Benson MJ, Kumar D. Rectal afferent hypersensitivity and compliance in irritable bowel syndrome: differences between diarrhoea-predominant and constipation-predominant subgroups. Eur J Gastroenterol Hepatol. 2006. 18(2):151–8. https://doi.org/10.1097/00042737-200602000-00007. PMID: 16394796.

  43. Larauche M, Mulak A, Taché Y. Stress and visceral pain: from animal models to clinical therapies. Exp Neurol. 2012. 233(1):49–67. https://doi.org/10.1016/j.expneurol.2011.04.020. Epub 2011 May 6. PMID: 21575632; PMCID: PMC3224675.

  44. W., J.n. and H.b. H.-J., Physiologie und Pathophysiologie viszeraler Schmerzen. Schmerz, 2002. 16: p. 429–445.

    Google Scholar 

  45. Johnson AC, Tran L, Schulkin J, Greenwood-Van Meerveld B. Importance of stress receptor-mediated mechanisms in the amygdala on visceral pain perception in an intrinsically anxious rat. Neurogastroenterol Motil. 2012. 24(5):479–86, e219. https://doi.org/10.1111/j.1365-2982.2012.01899.x. Epub 2012 Feb 26. PMID: 22364507; PMCID: PMC3461498.

  46. Dolan, R.J., Emotion, cognition, and behavior. Science, 2002. 298(5596): p. 1191–4.

    Google Scholar 

  47. Tillisch K, Mayer EA, Labus JS. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology. 2011. 140(1):91–100. https://doi.org/10.1053/j.gastro.2010.07.053. Epub 2010 Aug 7. PMID: 20696168; PMCID: PMC3253553.

  48. Elsenbruch S, Rosenberger C, Bingel U, Forsting M, Schedlowski M, Gizewski ER. Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli. Gastroenterology. 2010. 139(4):1310–9. https://doi.org/10.1053/j.gastro.2010.06.054. Epub 2010 Jun 22. PMID: 20600024.

  49. Harlow, H.F., The nature of love. American Psychologist, 1958. 13(12): p. 673–685.

    Google Scholar 

  50. Lopes, L.V., et al., Maternal deprivation affects the neuromuscular protein profile of the rat colon in response to an acute stressor later in life. J Proteomics, 2008. 71(1): p. 80–8.

    Google Scholar 

  51. Medland, J.E., et al., Early life adversity in piglets induces long-term upregulation of the enteric cholinergic nervous system and heightened, sex-specific secretomotor neuron responses. Neurogastroenterol Motil, 2016. 28(9): p. 1317–29.

    Google Scholar 

  52. Thapar (2020) Paediatric functional abdominal pain disorders, Nature Reviews 6: 89

    Google Scholar 

  53. Bolten M. (2019) Klinische Bindungsforschung. In: Schneider S., Margraf J. (eds) Lehrbuch der Verhaltenstherapie, Band 3. Abb. 4.1 /ISBN 978-3-662-57369-3. Berlin, Heidelberg: Springer

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Margarete Bolten .

Rights and permissions

Reprints and permissions

Copyright information

© 2022 Der/die Autor(en), exklusiv lizenziert an Springer-Verlag GmbH, DE, ein Teil von Springer Nature

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Bolten, M. (2022). Psychosoziale Aspekte der Nahrungsaufnahme, Verdauung und Ausscheidung. In: Funktionelle Magen-Darm-Störungen im Kindes- und Jugendalter. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-662-64253-5_2

Download citation

  • DOI: https://doi.org/10.1007/978-3-662-64253-5_2

  • Published:

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-662-64252-8

  • Online ISBN: 978-3-662-64253-5

  • eBook Packages: Medicine (German Language)

Publish with us

Policies and ethics