Vol 59, No 1 (2021)
Original paper
Published online: 2021-01-27

open access

Page views 1143
Article views/downloads 793
Get Citation

Connect on Social Media

Connect on Social Media

Reactivity of astrocytes in hippocampal CA1 area in rats after administration of habanero peppers

Jadwiga Jaworska-Adamu1, Aleksandra Krawczyk1, Karol Rycerz1, Marcin Golynski2
Pubmed: 33502751
Folia Histochem Cytobiol 2021;59(1):1-7.

Abstract

Introduction. Astrocytes react to microenvironmental changes. Their reactivity is manifested by an increase in glial fibrillary acidic protein (GFAP) and S100b protein levels, hypertrophy and hyperplasia. The aim of the study was to analyse immunoreactive GFAP (GFAP-IR) and S100b (S100b-IR) astrocytes of hippocampal CA1 area in adult rats intragastrically (i.g.) treated with habanero peppers.

Material and methods. Brains from 10 control rats (group C) and 10 rats receiving oil suspension of habanero fruits for 7 days (group I-7) or 28 days (group II-28) were used. Antibodies against GFAP and S100b were used for immunohistochemistry. Morphology and distribution of astrocytes was evaluated under light microscope and their density was quantitatively analysed.

Results. In the CA1 hippocampal area of group II-28 rats, GFAP-IR cells with numerous, branched processes were observed. S100b-IR astrocytes had delicate, single processes in comparison with cells without processes observed in groups I-7 and C. In groups I-7 and II-28, GFAP-IR astrocytes’ density significantly increased in SR — stratum radiatum of hippocampal CA1 area. In group I-7, a density of cells with the expression of S100b was significantly increased in SO — stratum oriens layer. In group II-28, the density of S100b-IR astrocytes was decreased.

Conclusions. Habanero peppers administrated to rats, especially for a longer time, caused reactive changes in the astrocytes in hippocampal CA1 area, and thus these glial cells may protect neurons against excitotoxic damage.

Article available in PDF format

View PDF Download PDF file

References

  1. Artinian J, Jordan A, Khlaifia A, et al. Regulation of Hippocampal Memory by mTORC1 in Somatostatin Interneurons. J Neurosci. 2019; 39(43): 8439–8456.
  2. Guerra-Gomes S, Sousa N, Pinto L, et al. Functional Roles of Astrocyte Calcium Elevations: From Synapses to Behavior. Front Cell Neurosci. 2017; 11: 427.
  3. Ota Y, Zanetti AT, Hallock RM. The role of astrocytes in the regulation of synaptic plasticity and memory formation. Neural Plast. 2013; 2013: 185463.
  4. Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev. 2018; 98(1): 239–389.
  5. Landis DM. The early reactions of non-neuronal cells to brain injury. Annu Rev Neurosci. 1994; 17: 133–151.
  6. Siracusa R, Fusco R, Cuzzocrea S. Astrocytes: Role and Functions in Brain Pathologies. Front Pharmacol. 2019; 10: 1114.
  7. Cano V, Valladolid-Acebes I, Hernández-Nuño F, et al. Morphological changes in glial fibrillary acidic protein immunopositive astrocytes in the hippocampus of dietary-induced obese mice. Neuroreport. 2014; 25(11): 819–822.
  8. Viola GG, Rodrigues L, Américo JC, et al. Morphological changes in hippocampal astrocytes induced by environmental enrichment in mice. Brain Res. 2009; 1274: 47–54.
  9. Okere CO, Waterhouse BD. Capsaicin increases GFAP and glutamine synthetase immunoreactivity in rat arcuate nucleus and median eminence. Neuroreport. 2004; 15(2): 255–258.
  10. Bennion D, Jensen T, Walther C, et al. Transient receptor potential vanilloid 1 agonists modulate hippocampal CA1 LTP via the GABAergic system. Neuropharmacology. 2011; 61(4): 730–738.
  11. Gibson HE, Edwards JG, Page RS, et al. TRPV1 channels mediate long-term depression at synapses on hippocampal interneurons. Neuron. 2008; 57(5): 746–759.
  12. Rycerz K, Krawczyk A, Jaworska-Adamu J, et al. Immunoreactivity of arcuate nucleus astrocytes in rats after intragastric administration of habanero peppers (Capsicum Chinese Jacq.). Pol J Vet Sci. 2016; 19(4): 809–817.
  13. Thompson RJ. A direct demonstration of functional TRPV1 in Cajal-Retzius cells. J Physiol. 2018; 596(16): 3451–3452.
  14. El Falougy H, Kubikova E, Benuska J. The microscopical structure of the hippocampus in the rat. Bratisl Lek Listy. 2008; 109(3): 106–110.
  15. Knierim JJ. The hippocampus. Curr Biol. 2015; 25(23): R1116–R1121.
  16. König JFR, Klippel RAA. stereotactic atlas of the forebrain and lower parts of the brain stem. Williams and Wilkins: Baltimore. ; 1963.
  17. Krawczyk A, Jaworska-Adamu J. Reactivity of astrocytes in the periaqueductal gray matter of rats treated with monosodium glutamate. Folia Histochem Cytobiol. 2020; 58(2): 147–155.
  18. Kimelberg HK. The problem of astrocyte identity. Neurochem Int. 2004; 45(2-3): 191–202.
  19. Hurtado-Zavala JI, Ramachandran B, Ahmed S, et al. TRPV1 regulates excitatory innervation of OLM neurons in the hippocampus. Nat Commun. 2017; 8: 15878.
  20. Li HB, Mao RR, Zhang JC, et al. Antistress effect of TRPV1 channel on synaptic plasticity and spatial memory. Biol Psychiatry. 2008; 64(4): 286–292.
  21. Magó Á, Weber JP, Ujfalussy BB, et al. Synaptic Plasticity Depends on the Fine-Scale Input Pattern in Thin Dendrites of CA1 Pyramidal Neurons. J Neurosci. 2020; 40(13): 2593–2605.
  22. Ruan YW, Zou B, Fan Y, et al. Dendritic plasticity of CA1 pyramidal neurons after transient global ischemia. Neuroscience. 2006; 140(1): 191–201.
  23. Steward O, Torre ER, Tomasulo R, et al. Neuronal activity up-regulates astroglial gene expression. Proc Natl Acad Sci U S A. 1991; 88(15): 6819–6823.
  24. Middeldorp J, Hol EM. GFAP in health and disease. Prog Neurobiol. 2011; 93(3): 421–443.
  25. Barker AJ, Ullian EM. Astrocytes and synaptic plasticity. Neuroscientist. 2010; 16(1): 40–50.
  26. Ventura R, Harris KM. Three-dimensional relationships between hippocampal synapses and astrocytes. J Neurosci. 1999; 19(16): 6897–6906.
  27. Hawrylak N, Chang FL, Greenough WT. Astrocytic and synaptic response to kindling in hippocampal subfield CA1. II. Synaptogenesis and astrocytic process increases to in vivo kindling. Brain Res 1993; 603: 309-316. ; 8461983.
  28. Armstrong DD. The neuropathology of temporal lobe epilepsy. J Neuropathol Exp Neurol. 1993; 52(5): 433–443.
  29. Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer's disease. Trends Neurosci. 2008; 31(9): 454–463.
  30. Shobha K, Vijayalakshmi K, Alladi PA, et al. Altered in-vitro and in-vivo expression of glial glutamate transporter-1 following exposure to cerebrospinal fluid of amyotrophic lateral sclerosis patients. J Neurol Sci. 2007; 254(1-2): 9–16.
  31. Zhou J, Sutherland ML. Glutamate transporter cluster formation in astrocytic processes regulates glutamate uptake activity. J Neurosci. 2004; 24(28): 6301–6306.
  32. Sullivan SM, Sullivan RKP, Miller SM, et al. Phosphorylation of GFAP is associated with injury in the neonatal pig hypoxic-ischemic brain. Neurochem Res. 2012; 37(11): 2364–2378.
  33. Michetti F, D'Ambrosi N, Toesca A, et al. The S100B story: from biomarker to active factor in neural injury. J Neurochem. 2019; 148(2): 168–187.
  34. Tramontina F, Tramontina AC, Souza DF, et al. Glutamate uptake is stimulated by extracellular S100B in hippocampal astrocytes. Cell Mol Neurobiol. 2006; 26(1): 81–86.
  35. Sakatani S, Seto-Ohshima A, Shinohara Y, et al. Neural-activity-dependent release of S100B from astrocytes enhances kainate-induced gamma oscillations in vivo. J Neurosci. 2008; 28(43): 10928–10936.
  36. Abdel-Salam OME, Sleem AA, Sayed MA, et al. Capsaicin Exerts Anti-convulsant and Neuroprotective Effects in Pentylenetetrazole-Induced Seizures. Neurochem Res. 2020; 45(5): 1045–1061.
  37. Donato R. Intracellular and extracellular roles of S100 proteins. Microsc Res Tech. 2003; 60(6): 540–551.
  38. Huang M, Cheng G, Tan H, et al. Capsaicin protects cortical neurons against ischemia/reperfusion injury via down-regulating NMDA receptors. Exp Neurol. 2017; 295: 66–76.
  39. Yasuda Y, Tateishi N, Shimoda T, et al. Relationship between S100beta and GFAP expression in astrocytes during infarction and glial scar formation after mild transient ischemia. Brain Res. 2004; 1021(1): 20–31.