PL EN


Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników
2020 | 80 | 2 |

Tytuł artykułu

Intracerebroventricular streptozotocin induces behavioral impairments and increases short‑term C3 gene expression in the hippocampus of Wistar rats

Warianty tytułu

Języki publikacji

EN

Abstrakty

EN
A non‑transgenic rat model based on intracerebroventricular injection of streptozotocin (STZ) has been used as an animal model to investigate mechanisms associated to the late onset of sporadic Alzheimer’s disease, such as anatomical and behavioral impairments. However, molecular aspects related to gene expression, mainly in the hippocampus, require more investigation. Thus, this study evaluated the early and late cognitive functions and hippocampal gene expression after STZ administration. Male Wistar rats were divided into 4 groups: STZ (injected bilaterally), control group for the early memory function evaluation (1 month after surgery = phase 1, same volume of vehicle), and the same treatment for the late memory function evaluation (4 months after surgery = phase 2). The animals were observed in the elevated plus maze to assess behaviors related to anxiety, risk‑assessment and fear‑related memories. The behavioral tests were followed by brain removal and hippocampal dissection for RNA extraction and qRT‑PCR to assess the expression levels of 4 Alzheimer’s disease related genes: Mapt, Apoe, C3 and Ps‑1. Animals from both phases showed increased time percentage and number of entries into the open arms, indicating risk behavior associated with anxiety, and an increased time percentage in the center square for both exposures (re‑test) when compared to the control group, suggesting working memory impairment related to an aversive event. Statistical analyses indicated that the STZ group presented alterations in anxiety, memory and risk assessment responses. Additionally, one month after STZ administration, C3 gene assays revealed an increased expression. Therefore, current data indicate that neuroinflammatory events linked to the expression of pro inflammatory cytokines such as C3 are related to memory, anxiety and decision‑making alterations.

Słowa kluczowe

Wydawca

-

Rocznik

Tom

80

Numer

2

Opis fizyczny

p.168-171,fig.,ref.

Twórcy

  • Post Graduation Program in Biotechnology, Universidade Positivo, Curitiba - PR, Brasil
autor
  • Post Graduation Program in Biotechnology, Universidade Positivo, Curitiba - PR, Brasil
  • Post Graduation Program in Biotechnology, Universidade Positivo, Curitiba - PR, Brasil
autor
  • Post Graduation Program in Biotechnology, Universidade Positivo, Curitiba - PR, Brasil

Bibliografia

  • Bao J, Mahaman YAR, Liu R, Wang JZ, Zhang Z, Zhang B, Wang X (2017) Sex Differences in the cognitive and hippocampal effects of streptozotocin in an animal model of sporadic AD. Front Aging Neurosci 9: 1–12.
  • Bartolotti N, Lazarov O (2016) Lifestyle and Alzheimer’s Disease. In: The Role of Environmental Factors in Disease Development. Genes, Environment and Alzheimer’s Disease. Elsevier Inc. p. 197–237.
  • Bertoglio LJ, Carobrez AP (2000) Previous maze experience required to increase open arms avoidance in rats submitted to the elevated plus‑maze model of anxiety. Behav Brain Res 108: 197–203.
  • Bertoglio LJ, Carobrez AP (2002) Prior maze experience required to alter midazolam effects in rats submitted to the elevated plus‑maze. Pharmacol Biochem Behav 72: 449–455.
  • Blanchard DC, Blanchard RJ, Rodgers RJ (1991) Risk assessment and animal models of anxiety. In: Animal Models in Psychopharmacology (Olivier B, Mos J, Slangen JL, Eds.). Basel, Birkhauser, p. 117–134.
  • Blanchard RJ, Yudko EB, Rodgers RJ, Blanchard DC (1993) Defense system psychopharmacology: an ethological approach to the pharmacology of fear and anxiety. Behav Brain Res 58: 155–165.
  • Blanco E, Campos‑Sandoval JA, Palomino A, Luque‑Rojas MJ, Bilbao A, Suárez J, Márquez J, de Fonseca FR (2012) Cocaine modulates both glutaminase gene expression and glutaminase activity in the brain of co‑ caine‑sensitized mice. Psychopharmacology 219: 933–944.
  • Carobrez AP, Bertoglio LJ (2005) Ethological and temporal analysis of anxiety‑like behavior: the elevated plus‑maze model 20 years on. Neurosci Biobehav Rev 29: 1193–1205.
  • Carobrez AP, Teixeira K V, Graeff FG (2001) Modulation of defensive behavior by periaqueductal gray NMDA/glycine‑B receptor. Neurosci Biobehav Rev 25: 697–709.
  • Chen Y, Liang Z, Blanchard J, Dai CL, Sun S, Lee MH, Grundke‑Iqbal I, Iqbal K, Liu F, Gong CX (2012) A non‑transgenic mouse model (icv‑STZ mouse) of Alzheimer’s disease: similarities to and differences from the trans‑ genic model (3xTg‑AD mouse). Mol Neurobiol 47: 1–15.
  • Cuchillo‑Ibanez I, Seereeram A, Byers HL, Leung KY, Ward M, Anderton BH, Hanger DP (2008) Phosphorylation of TAU regulates its axonal transport by controlling its binding to kinesin. FASEB J 22: 3186–3195.
  • Da Silva, I, Corbellini, J, Pfutzenreuter G, Maranho L, Pincerati M (2019) Leaf extract of Eugenia uniflora L. prevents episodic memory impairment induced by streptozotocin in rats. Pharmacognosy Res 11: 329–332.
  • Ding B, Xi Y, Gao M, Li Z, Xu C, Fan S, He W (2014) Gene expression profiles of entorhinal cortex in Alzheimer’s disease. Am J Alzheimer’s Dis Other Demen 29: 526–532.
  • Dorszewska J, Prendecki M, Oczkowska A, Dezor M, Kozubski W (2016) Molecular basis of familial and sporadic Alzheimer’s disease. Curr Alzheimer Res 13: 952–963.
  • Dubey M, Chaudhury P, Kabiru H, Shea TB (2008) TAU inhibits anterograde axonal transport and perturbs stability in growing axonal neurites in part by displacing kinesin cargo: Neurofilaments attenuate TAU‑mediated neurite instability. Cell Motil Cytoskeleton 65: 89–99.
  • Duff K, Eckman C, Zehr C, Yu X, Prada CM, Perez‑Tur J, Hutton M, Buee L, Harigaya Y, Yager D, Morgan D, Gordon MN, Holcomb L, Refolo L, Zenk B, Hardy J, Younkin S (1996) Increased amyloid‑beta42(43) in brains of mice expressing mutant presenilin 1. Nature 383: 710–713.
  • Elder GA, Gama Sosa MA, De Gasperi R (2010) Transgenic mouse models of Alzheimer’s disease. Mt Sinai J Med 77: 69–81.
  • File SE, Mabbutt PS, Hitchott PK (1990) Characterization of phenomenon of ‘one‑trial tolerance’ to the anxiolytic effect of chlordiazepoxide in the elevated plus‑maze. Psychopharmacology 102: 98–101.
  • Flanigan TJ, Xue Y, Rao SK, Dhanushkodi A, McDonald MP (2014) Abnormal vibrissa‑related behavior and loss of barrel field inhibitory neurons in 5xFAD transgenics. Genes Brain Behav 13: 488–500.
  • Goedert  M, Sisodia SS, Price DL (1991) Neurofibrillary tangles and beta‑amyloid deposits in Alzheimer’s disease. Curr Opin Neurobiol 1: 441–447.
  • Gong CX, Liu F, Grundke‑Iqbal I, Iqbal K (2006) Impaired brain glucose metabolism leads to Alzheimer neurofibrillary degeneration through a decrease in tau O‑GlcNAcylation. J Alzheimers Dis 9: 1–12.
  • Grieb P (2016) Intracerebroventricular streptozotocin injections as a model of Alzheimer’s disease: in search of a relevant mechanism. Mol Neurobiol 53: 1741–1752.
  • Grünblatt E, Hoyer S, Riederer P (2004) Gene expression profile in streptozotocin rat model for sporadic Alzheimer’s disease. J Neural Transm 111: 367–386.
  • Grünblatt E, Salkovic‑Petrisic M, Osmanovic J, Riederer P, Hoyer S (2007) Brain insulin system dysfunction in streptozotocin intracerebroventricularly treated rats generates hyperphosphorylated TAU protein. J Neurochem 101: 757–770.
  • Gupta S, Yadav K, Mantri SS, Singhal NK, Ganesh S, Sandhir R (2018) Evidence for compromised insulin signaling and neuronal vulnerability in experimental model of sporadic Alzheimer’s disease. Mol Neurobiol 55: 8916–8935.
  • Hatami H, Hossainpour‑Faizi MA, Azarfarin M, Azarfam P (2010) Chronic ecstasy use increases neurotrophin‑4 gene expression and protein levels in the rat brain. Pharmacol Rep 62: 998–1004.
  • Heiss WD, Szelies B, Kessler J, Herholz K (1991) Abnormalities of energy metabolism in Alzheimer’s disease studied with PET. Ann N Y Acad Sci 640: 65–71.
  • Holtzman DM, Bales KR, Tenkova T, Fagan AM, Parsadanian M, Sartorius LJ, Mackey B, Olney J, McKeel D, Wozniak D, Paul SM (2000) Apolipoprotein E isoform‑dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci 97: 2892–2897.
  • Hosseinzadeh S, Zahmatkesh M, Heidari M, Hassanzadeh GR, Karimian M, Sarrafnejad A, Zarrindast MR (2015) Hippocampal DHCR24 down regulation in a rat model of streptozotocin‑induced cognitive decline. Neurosci Lett 587: 107–112.
  • Iqbal K, Grundke‑Iqbal I (2005) Metabolic/signal transduction hypothesis of Alzheimer’s disease and other tauopathies. Acta Neuropathol 109: 25–31.
  • Jawhar S, Trawicka A, Jenneckens C, Bayer TA, Wirths O (2012) Motor deficits, neuron loss, and reduced anxiety coinciding with axonal degeneration and intraneuronal AB aggregation in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging 33: 196.
  • Kamat P, Kalani A, Rai S, Tota S, Kumar A, Ahmad A (2016) Streptozotocin intracerebroventricular‑induced neurotoxicity and brain insulin resistance: a therapeutic intervention for treatment of sporadic Alzheimer’s disease (sAD)‑like pathology. Mol Neurobiol 53: 4548–4562.
  • Kamat PK (2015) Streptozotocin induced Alzheimer’s disease like changes and the underlying neural degeneration and regeneration mechanism. Neural Regen Res 10: 1050–1052.
  • Kar S, Slowikowski SPM, Westaway D, Mount HTJ (2004) Interactions between beta‑amyloid and central cholinergic neurons: implications for Alzheimer’s disease. J Psychiatry Neurosci 29: 427–441.
  • Lamprea MR, Cardenas FP, Silveira R, Morato S, Walsh TJ (2000) Dissociation of memory and anxiety in a repeated elevated plus maze paradigm: Forebrain cholinergic mechanisms. Behav Brain Res 117: 97–105.
  • Lester‑Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR, de la Monte SM (2006) Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadic Alzheimer’s disease. J Alzheimers Dis 9: 13–33.
  • Lister RG (1987) The use of a plus‑maze to measure anxiety in the mouse. Psychopharmacology 92: 180–185.
  • Liu CC, Kanekiyo T, Xu H, Bu G (2013) Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 9: 106–118.
  • Maier M, Peng Y, Jiang L, Seabrook TJ, Carroll MC, Lemere CA (2008) Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci 28: 6333–6341.
  • Miller JA, Woltjer RL, Goodenbour JM, Horvath S, Geschwind DH (2013) Genes and pathways underlying regional and cell type changes in Alzheimer’s disease. Genome Med 5: 48.
  • Montgomery KC, Monkman J (1955) The relation between fear and exploratory behavior. J Comp Physiol Psychol 48: 132–136.
  • Mutlu O, Akar F, Celikyurt IK, Tanyeri P, Ulak G, Erden F (2015) 7‑NI and ODQ disturbs memory in the elevated plus maze, Morris water maze, and radial arm maze tests in mice. Drug Target Insights 9: 1–8.
  • Nelson PT, Braak H, Markesbery WR (2009) Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship. J Neuropathol Exp Neurol 68: 1–14.
  • Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet‑Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R (2006) Intraneuronal beta‑amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci 26: 10129–10140.
  • Paxinos G, Watson C (2013) The Rat Brain in Stereotaxic Coordinates, Academic Press, Sydney. Pericak‑Vance MA, Bebout JL, Gaskell PC, Yamaoka LH, Hung WY, Alberts MJ, Walker AP, Bartlett RJ, Haynes CA, Welsh KA (1991) Linkage studies in familial Alzheimer disease: evidence for chromosome 19 linkage. Am J Hum Genet 48: 1034–1050.
  • Rai S, Kamat P, Nath C, Shukla R (2014) Glial activation and post‑synaptic neurotoxicity: The key events in Streptozotocin (ICV) induced memory impairment in rats. Pharmacol Biochemistry Behavior 117: 104–117.
  • Rasmussen DD, Mitton DR, Green J, Puchalski S (2001) Chronic daily ethanol and withdrawal: 2. Behavioral changes during prolonged abstinence. Alcohol Clin Exp Res 25: 999–1005.
  • Ricceri  L, Luisa  M, Anna  M, Patrizia P, Annamaria C, Roberta S, Paola P, Maria L S, Monica L, Gemma C (2004) Cognitive and neurological deficits induced by early and prolonged basal forebrain cholinergic hypofunction in rats. Experimental Neurol 189: 162–172.
  • Rodgers RJ, Cao BJ, Dalvi A, Holmes A (1997) Animal models of anxiety: An ethological perspective. Brazilian J Med Biol Res 30: 289–304.
  • Salkovic‑Petrisic M, Osmanovic‑Barilar J, Brückner MK, Hoyer S, Arendt T, Riederer P (2011) Cerebral amyloid angiopathy in streptozotocin rat model of sporadic Alzheimer’s disease: A long‑term follow up study. J Neural Transm 118: 765–772.
  • Santos TO, Mazucanti CHY, Xavier GF, da Silva TA (2012) Early and late neurodegeneration and memory disruption after intracerebroventricular streptozotocin. Physiol Behav 107: 401–413.
  • Sharma AC, Kulkarni SK (1992) Evaluation of learning and memory mechanisms employing elevated plus‑maze in rats and mice. Prog Neuropsy‑ chopharmacol Biol Psychiatry 16: 117–125.
  • Shoham S, Bejar C, Kovalev E, Weinstock  M (2003) Intracerebroventricular injection of streptozotocin causes neurotoxicity to myelin that contributes to spatial memory deficits in rats. Exp Neurol 184: 1043–1052.
  • Soria‑Fregozo C, Pérez‑Vega MI, González‑Burgos I, Feria‑Velasco A, Beas‑Zárate C (2008) Prefrontal serotonergic denervation induces in‑crease in the density of 5‑HT2A receptors in adult rat prefrontal cortex. Neurochem Res 33: 2350–2357.
  • Tanyeri P (2014) Zaprinast and rolipram enhances spatial and emotional memory in the elevated plus maze and passive avoidance tests and diminishes exploratory activity in naive mice. Med Sci Monit Basic Res 20: 105–111.
  • Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW (1975) A protein factor essential for microtubule assembly. Proc Natl Acad Sci 72: 1858–1862.
  • Xiang Z, Haroutunian V, Ho L, Purohit D, Pasinetti GM (2006) Microglia activation in the brain as inflammatory biomarker of Alzheimer’s disease neuropathology and clinical dementia. Dis Markers 22: 95–102.
  • Yamada M, Iwabuchi T, Takahashi K, Kurahashi C, Ohata H, Honda K (2005) Identification and expression of frizzled‑3 protein in rat frontal cortex after antidepressant and electroconvulsive treatment. J Pharmacol Sci 99: 239–246.

Typ dokumentu

Bibliografia

Identyfikatory

Identyfikator YADDA

bwmeta1.element.agro-9e0d35e6-b3ea-4d88-975b-cbc8b6b5c93d
JavaScript jest wyłączony w Twojej przeglądarce internetowej. Włącz go, a następnie odśwież stronę, aby móc w pełni z niej korzystać.