open access

Vol 81, No 3 (2022)
Original article
Submitted: 2021-04-27
Accepted: 2021-07-17
Published online: 2021-08-03
Get Citation

Preventive effects of bone marrow-derived mesenchymal stem cell transplantation in a D-galactose-induced brain aging in rats

G. El-Akabawy123, K. Aabed4, L. A. Rashed5, S. N. Amin67, I. AlSaati2, M. Al-Fayez8
·
Pubmed: 34355788
·
Folia Morphol 2022;81(3):632-649.
Affiliations
  1. Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman, United Arab Emirates
  2. Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
  3. Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
  4. Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
  5. Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
  6. Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
  7. Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqaa, Jordan
  8. Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia

open access

Vol 81, No 3 (2022)
ORIGINAL ARTICLES
Submitted: 2021-04-27
Accepted: 2021-07-17
Published online: 2021-08-03

Abstract

Background: Aging is a complex process accompanied by numerous morphological, functional, and metabolic impairments in the brain, and a critical risk factor involved in the increasing incidence of neurodegenerative diseases. Few studies have evaluated the efficacy of different sources of mesenchymal stem cells (MSCs) in ameliorating the early morphological and functional alterations in the aging brain. This study, for the first time, evaluated the potential efficacy of intravenous injection of bone marrow-derived mesenchymal stem cells (BMMSCs) in a D-galactose-induced rat model of brain aging.
Materials and methods: BMMSCs (1 × 106) were intravenously injected into brain aging model rats once every 2 weeks for 8 weeks.
Results: The transplanted cells survived and migrated to the brain, and differentiated into astrocytes and neurons, including choline acetyltransferase neurons. BMMSC transplantation improved locomotor activity and cognitive functions, restored cholinergic system function, protected atrophic cholinergic neurons in the basal forebrain, induced antioxidative effects and restored neurotrophic factors, and modulated hippocampal synaptic plasticity by upregulating PSD95 and Egr1 expression.
Conclusions: Our findings demonstrated the efficacy of BMMSC injection in an aging rat model and suggest that these cells may be developed into an effective cell therapy for the aging brain.

Abstract

Background: Aging is a complex process accompanied by numerous morphological, functional, and metabolic impairments in the brain, and a critical risk factor involved in the increasing incidence of neurodegenerative diseases. Few studies have evaluated the efficacy of different sources of mesenchymal stem cells (MSCs) in ameliorating the early morphological and functional alterations in the aging brain. This study, for the first time, evaluated the potential efficacy of intravenous injection of bone marrow-derived mesenchymal stem cells (BMMSCs) in a D-galactose-induced rat model of brain aging.
Materials and methods: BMMSCs (1 × 106) were intravenously injected into brain aging model rats once every 2 weeks for 8 weeks.
Results: The transplanted cells survived and migrated to the brain, and differentiated into astrocytes and neurons, including choline acetyltransferase neurons. BMMSC transplantation improved locomotor activity and cognitive functions, restored cholinergic system function, protected atrophic cholinergic neurons in the basal forebrain, induced antioxidative effects and restored neurotrophic factors, and modulated hippocampal synaptic plasticity by upregulating PSD95 and Egr1 expression.
Conclusions: Our findings demonstrated the efficacy of BMMSC injection in an aging rat model and suggest that these cells may be developed into an effective cell therapy for the aging brain.

Get Citation

Keywords

bone marrow-mesenchymal stem cells, D-galactose, rat, brain

About this article
Title

Preventive effects of bone marrow-derived mesenchymal stem cell transplantation in a D-galactose-induced brain aging in rats

Journal

Folia Morphologica

Issue

Vol 81, No 3 (2022)

Article type

Original article

Pages

632-649

Published online

2021-08-03

Page views

5075

Article views/downloads

1361

DOI

10.5603/FM.a2021.0073

Pubmed

34355788

Bibliographic record

Folia Morphol 2022;81(3):632-649.

Keywords

bone marrow-mesenchymal stem cells
D-galactose
rat
brain

Authors

G. El-Akabawy
K. Aabed
L. A. Rashed
S. N. Amin
I. AlSaati
M. Al-Fayez

References (95)
  1. Abdullah RH, Yaseen NY, Salih SM, et al. Induction of mice adult bone marrow mesenchymal stem cells into functional motor neuron-like cells. J Chem Neuroanat. 2016; 77: 129–142.
  2. Albin RL, Bohnen NI, Muller ML, et al. Regional vesicular acetylcholine transporter distribution in human brain: A [18 F]fluoroethoxybenzovesamicol positron emission tomography study. J Comp Neurol. 2018; 526(17): 2884–2897.
  3. Artegiani B, Calegari F. Age-related cognitive decline: can neural stem cells help us? Aging (Albany NY). 2012; 4(3): 176–186.
  4. Badyra B, Sułkowski M, Milczarek O, et al. Mesenchymal stem cells as a multimodal treatment for nervous system diseases. Stem Cells Transl Med. 2020; 9(10): 1174–1189.
  5. Bae KS, Park JB, Kim HS, et al. Neuron-like differentiation of bone marrow-derived mesenchymal stem cells. Yonsei Med J. 2011; 52(3): 401–412.
  6. Bai WF, Zhang Y, Xu W, et al. Isolation and characterization of neural progenitor cells from bone marrow in cell replacement therapy of brain injury. Front Cell Neurosci. 2020; 14: 49.
  7. Bali P, Lahiri DK, Banik A, et al. Potential for stem cells therapy in Alzheimer's disease: do neurotrophic factors play critical role? Curr Alzheimer Res. 2017; 14(2): 208–220.
  8. Barrientos RM, Kitt MM, Watkins LR, et al. Neuroinflammation in the normal aging hippocampus. Neuroscience. 2015; 309: 84–99.
  9. Bekdash RA. The cholinergic system, the adrenergic system and the neuropathology of Azheimer's disease. Int J Mol Sci. 2021; 22(3).
  10. Berebichez-Fridman R, Montero-Olvera PR. Sources and clinical applications of mesenchymal stem cells: state-of-the-art review. Sultan Qaboos Univ Med J. 2018; 18(3): e264–e277.
  11. Bishop NA, Lu T, Yankner BA. Neural mechanisms of ageing and cognitive decline. Nature. 2010; 464(7288): 529–535.
  12. Borkowska P, Fila-Danilow A, Paul-Samojedny M, et al. Differentiation of adult rat mesenchymal stem cells to GABAergic, dopaminergic and cholinergic neurons. Pharmacol Rep. 2015; 67(2): 179–186.
  13. Bortolotti F, Ukovich L, Razban V, et al. In vivo therapeutic potential of mesenchymal stromal cells depends on the source and the isolation procedure. Stem Cell Reports. 2015; 4(3): 332–339.
  14. Boskovic Z, Meier S, Wang Y, et al. Regulation of cholinergic basal forebrain development, connectivity, and function by neurotrophin receptors. Neuronal Signal. 2019; 3(1): NS20180066.
  15. Budni J, Bellettini-Santos T, Mina F, et al. The involvement of BDNF, NGF and GDNF in aging and Alzheimer's disease. Aging Dis. 2015; 6(5): 331–341.
  16. Bustos FJ, Ampuero E, Jury N, et al. Epigenetic editing of the Dlg4/PSD95 gene improves cognition in aged and Alzheimer's disease mice. Brain. 2017; 140(12): 3252–3268.
  17. Cao N, Liao T, Liu J, et al. Clinical-grade human umbilical cord-derived mesenchymal stem cells reverse cognitive aging via improving synaptic plasticity and endogenous neurogenesis. Cell Death Dis. 2017; 8(8): e2996.
  18. Casu MA, Wong TP, De Koninck Y, et al. Aging causes a preferential loss of cholinergic innervation of characterized neocortical pyramidal neurons. Cereb Cortex. 2002; 12(3): 329–337.
  19. Daniele S, Giacomelli C, Martini C. Brain ageing and neurodegenerative disease: The role of cellular waste management. Biochem Pharmacol. 2018; 158: 207–216.
  20. Daulatzai MA. Early stages of pathogenesis in memory impairment during normal senescence and Alzheimer's disease. J Alzheimers Dis. 2010; 20(2): 355–367.
  21. Duclot F, Kabbaj M. The role of early growth response 1 (EGR1) in brain plasticity and neuropsychiatric disorders. Front Behav Neurosci. 2017; 11: 35.
  22. Ellman G. Tissue sulfhydryl groups. Arch Biochem Biophys. 1959; 82(1): 70–77.
  23. Erickson KI, Prakash RS, Voss MW, et al. Brain-derived neurotrophic factor is associated with age-related decline in hippocampal volume. J Neurosci. 2010; 30(15): 5368–5375.
  24. Fatemi I, Khaluoi A, Kaeidi A, et al. Protective effect of metformin on D-galactose-induced aging model in mice. Iran J Basic Med Sci. 2018; 21(1): 19–25.
  25. Ge M, Zhang Y, Hao Q, et al. Effects of mesenchymal stem cells transplantation on cognitive deficits in animal models of Alzheimer's disease: a systematic review and meta-analysis. Brain Behav. 2018; 8(7): e00982.
  26. Gemma C, Vila J, Bachstetter A, Bickford PC. Oxidative stress and the aging brain: from theory to prevention. In: riddle DR (ed.). Brain aging: models, methods, and mechanisms. Frontiers in neuroscience. Boca Raton, FL 2007.
  27. Grosse G, Djalali S, Deng DR, et al. Area-specific effects of brain-derived neurotrophic factor (BDNF) genetic ablation on various neuronal subtypes of the mouse brain. Brain Res Dev Brain Res. 2005; 156(2): 111–126.
  28. Grothe M, Heinsen H, Teipel SJ. Atrophy of the cholinergic Basal forebrain over the adult age range and in early stages of Alzheimer's disease. Biol Psychiatry. 2012; 71(9): 805–813.
  29. Hajjar I, Hayek SS, Goldstein FC, et al. Oxidative stress predicts cognitive decline with aging in healthy adults: an observational study. J Neuroinflammation. 2018; 15(1): 17.
  30. Hampel H, Mesulam MM, Cuello AC, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain. 2018; 141(7): 1917–1933.
  31. Harada CN, Natelson Love MC, Triebel KL. Normal cognitive aging. Clin Geriatr Med. 2013; 29(4): 737–752.
  32. Hernandez CM, Gearhart DA, Parikh V, et al. Comparison of galantamine and donepezil for effects on nerve growth factor, cholinergic markers, and memory performance in aged rats. J Pharmacol Exp Ther. 2006; 316(2): 679–694.
  33. Hernández R, Jiménez-Luna C, Perales-Adán J, et al. Differentiation of human mesenchymal stem cells towards neuronal lineage: clinical trials in nervous system disorders. Biomol Ther (Seoul). 2020; 28(1): 34–44.
  34. Hou Y, Dan X, Babbar M, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019; 15(10): 565–581.
  35. Hu S, Li CSR. Age-Related structural and functional changes of the hippocampus and the relationship with inhibitory control. Brain Sci. 2020; 10(12).
  36. Hunsberger J, Rao M, Kurtzberg J, et al. Accelerating stem cell trials for Alzheimer's disease. Lancet Neurol. 2016; 15(2): 219–230.
  37. Ibrahim WW, Abdelkader NF, Ismail HM, et al. Escitalopram ameliorates cognitive impairment in D-galactose-injected ovariectomized rats: modulation of JNK, GSK-3beta, and ERK signalling pathways. Sci Rep. 2019; 9(1): 10056.
  38. Isaev NK, Stelmashook EV, Genrikhs EE. Neurogenesis and brain aging. Rev Neurosci. 2019; 30(6): 573–580.
  39. Kim D, Kyung J, Park D, et al. Health span-extending activity of human amniotic membrane- and adipose tissue-derived stem cells in F344 rats. Stem Cells Transl Med. 2015; 4(10): 1144–1154.
  40. Kim Y, Kim H, Cho H, et al. Direct comparison of human mesenchymal stem cells derived from adipose tissues and bone marrow in mediating neovascularization in response to vascular ischemia. Cell Physiol Biochem. 2007; 20(6): 867–876.
  41. Lee HJu, Lee JK, Lee H, et al. Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer's disease mouse model through modulation of neuroinflammation. Neurobiol Aging. 2012; 33(3): 588–602.
  42. Li Z, Zhang Z, Ren Y, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 2021; 22(2): 165–187.
  43. Limke TL, Rao MS. Neural stem cells in aging and disease. J Cell Mol Med. 2002; 6(4): 475–496.
  44. Liu X, Wu C, Han D, et al. Partially hydrolyzed guar gum attenuates D-galactose-Induced oxidative stress and restores gut microbiota in rats. Int J Mol Sci. 2019; 20(19).
  45. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001; 25(4): 402–408.
  46. Lopez-Leon M, Reggiani PC, Claudia B, et al. Regenerative medicine for the aging brain. Enliven J Stem Cell Res Regen Med. 2014; 01(01).
  47. Lushchak VI. Interplay between bioenergetics and oxidative stress at normal brain aging. Aging as a result of increasing disbalance in the system oxidative stress-energy provision. Pflugers Arch. 2021; 473(5): 713–722.
  48. Ma S, Zang T, Liu ML, et al. Aging-relevant human basal forebrain cholinergic neurons as a cell model for Alzheimer's disease. Mol Neurodegener. 2020; 15(1): 61.
  49. Mesulam M. Cholinergic aspects of aging and Alzheimer's disease. Biol Psychiatry. 2012; 71(9): 760–761.
  50. Miranda M, Morici JF, Zanoni MB, et al. Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci. 2019; 13: 363.
  51. Molinari C, Morsanuto V, Ruga S, et al. The role of BDNF on aging-modulation markers. Brain Sci. 2020; 10(5).
  52. Morrison JH, Baxter MG. The ageing cortical synapse: hallmarks and implications for cognitive decline. Nat Rev Neurosci. 2012; 13(4): 240–250.
  53. Mota C, Taipa R, das Neves SP, et al. Structural and molecular correlates of cognitive aging in the rat. Sci Rep. 2019; 9(1): 2005.
  54. Naaldijk Y, Jäger C, Fabian C, et al. Effect of systemic transplantation of bone marrow-derived mesenchymal stem cells on neuropathology markers in APP/PS1 Alzheimer mice. Neuropathol Appl Neurobiol. 2017; 43(4): 299–314.
  55. Naghdi M, Tiraihi T, Namin SA, et al. Transdifferentiation of bone marrow stromal cells into cholinergic neuronal phenotype: a potential source for cell therapy in spinal cord injury. Cytotherapy. 2009; 11(2): 137–152.
  56. Nakano M, Kubota K, Kobayashi E, et al. Bone marrow-derived mesenchymal stem cells improve cognitive impairment in an Alzheimer's disease model by increasing the expression of microRNA-146a in hippocampus. Sci Rep. 2020; 10(1): 10772.
  57. Nam SM, Seo M, Seo JS, et al. Ascorbic acid mitigates D-galactose-induced brain aging by increasing hippocampal neurogenesis and improving memory function. Nutrients. 2019; 11(1).
  58. Nasiri E, Alizadeh A, Roushandeh AM, et al. Melatonin-pretreated adipose-derived mesenchymal stem cells efficeintly improved learning, memory, and cognition in an animal model of Alzheimer's disease. Metab Brain Dis. 2019; 34(4): 1131–1143.
  59. Neves AF, Camargo C, Premer C, et al. Intravenous administration of mesenchymal stem cells reduces Tau phosphorylation and inflammation in the 3xTg-AD mouse model of Alzheimer's disease. Exp Neurol. 2021; 341: 113706.
  60. Oh SeH, Kim HaNa, Park HJ, et al. Mesenchymal stem cells increase hippocampal neurogenesis and neuronal differentiation by enhancing the wnt signaling pathway in an Alzheimer's disease model. Cell Transplant. 2015; 24(6): 1097–1109.
  61. Park D, Yang G, Bae DK, et al. Human adipose tissue-derived mesenchymal stem cells improve cognitive function and physical activity in ageing mice. J Neurosci Res. 2013; 91(5): 660–670.
  62. Penner MR, Parrish RR, Hoang LT, et al. Age-related changes in Egr1 transcription and DNA methylation within the hippocampus. Hippocampus. 2016; 26(8): 1008–1020.
  63. Peters R. Ageing and the brain. Postgrad Med J. 2006; 82(964): 84–88.
  64. Piccardi L, Curcio G, Palermo L, et al. Ageing and neurodegenerative disorders. Behav Neurol. 2015; 2015: 149532.
  65. Polisetti N, Chaitanya VG, Babu PP, et al. Isolation, characterization and differentiation potential of rat bone marrow stromal cells. Neurol India. 2010; 58(2): 201–208.
  66. Popescu BO, Toescu EC, Popescu LM, et al. Blood-brain barrier alterations in ageing and dementia. J Neurol Sci. 2009; 283(1-2): 99–106.
  67. Qi Y, Zhang F, Song Ge, et al. Cholinergic neuronal differentiation of bone marrow mesenchymal stem cells in rhesus monkeys. Sci China Life Sci. 2010; 53(5): 573–580.
  68. Qin C, Lu Y, Wang K, et al. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer's disease: a meta-analytic review on potential mechanisms. Transl Neurodegener. 2020; 9(1): 20.
  69. Ramalho BD, Almeida FM, Sales CM, et al. Injection of bone marrow mesenchymal stem cells by intravenous or intraperitoneal routes is a viable alternative to spinal cord injury treatment in mice. Neural Regen Res. 2018; 13(6): 1046–1053.
  70. Rangan GK, Tesch GH. Quantification of renal pathology by image analysis. Nephrology (Carlton). 2007; 12(6): 553–558.
  71. Reeve A, Simcox E, Turnbull D. Ageing and Parkinson's disease: why is advancing age the biggest risk factor? Ageing Res Rev. 2014; 14: 19–30.
  72. Schliebs R, Arendt T. The cholinergic system in aging and neuronal degeneration. Behav Brain Res. 2011; 221(2): 555–563.
  73. Shekari A, Fahnestock M. Retrograde axonal transport of BDNF and proNGF diminishes with age in basal forebrain cholinergic neurons. Neurobiol Aging. 2019; 84: 131–140.
  74. Shen J, Tsai YT, Dimarco NM, et al. Transplantation of mesenchymal stem cells from young donors delays aging in mice. Sci Rep. 2011; 1: 67.
  75. Sikora E, Bielak-Zmijewska A, Dudkowska M, et al. Cellular senescence in brain aging. Front Aging Neurosci. 2021; 13: 646924.
  76. Simpson JE, Wharton SB, Cooper J, et al. Alterations of the blood-brain barrier in cerebral white matter lesions in the ageing brain. Neurosci Lett. 2010; 486(3): 246–251.
  77. Stepanichev M, Nedogreeva O, Gulyaeva N. Cholinergic degeneration in early stages of Alzheimer’s disease: loss of cholinergic phenotype or loss of cells? Alzheimers Dement Cogn Neurol. 2017; 1(2).
  78. Sun C, Shao J, Su Le, et al. Cholinergic neuron-like cells derived from bone marrow stromal cells induced by tricyclodecane-9-yl-xanthogenate promote functional recovery and neural protection after spinal cord injury. Cell Transplant. 2013; 22(6): 961–975.
  79. Tapia-Arancibia L, Aliaga E, Silhol M, et al. New insights into brain BDNF function in normal aging and Alzheimer disease. Brain Res Rev. 2008; 59(1): 201–220.
  80. Tropel P, Platet N, Platel JC, et al. Functional neuronal differentiation of bone marrow-derived mesenchymal stem cells. Stem Cells. 2006; 24(12): 2868–2876.
  81. Utkin YN. Aging affects nicotinic acetylcholine receptors in brain. Cent Nerv Syst Agents Med Chem. 2019; 19(2): 119–124.
  82. Vallianatou T, Shariatgorji M, Nilsson A, et al. Molecular imaging identifies age-related attenuation of acetylcholine in retrosplenial cortex in response to acetylcholinesterase inhibition. Neuropsychopharmacology. 2019; 44(12): 2091–2098.
  83. Voigt RM, Raeisi S, Yang J, et al. Systemic brain derived neurotrophic factor but not intestinal barrier integrity is associated with cognitive decline and incident Alzheimer's disease. PLoS One. 2021; 16(3): e0240342.
  84. Volkman R, Offen D. Concise review: mesenchymal stem cells in neurodegenerative diseases. Stem Cells. 2017; 35(8): 1867–1880.
  85. Wang X, Ma S, Yang Bo, et al. Resveratrol promotes hUC-MSCs engraftment and neural repair in a mouse model of Alzheimer's disease. Behav Brain Res. 2018; 339: 297–304.
  86. Wilkins A, Kemp K, Ginty M, et al. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res. 2009; 3(1): 63–70.
  87. Wills ED. Evaluation of lipid peroxidation in lipids and biologicalmembranes. In: Snell K., Mullock B. (eds.), Biochemical toxicology: a practicalapproach. Oxford, London 1987.
  88. Wu QY, Li J, Feng ZT, et al. Bone marrow stromal cells of transgenic mice can improve the cognitive ability of an Alzheimer's disease rat model. Neurosci Lett. 2007; 417(3): 281–285.
  89. Yi TG, Cho YK, Lee HJ, et al. A novel immunomodulatory mechanism dependent on acetylcholine secreted by human bone marrow-derived mesenchymal stem cells. Int J Stem Cells. 2019; 12(2): 315–330.
  90. Yun HM, Kim HS, Park KR, et al. Placenta-derived mesenchymal stem cells improve memory dysfunction in an Aβ1-42-infused mouse model of Alzheimer's disease. Cell Death Dis. 2013; 4: e958.
  91. Zappa Villar MF, Lehmann M, García MG, et al. Mesenchymal stem cell therapy improves spatial memory and hippocampal structure in aging rats. Behav Brain Res. 2019; 374: 111887.
  92. Zhang XM, Ouyang YJ, Yu BQ, et al. Therapeutic potential of dental pulp stem cell transplantation in a rat model of Alzheimer's disease. Neural Regen Res. 2021; 16(5): 893–898.
  93. Zhou G, Xiong W, Zhang X, et al. Retrieval of consolidated spatial memory in the water maze is correlated with expression of pCREB and egr1 in the hippocampus of aged mice. Dement Geriatr Cogn Dis Extra. 2013; 3(1): 39–47.
  94. Zia A, Pourbagher-Shahri AM, Farkhondeh T, et al. Molecular and cellular pathways contributing to brain aging. Behav Brain Funct. 2021; 17(1): 6.
  95. Zilka N, Zilkova M, Kazmerova Z, et al. Mesenchymal stem cells rescue the Alzheimer's disease cell model from cell death induced by misfolded truncated tau. Neuroscience. 2011; 193: 330–337.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Grupa Via Medica, Świętokrzyska 73, 80–180 Gdańsk, Poland

tel.: +48 58 320 94 94, faks: +48 58 320 94 60, e-mail: viamedica@viamedica.pl