Disturbance in Expression of Lactate Transporters in Brain Cells under Acute Toxic Effect of Beta-Amyloid In Vitro and In Vivo

Cover Page

Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription Access

Abstract

Decreased energy metabolism in the brain correlates with cognitive impairment in Alzheimer’s disease. Accumulating experimental data indicate that lactate transporters and monocarboxylate transporters (MCTs) are directly involved in cerebral energy metabolism. However, to date, changes in lactate levels and MCT content in Alzheimer’s disease remain unclear. The aim of the study was to study the content of lactate and of its transporters – MCT1 and MCT2 in cells of neuronal, astroglial and endothelial nature under acute toxic effects of beta-amyloid (Aβ1–42) in vitro and in vivo. Under conditions of acute toxic action of Aβ1–42 in vivo, a significant (P ≤ 0.05) decrease in the level of lactate in the hippocampal tissue and an increase (P ≤ 0.05) in the dialysate were found. At the same time, a low (P ≤ 0.05) levels of MCT1 and MCT2 was set. In vitro, significantly high (P ≤ 0.05) production of lactate by astrocytes was revealed, coupled with low (P ≤ 0.05) level of MCT2 on neurons. Thus, it was found that Aβ1–42 causes a decrease in the level of lactate in the hippocampal tissue and an increase in its level in dialysate in vivo, which correlates with the impaired level of MCT1 and MCT2. This indicates a violation of energy metabolism due to the acute toxic effect of Aβ1–42. At the same time, the revealed increase in the production of lactate by astrocytes in vitro may indicate the inclusion of a compensatory mechanism aimed at maintaining the astrocyte-neuronal interaction.

About the authors

Ya. V. Gorina

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University

Author for correspondence.
Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

E. V. Kharitonova

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

E. D. Khilazheva

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

A. A. Semenova

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

A. V. Morgun

Department of Outpatient Pediatrics and Propaedeutics of Childhood Diseases with a Postgraduate Course,
Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

Yu. K. Komleva

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University; Center for Collective Use Molecular and Cellular Technologies, Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk; Russia, 660022, Krasnoyarsk

O. L. Lopatina

Department of Outpatient Pediatrics and Propaedeutics of Childhood Diseases with a Postgraduate Course,
Voino-Yasenetsky Krasnoyarsk State Medical University

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk

A. B. Salmina

Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University; Laboratory of Neurobiology and Tissue Engineering, Brain Institute, Scientific Center of Neurology

Email: yana_20@bk.ru
Russia, 660022, Krasnoyarsk; Russia, 125367, Moscow

References

  1. Горина Я.В., Комлева Ю.К., Лопатина О.Л., Черных А.И., Салмина А.Б. 2017. Влияние инсулинорезистентности на нарушение метаболизма глюкозы в миндалине головного мозга при экспериментальной болезни Альцгеймера. Бюлл. сибирской мед. № 6. С. 1. (Gorina Ya.V., Komleva Yu.K., Lopatina O.L., Chernykh A.I., Salmina A.B. 2017. Influence of insulin resistance on increased risk of brain amygdala development in experimental Alzheimer’s disease. Bulletin of Siberian medicine. № 6. Р. 1.)
  2. Комлева Ю.К., Малиновская Н.А., Горина Я.В., Лопатина О.Л., Волкова В.В., Салмина А.Б. 2015. Экспрессия молекул CD38 и CD157 в ольфакторных луковицах головного мозга при экспериментальной болезни Альцгеймера. Сибирское медицинское обозрение. № 5. С. 45. (Komleva Yu.K., Malinovskaya N.A., Gorina Ya.V., Lopatina O.L., Volkova V.V., Salmina A.B. 2015. Expression of CD38 and CD157 molecules in the olfactory bulbs of the brain in experimental Alzheimer’s disease. Siberian Med. Rev. № 5. Р. 45.)
  3. Лобзин В.Ю., Одинак М.М., Фокин В.А., Воробьев С.В., Емелин А.Ю., Лупанов И.А., Кудяшева А.В., Соколов А.В. 2013. Метаболические изменения головного мозга при болезни Альцгеймера, сосудистой и смешанной деменции. Биомед. журнал медлайн.ру. С. 1085. (Lobzin V.Yu., Odinak M.M., Fokin V.A., Vorobyov S.V., Emelin A.Yu., Lupanov I.A., Kudyasheva A.V., Sokolov A.V. 2013. Brain metabolic changes in Alzheimer’s disease, vascular and mixed dementia. Biomed. J. Medline.ru. P. 1085.)
  4. Моргун А.В., Кувачева Н.В., Комлева Ю.К., Кутищева И.А., Окунева О.С., Дробушевская А.И., Хилажева Е.Д., Черепанов С.М., Салмина А.Б. 2013. Дифференцировка эмбриональных прогениторных клеток мозга крыс в астроциты и нейроны. Сибирское мед. обозрение. № 6. С. 9. (Morgun A.V., Kuvacheva N.V., Komleva Yu.K., Kutishcheva I.A., Okuneva O.S., Drobushevskaya A.I., Khilazheva E.D., Cherepanov S.M., Salmina A.B. 2013. Differentiation of rat brain embryonic progenitor cells into astrocytes and neurons. Siberian Med. Rev. № 6. Р. 9.)
  5. Allaman I., Gavillet M., Bélanger M., Laroche T., Viertl D., Lashuel H.A., Magistretti P.J. 2010. Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J. Neurosci. V. 30. P. 3326. https://doi.org/10.1523/JNEUROSCI.5098-09.2010
  6. Bartolotti N., Lazarov O. 2019. CREB signals as PBMC-based biomarkers of cognitive dysfunction: a novel perspective of the brain-immune axisBrain Behav. Immun. V. 78. P. 9. https://doi.org/10.1016/j.bbi.2019.01.004
  7. Bekinschtein P., Cammarota M., Katche C., Slipczuk L., Rossato J.I., Goldin A., Izquierdo I., Medina J.H. 2008. BDNF is essential to promote persistence of long-term memory storage. Proc. Natl. Acad. Sci. USA. V. 105. P. 2711. https://doi.org/10.1073/pnas.0711863105
  8. Bergersen L.H. 2015. Lactate transport and signaling in the brain: potential therapeutic targets and roles in body-brain interaction. J. Cereb. Blood Flow. Metab. V. 35. P. 176. https://doi.org/10.1038/jcbfm.2014.206
  9. Berthet C., Castillo X., Magistretti P.J., Hirt L. 2012. New evidence of neuroprotection by lactate after transient focal cerebral ischaemia: extended benefit after intracerebroventricular injection and efficacy of intravenous administration. Cerebrovasc Dis. V. 34. P. 329. https://doi.org/10.1159/000343657
  10. Bolaños J.P., Almeida A., Moncada S. 2010. Glycolysis: a bioenergetic or a survival pathway? Trends Biochem. Sci. V. 35. P. 145. https://doi.org/10.1016/j.tibs.2009.10.006
  11. Bondi M.W., Edmonds E.C., Salmon D.P. 2017. Alzheimer’s disease: past, present, and future. J. Int. Neuropsychol. Soc. V. 23. P. 818. https://doi.org/10.1017/S135561771700100X
  12. Boury-Jamot B., Carrard A., Martin J.L., Halfon O., Magistretti P.J., Boutrel B. 2016. Disrupting astrocyte-neuron lactate transfer persistently reduces conditioned responses to cocaine. Mol. Psychiatry. V. 21. P. 1070. https://doi.org/10.1038/mp.2015.157
  13. Coco M., Caggia S., Musumeci G., Perciavalle V., Graziano A.C.E., Pannuzzo G., Cardile V. 2013. Sodium L-lactate differently affects brain-derived neurothrophic factor, inducible nitric oxide synthase, and heat shock protein 70 kDa production in human astrocytes and SH-SY5Y cultures. J. Neurosci. Res. V. 91. P. 313. https://doi.org/10.1002/jnr.23154
  14. Correia S.C., Santos R.X., Carvalho C., Cardoso S., Candeias E., Santos M.S., Oliveira C.R., Moreira P.I. 2012. Insulin signaling, glucose metabolism and mitochondria: major players in Alzheimer’s disease and diabetes interrelation. Brain Res. V. 1441. P. 64. https://doi.org/10.1016/j.brainres.2011.12.063
  15. Croteau E., Castellano C.A., Fortier M., Bocti C., Fulop T., Paquet N., Cunnane S.C. 2018. A cross-sectional comparison of brain glucose and ketone metabolism in cognitively healthy older adults, mild cognitive impairment and early Alzheimer’s disease. Exp. Gerontol. V.107. P.18. https://doi.org/10.1016/j.exger.2017.07.004
  16. Cunnane S., Nugent S., Roy M., Courchesne-Loyer A., Croteau E., Tremblay S., Castellano A., Pifferi F., Bocti C., Paquet N., Begdouri H., Bentourkia M., Turcotte E., Allard M., Barberger-Gateau P., Fulop T., Rapoport S.I. 2011. Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition. V. 27. P. 3. https://doi.org/10.1016/j.nut.2010.07.021
  17. Ding F., Yao J., Rettberg J.R., Chen S., Brinton R.D. 2013. Early decline in glucose transport and metabolism precedes shift to ketogenic system in female aging and Alzheimer’s mouse brain: implication for bioenergetic intervention. PLoS One. V. 8. P. e79977. https://doi.org/10.1371/journal.pone.0079977
  18. Encinas J.M., Enikolopov G. 2008. Identifying and quantitating neural stem and progenitor cells in the adult brain. Methods Cell Biol. V. 85. P. 243. https://doi.org/10.1016/S0091-679X(08)85011-X
  19. Epelbaum S., Youssef I., Lacor P.N., Chaurand P., Duplus E., Brugg B., Duyckaerts C., Delatour B. 2015. Acute amnestic encephalopathy in amyloid-β oligomer-injected mice is due to their widespread diffusion in vivo. Neurobiol Aging. V. 36. P. 2043. https://doi.org/10.1016/j.neurobiolaging.2015.03.005
  20. Falkowska A., Gutowska I., Goschorska M., Nowacki P., Chlubek D., Baranowska-Bosiacka I. 2015. Energy metabolism of the brain, including the cooperation between astrocytes and neurons, especially in the context of glycogen metabolism. Int. J. Mol. Sci. V. 16. P. 25959. https://doi.org/10.3390/ijms161125939
  21. Forlenza O.V., Diniz B.S., Gattaz W.F. 2010. Diagnosis and biomarkers of predementia in Alzheimer’s disease. BMC Med. V. 8. P. 89. https://doi.org/10.1186/1741-7015-8-89
  22. Gordon G.R., Howarth C., MacVicar B.A. 2016. Bidirectional control of blood flow by astrocytes: a role for tissue oxygen and other metabolic factors. Adv. Exp. Med. Biol. V. 903. P. 209. https://doi.org/10.1007/978-1-4899-7678-9_15
  23. Harris J.J., Attwell D. 2012. The energetics of CNS white matter. J. Neurosci. V. 32. P. 356. https://doi.org/10.1523/JNEUROSCI.3430-11.2012
  24. Harris R.A. 2017. Cerebral lactate metabolism and memory: Implications for Alzheimer’s disease. Electronic Thesis and Dissertation Repository. P. 4529.
  25. Hashimoto T., Hussien R., Oommen S., Gohil K., Brooks G.A. 2007. Lactate sensitive transcription factor network in L6 cells: activation of MCT1 and mitochondrial biogenesis. FASEB J. V. 21. P. 2602. https://doi.org/10.1096/fj.07-8174com
  26. Hui S., Ghergurovich J.M., Morscher R.J., Jang C., Teng X., Lu W., Esparza L.A., Reya T., Le Z., Yanxiang Guo J., White E., Rabinowitz J.D. 2017. Glucose feeds the TCA cycle via circulating lactateNature. V. 551. P. 115. https://doi.org/10.1038/nature24057
  27. Jin N., Qian W., Yin X., Zhang L., Iqbal K., Grundke-Iqbal I., Gong C.-X., Liu F. 2013. CREB regulates the expression of neuronal glucose transporter 3: a possible mechanism related to impaired brain glucose uptake in Alzheimer’s disease. Nucleic Acids Res. V. 41. P. 3240. https://doi.org/10.1093/nar/gks1227
  28. Koenig M.K. 2008. Presentation and diagnosis of mitochondrial disorders in children. Pediatr. Neurol. V. 38. P. 305. https://doi.org/10.1016/j.pediatrneurol
  29. Liguori C., Chiaravalloti A., Sancesario G., Stefani A., Sancesario G.M., Mercuri N.B., Schillaci O., Pierantozzi M. 2016. Cerebrospinal fluid lactate levels and brain [18F]FDG PET hypometabolism within the default mode network in Alzheimer’s disease. Eur. J. Nucl. Med. Mol. Imaging. V. 43. P. 2040. https://doi.org/10.1007/s00259-016-3417-2
  30. Liguori C., Stefani A., Sancesario G., Sancesario G.M., Marciani M.G., Pierantozzi M. 2015. CSF lactate levels, τ proteins, cognitive decline: a dynamic relationship in Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry. V. 86. P. 655. https://doi.org/10.1136/jnnp-2014-308577
  31. Liu Y., Xue Q., Tang Q., Hou M., Qi H., Chen G., Chen W., Zhang J., Chen Y., Xu X. 2013. A simple method for isolating and culturing the rat brain microvascular endothelial cells. Microvasc. Res. V. 90. P. 199. https://doi.org/10.1016/j.mvr.2013.08.004
  32. Lu W., Huang J., Sun S., Huang S., Gan S., Xu J., Yang M., Xu S., Jiang X. 2015. Changes in lactate content and monocarboxylate transporter 2 expression in Aβ25−35-treated rat model of Alzheimer’s disease. Neurol Sci. V. 36. P. 871. https://doi.org/10.1007/s10072-015-2087-3
  33. Mohamed A., Posse de Chaves E. 2011. Aβ internalization by neurons and glia. Int. J. Alzheimers Dis. P. 127984. https://doi.org/10.4061/2011/127984
  34. Moreira T.J., Pierre K., Maekawa F., Repond C., Cebere A., Liljequist S., Pellerin L. 2009. Enhanced cerebral expression of MCT1 and MCT2 in a rat ischemia model occurs in activated microglial cells. J. Cereb. Blood Flow Metab. V. 29. P. 1273. https://doi.org/10.1038/jcbfm.2009.50
  35. Mosconi L., Mistur R., Switalski R., Tsui W.H., Glodzik L., Li Y., Pirraglia E., De Santi S., Reisberg B., Wisniewski T., de Leon M. 2009. FDG-PET changes in brain glucose metabolism from normal cognition to pathologically verified Alzheimer’s disease. J. Eur. J. Nucl. Med. Mol. Imaging. V. 36. P. 811. https://doi.org/10.1007/s00259-008-1039-z
  36. Muraleedharan R., Gawali M.V., Tiwari D., Sukumaran A., Oatman N., Anderson J., Nardini D., Bhuiyan M.A.N., Tkáč I., Ward A.L., Kundu M., Waclaw R., Chow L.M., Gross C., Rao R., Schirmeier S., Dasgupta B. 2020. AMPK-Regulated Astrocytic lactate shuttle plays a non-cell-autonomous role in neuronal survival. Cell Rep. V. 32. P. 108092. https://doi.org/10.1016/j.celrep.2020.108092
  37. Nagase M., Takahashi Y., Watabe A.M., Kubo Y., Kato F. 2014. On-site energy supply at synapses through monocarboxylate transporters maintains excitatory synaptic transmission. J. Neurosci. V. 34. P. 2605. https://doi.org/10.1523/JNEUROSCI.4687-12.2014
  38. Newman L.A., Korol D.L., Gold P.E. 2011. Lactate produced by glycogenolysis in astrocytes regulates memory processing. PLoS One. V. 6. P. e28427. https://doi.org/0.1371/journal.pone.0028427
  39. Nielsen H.M., Veerhuis R., Holmqvist B., Janciauskiene S. 2009. Binding and uptake of A beta1-42 by primary human astrocytes in vitro. Glia. V. 57. P. 978. https://doi.org/10.1002/glia.20822
  40. Pérez-Escuredo J., Van Hée V.F., Sboarina M., Falces J., Payen V.L., Pellerin L., Sonveaux P. 2016. Monocarboxylate transporters in the brain and in cancer Biochim. Biophys. Acta. V. 1863. P. 2481. https://doi.org/10.1016/j.bbamcr.2016.03.013
  41. Pierre K., Pellerin L. 2005. Monocarboxylate transporters in the central nervous system: distribution, regulation and function. J. Neurochem. V. 94. P. 1. https://doi.org/10.1111/j.1471-4159.2005.03168.x
  42. Pinheiro C., Longatto-Filho A., Azevedo-Silva J., Casal M., Schmitt F.C., Baltazar F. 2012. Role of monocarboxylate transporters in human cancers: state of the art. J. Bioenerg. Biomembr. V. 44. P. 127–139. https://doi.org/10.1007/s10863-012-9428-1
  43. Salmina A.B., Kuvacheva N.V., Morgun A.V., Komleva Y.K., Pozhilenkova E.A., Lopatina O.L., Gorina Y.V., Taranushenko T.E., Petrova L.L. 2015. Glycolysis-mediated control of blood-brain barrier development and function. Int. J. Biochem. Cell Biol. V. 64. P. 174. https://doi.org/10.1016/j.biocel.2015.04.005
  44. Shin B.K., Kang S., Kim D.S., Park S. 2018. Intermittent fasting protects against the deterioration of cognitive function, energy metabolism and dyslipidemia in Alzheimer’s disease-induced estrogen deficient rats. Exp. Biol. Med. (Maywood). V. 243. P. 334. https://doi.org/10.1177/1535370217751610
  45. Shin Y., Choi S.H., Kim E., Bylykbashi E., Kim J.A., Chung S., Kim D.Y., Kamm R.D., Tanzi R.E. 2019. Blood-brain barrier dysfunction in a 3D in vitro model of Alzheimer’s disease. Adv. Sci. V. 6. P.1900962. https://doi.org/10.1002/advs.201900962
  46. Sipos E., Kurunczi A., Kasza A., Horvath J., Felszeghy K., Laroche S., Toldi J., Parducz A., Penke B., Penke Z. 2007. Beta-amyloid pathology in the entorhinal cortex of rats induces memory deficits: implications for Alzheimer’s disease. Neurosci. V. 147. P. 28. https://doi.org/10.1016/j.neuroscience.2007.04.011
  47. Suzuki A., Stern S.A., Bozdagi O., Huntley G.W., Walker R.H., Magistretti P.J., Alberini C.M. 2011. Astrocyte-neuron lactate transport is required for long-term memory formation. Cell. V. 144. P. 810. https://doi.org/10.1016/j.cell.2011.02.018
  48. Tadi M., Allaman I., Lengacher S., Grenningloh G., Magistretti P.J. 2015. Learning-induced gene expression in the hippocampus reveals a role of neuron-astrocyte metabolic coupling in long term memory. PLoS One. 2015. V. 10. P. e0141568. https://doi.org/10.1371/journal.pone.0141568
  49. Tang B.L. 2018. Brain activity-induced neuronal glucose uptake/glycolysis: is the lactate shuttle not required? Brain Res. Bull. V. 137. P. 225. https://doi.org/10.1016/j.brainresbull.2017.12.010
  50. Wang Y., Shang Y., Mishra A., Bacon E., Yin F., Brinton R. 2020. Midlife chronological and endocrinological transitions in brain metabolism: system biology basis for increased Alzheimer’s risk in female brain. Sci. Rep. V. 10. P. 8528. https://doi.org/10.1038/s41598-020-65402-5
  51. Yamanishi S., Katsumura K., Kobayashi T., Puro D.G. 2006. Extracellular lactate as a dynamic vasoactive signal in the rat retinal microvasculature. Am. J. Physiol. Heart Circ. Physiol. V. 290. P. 925H. https://doi.org/10.1152/ajpheart.01012.2005
  52. Zhang M., Cheng X., Dang R., Zhang W., Zhang J., Yao Z. 2018. Lactate deficit in an Alzheimer disease mouse model: the relationship with neuronal damage. J. Neuropathol. Exp. Neurol. V. 77. P. 1163. https://doi.org/10.1093/jnen/nly102

Supplementary files


Copyright (c) 2023 Я.В. Горина, Е.В. Харитонова, Е.Д. Хилажева, А.А. Семенова, А.В. Моргун, Ю.К. Комлева, О.Л. Лопатина, А.Б. Салмина

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies