Publication Cover
Experimental Aging Research
An International Journal Devoted to the Scientific Study of the Aging Process
Volume 46, 2020 - Issue 2
266
Views
2
CrossRef citations to date
0
Altmetric
Articles

Platelet-Rich Plasma (PRP) is a Potential Self-Sourced Cognition Booster in Elderly Mice

ORCID Icon & ORCID Icon
Pages 139-153 | Received 12 Jun 2019, Accepted 10 Dec 2019, Published online: 15 Jan 2020

References

  • Alcaraz, J., Oliver, A., & Sánchez, J. M. (2015a). Platelet-Rich Plasma in a patient with cerebral palsy. The American Journal of Case Reports, 16, 469–472. doi:10.12659/AJCR.893805
  • Alcaraz, J., Oliver, A., Sánchez, J. M. J. M., & Lajara, J. J. (2015b). Clinical use of platelet-rich plasma: A new dimension in regenerative medicine. Medical Science Review, 2, 111–120. doi:10.12659/MSRev.895455
  • Almutairi, M. M. A., Gong, C., Xu, Y. G., Chang, Y., & Shi, H. (2016). Factors controlling permeability of the blood–brain barrier. Cellular and Molecular Life Sciences, 73(1), 57–77. doi:10.1007/s00018-015-2050-8
  • Anitua, E., Pascual, C., Antequera, D., Bolos, M., Padilla, S., Orive, G., & Carro, E. (2014). Plasma rich in growth factors (PRGF-Endoret) reduces neuropathologic hallmarks and improves cognitive functions in an Alzheimer’s disease mouse model. Neurobiology of Aging, 35(7), 1582–1595. doi:10.1016/J.NEUROBIOLAGING.2014.01.009
  • Anitua, E., Pascual, C., Pérez-Gonzalez, R., Antequera, D., Padilla, S., Orive, G., & Carro, E. (2013). Intranasal delivery of plasma and platelet growth factors using PRGF-endoret system enhances neurogenesis in a mouse model of Alzheimer’s disease. PloS One, 8(9), e73118. doi:10.1371/journal.pone.0073118
  • Anitua, E., Pascual, C., Pérez-Gonzalez, R., Orive, G., & Carro, E. (2015). Intranasal PRGF-Endoret enhances neuronal survival and attenuates NF-κB-dependent inflammation process in a mouse model of Parkinson’s disease. Journal of Controlled Release, 203, 170–180. doi:10.1016/J.JCONREL.2015.02.030
  • Anitua, E., Pino, A., & Orive, G. (2017). Opening new horizons in regenerative dermatology using platelet-based autologous therapies. International Journal of Dermatology, 56(3), 247–251. doi:10.1111/ijd.13510
  • Anitua, E., Sánchez, M., Orive, G., & Andía, I. (2007). The potential impact of the preparation rich in growth factors (PRGF) in different medical fields. Biomaterials, 28(31), 4551–4560. doi:10.1016/J.BIOMATERIALS.2007.06.037
  • Balasubramanian, P., & Longo, V. D. (2016). Growth factors, aging and age-related diseases. Growth Hormone & IGF Research : Official Journal of the Growth Hormone Research Society and the International IGF Research Society, 28, 66–68. doi:10.1016/j.ghir.2016.01.001
  • Barton, S. M., Janve, V. A., McClure, R., Anderson, A., Matsubara, J. A., Gore, J. C., & Pham, W. (2019). Lipopolysaccharide induced opening of the blood brain barrier on aging 5XFAD mouse model. Journal of Alzheimer’s Disease, 67(2), 503–513. doi:10.3233/JAD-180755
  • Bilgic, Y., Demir, E. A., Bilgic, N., Dogan, H., Tutuk, O., & Tumer, C. (2018). Detrimental effects of chia (Salvia hispanica L.) seeds on learning and memory in aluminum chloride‑induced experimental Alzheimer’s disease. Acta Neurobiologiae Experimentalis, 78(4), 322–331. doi:10.21307/ane-2018-031
  • Bimonte, H. A., Nelson, M. E., & Granholm, A.-C. E. (2003). Age-related deficits as working memory load increases: Relationships with growth factors. Neurobiology of Aging, 24(1), 37–48. doi:10.1016/S0197-4580(02)00015-5
  • Bland, S. T., Tamlyn, J. P., Barrientos, R. M., Greenwood, B. N., Watkins, L. R., Campeau, S., … Maier, S. F. (2007). Expression of fibroblast growth factor-2 and brain-derived neurotrophic factor mRNA in the medial prefrontal cortex and hippocampus after uncontrollable or controllable stress. Neuroscience, 144(4), 1219–1228. doi:10.1016/J.NEUROSCIENCE.2006.11.026
  • Bordner, K. A., Kitchen, R. R., Carlyle, B., George, E. D., Mahajan, M. C., Mane, S. M., … Simen, A. A. (2011). Parallel declines in cognition, motivation, and locomotion in aging mice: Association with immune gene upregulation in the medial prefrontal cortex. Experimental Gerontology, 46(8), 643–659. doi:10.1016/j.exger.2011.03.003
  • Bowman, G. L., Dayon, L., Kirkland, R., Wojcik, J., Peyratout, G., Severin, I. C., … Popp, J. (2018). Blood-brain barrier breakdown, neuroinflammation, and cognitive decline in older adults. Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 14(12), 1640–1650. doi:10.1016/j.jalz.2018.06.2857
  • Brooks, S. P., Pask, T., Jones, L., & Dunnett, S. B. (2005). Behavioural profiles of inbred mouse strains used as transgenic backgrounds. II: Cognitive tests. Genes, Brain and Behavior, 4(5), 307–317. doi:10.1111/j.1601-183X.2004.00109.x
  • Burke, S. N., & Barnes, C. A. (2006). Neural plasticity in the ageing brain. Nature Reviews Neuroscience, 7(1), 30–40. doi:10.1038/nrn1809
  • Carro, E., Spuch, C., Trejo, J. L., Antequera, D., & Torres-Aleman, I. (2005). Choroid plexus megalin is involved in neuroprotection by serum insulin-like growth factor I. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 25(47), 10884–10893. doi:10.1523/JNEUROSCI.2909-05.2005
  • Cham, R., Studenski, S. A., Perera, S., & Bohnen, N. I. (2008). Striatal dopaminergic denervation and gait in healthy adults. Experimental Brain Research, 185(3), 391–398. doi:10.1007/s00221-007-1161-3
  • Ciechanowski, P. S., Katon, W. J., & Russo, J. E. (2000). Depression and diabetes: Impact of depressive symptoms on adherence, function, and costs. Archives of Internal Medicine, 160(21), 3278–3285. doi:10.1001/archinte.160.21.3278
  • Cryan, J. F., Mombereau, C., & Vassout, A. (2005). The tail suspension test as a model for assessing antidepressant activity: Review of pharmacological and genetic studies in mice. Neuroscience & Biobehavioral Reviews, 29(4–5), 571–625. doi:10.1016/J.NEUBIOREV.2005.03.009
  • De Geyter, D., De Smedt, A., Stoop, W., De Keyser, J., & Kooijman, R. (2016). Central IGF-I receptors in the brain are instrumental to neuroprotection by systemically injected IGF-I in a rat model for ischemic stroke. CNS Neuroscience and Therapeutics, 22(7), 611–616. doi:10.1111/cns.12550
  • Deak, F., & Sonntag, W. E. (2012). Aging, synaptic dysfunction, and insulin-like growth factor (IGF)-1. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 67A(6), 611–625. doi:10.1093/gerona/gls118
  • Demir, E. A., Ozturk, A., Tutuk, O., Dogan, H., & Tumer, C. (2019). Anticonvulsive and behavior modulating effects of sophoretin and rutoside. Biologia Futura, 70(3), 251–259. doi:10.1556/019.70.2019.29
  • Demir, E. A., Tutuk, O., Dogan, H., & Tumer, C. (2019). Depression in Alzheimer’s disease: The roles of cholinergic and serotonergic systems. In T. Wisniewski (Ed.), Alzheimer’s disease (1st ed. pp. 223–235). Brisbane, AU: Codon Publications. doi:10.15586/alzheimersdisease.2019
  • Dobolyi, A., Vincze, C., Pál, G., & Lovas, G. (2012). The neuroprotective functions of transforming growth factor beta proteins. International Journal of Molecular Sciences, 13(7), 8219–8258. doi:10.3390/ijms13078219
  • Dohan Ehrenfest, D. M., Andia, I., Zumstein, M. A., Zhang, C.-Q., Pinto, N. R., & Bielecki, T. (2014). Classification of platelet concentrates (Platelet-Rich Plasma-PRP, Platelet-Rich Fibrin-PRF) for topical and infiltrative use in orthopedic and sports medicine: Current consensus, clinical implications and perspectives. Muscles, Ligaments and Tendons Journal, 4(1), 3–9. doi:10.11138/mltj/2014.4.1.0013
  • Duong, Q.-V., Kintzing, M. L., Kintzing, W. E., Abdallah, I. M., Brannen, A. D., & Kaddoumi, A. (2019). Plasma Rich in Growth Factors (PRGF) disrupt the blood-brain barrier integrity and elevate amyloid pathology in the brains of 5XFAD mice. International Journal of Molecular Sciences, 20, 6. doi:10.3390/ijms20061489
  • Farrall, A. J., & Wardlaw, J. M. (2009). Blood–brain barrier: Ageing and microvascular disease – Systematic review and meta-analysis. Neurobiology of Aging, 30(3), 337–352. doi:10.1016/J.NEUROBIOLAGING.2007.07.015
  • Franklin, S. P., Birdwhistell, K. E., Strelchik, A., Garner, B. C., & Brainard, B. M. (2017). Influence of cellular composition and exogenous activation on growth factor and cytokine concentrations in canine Platelet-Rich Plasmas. Frontiers in Veterinary Sciences, 4, 40. doi:10.3389/fvets.2017.00040
  • Friard, O., & Gamba, M. (2016). BORIS: A free, versatile open-source event-logging software for video/audio coding and live observations. Methods in Ecology and Evolution, 7(11), 1325–1330. doi:10.1111/2041-210X.12584
  • Gaceb, A., Barbariga, M., Özen, I., & Paul, G. (2018). The pericyte secretome: Potential impact on regeneration. Biochimie, 155, 16–25. doi:10.1016/J.BIOCHI.2018.04.015
  • Gaceb, A., Özen, I., Padel, T., Barbariga, M., & Paul, G. (2018). Pericytes secrete pro-regenerative molecules in response to platelet-derived growth factor-BB. Journal of Cerebral Blood Flow and Metabolism: Official Journal of the International Society of Cerebral Blood Flow and Metabolism, 38(1), 45–57. doi:10.1177/0271678X17719645
  • Gazit, N., Vertkin, I., Shapira, I., Helm, M., Slomowitz, E., Sheiba, M., … Slutsky, I. (2016). IGF-1 receptor differentially regulates spontaneous and evoked transmission via mitochondria at hippocampal synapses. Neuron, 89(3), 583–597. doi:10.1016/j.neuron.2015.12.034
  • Goldstein, C. M., Gathright, E. C., & Garcia, S. (2017). Relationship between depression and medication adherence in cardiovascular disease: The perfect challenge for the integrated care team. Patient Preference and Adherence, 11, 547–559. doi:10.2147/PPA.S127277
  • Hamezah, H. S., Durani, L. W., Ibrahim, N. F., Yanagisawa, D., Kato, T., Shiino, A., … Tooyama, I. (2017). Volumetric changes in the aging rat brain and its impact on cognitive and locomotor functions. Experimental Gerontology, 99, 69–79. doi:10.1016/J.EXGER.2017.09.008
  • Harada, C. N., Natelson Love, M. C., & Triebel, K. L. (2013). Normal cognitive aging. Clinics in Geriatric Medicine, 29(4), 737–752. doi:10.1016/j.cger.2013.07.002
  • Hughes, C. M. (2004). Medication non-adherence in the elderly. Drugs & Aging, 21(12), 793–811. doi:10.2165/00002512-200421120-00004
  • Kniewallner, K. M., Grimm, N., & Humpel, C. (2014). Platelet-derived nerve growth factor supports the survival of cholinergic neurons in organotypic rat brain slices. Neuroscience Letters, 574, 64–69. doi:10.1016/j.neulet.2014.05.033
  • Kumar, A., Bodhinathan, K., & Foster, T. C. (2009). Susceptibility to calcium dysregulation during brain aging. Frontiers in Aging Neuroscience, 1, 2. doi:10.3389/neuro.24.002.2009
  • Lichtenwalner, R. J., Forbes, M. E., Bennett, S. A., Lynch, C. D., Sonntag, W. E., & Riddle, D. R. (2001). Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis. Neuroscience, 107(4), 603–613. doi:10.1016/S0306-4522(01)00378-5
  • McLennan, I. S., Weible, M. W., Hendry, I. A., & Koishi, K. (2005). Transport of transforming growth factor-β2 across the blood–brain barrier. Neuropharmacology, 48(2), 274–282. doi:10.1016/j.neuropharm.2004.10.005
  • Mirelman, A., Maidan, I., Bernad-Elazari, H., Shustack, S., Giladi, N., & Hausdorff, J. M. (2017). Effects of aging on prefrontal brain activation during challenging walking conditions. Brain and Cognition, 115, 41–46. doi:10.1016/J.BANDC.2017.04.002
  • Mohapel, P., Frielingsdorf, H., Häggblad, J., Zachrisson, O., & Brundin, P. (2005). Platelet-Derived Growth Factor (PDGF-BB) and Brain-Derived Neurotrophic Factor (BDNF) induce striatal neurogenesis in adult rats with 6-hydroxydopamine lesions. Neuroscience, 132(3), 767–776. doi:10.1016/j.neuroscience.2004.11.056
  • Montagne, A., Barnes, S. R., Sweeney, M. D., Halliday, M. R., Sagare, A. P., Zhao, Z., … Zlokovic, B. V. (2015). Blood-brain barrier breakdown in the aging human hippocampus. Neuron, 85(2), 296–302. doi:10.1016/J.NEURON.2014.12.032
  • Pan, W., & Kastin, A. J. (1999). Entry of EGF into brain is rapid and saturable. Peptides, 20(9), 1091–1098. doi:10.1016/s0196-9781(99)00094-7
  • Park, R., Kook, S.-Y., Park, J.-C., & Mook-Jung, I. (2014). Aβ1–42 reduces P-glycoprotein in the blood–brain barrier through RAGE–NF-κB signaling. Cell Death & Disease, 5(6), e1299–e1299. doi:10.1038/cddis.2014.258
  • Picillo, M., Pivonello, R., Santangelo, G., Pivonello, C., Savastano, R., Auriemma, R., … Pellecchia, M. T. (2017). Serum IGF-1 is associated with cognitive functions in early, drug-naïve Parkinson’s disease. PLoS One, 12(10), e0186508. doi:10.1371/journal.pone.0186508
  • Rebentish, P. D., Umashetty, G., Kaur, H., Doizode, T., Kaslekar, M., & Chowdhury, S. (2016). Platelet-rich fibrin: A boon in regenerative endodontics. Minerva Stomatologica, 65(6), 385–392.
  • Saify, K. (2016). Efficacy of platelet components and cord blood in the treatment of Vitiligo. In 5th National Conference of Indian Society of Transfusion Medicine, Nov 18-19, 2016. Bhopal, India. Retrieved from http://istm.net.in/transmedcon2016-presentations/23.Efficacy of Platelet Components and Cord Blood.pdf
  • Salthouse, T. A. (2009). When does age-related cognitive decline begin? Neurobiology of Aging, 30(4), 507–514. doi:10.1016/j.neurobiolaging.2008.09.023
  • Sanchez, A., Wadhwani, S., & Grammas, P. (2010). Multiple neurotrophic effects of VEGF on cultured neurons. Neuropeptides, 44(4), 323–331. doi:10.1016/J.NPEP.2010.04.002
  • Sclafani, A. P. (2011). Safety, efficacy, and utility of platelet-rich fibrin matrix in facial plastic surgery. Archives of Facial Plastic Surgery, 13(4), 247. doi:10.1001/archfacial.2011.3
  • Shan, H., Messi, M. L., Zheng, Z., Wang, Z.-M., & Delbono, O. (2003). Preservation of motor neuron Ca2+ channel sensitivity to insulin-like growth factor-1 in brain motor cortex from senescent rat. The Journal of Physiology, 553(Pt 1), 49–63. doi:10.1113/jphysiol.2003.047746
  • Shen, J., Xu, G., Zhu, R., Yuan, J., Ishii, Y., Hamashima, T., … Sasahara, M. (2019). PDGFR-β restores blood-brain barrier functions in a mouse model of focal cerebral ischemia. Journal of Cerebral Blood Flow and Metabolism: Official Journal of the International Society of Cerebral Blood Flow and Metabolism, 39(8), 1501–1515. doi:10.1177/0271678X18769515
  • Shetty, A. K., Hattiangady, B., & Shetty, G. A. (2005). Stem/progenitor cell proliferation factors FGF-2, IGF-1, and VEGF exhibit early decline during the course of aging in the hippocampus: Role of astrocytes. Glia, 51(3), 173–186. doi:10.1002/glia.20187
  • Siddiqui, S., Fang, M., Ni, B., Lu, D., Martin, B., & Maudsley, S. (2012). Central role of the EGF receptor in neurometabolic aging. International Journal of Endocrinology, 2012, 739428. doi:10.1155/2012/739428
  • Sonntag, W. E., Bennett, S. A., Khan, A. S., Thornton, P. L., Xu, X., Ingram, R. L., & Brunso-Bechtold, J. K. (2000). Age and insulin-like growth factor-1 modulate N-methyl-D-aspartate receptor subtype expression in rats. Brain Research Bulletin, 51(4), 331–338. doi:10.1016/S0361-9230(99)00259-2
  • Sonntag, W. E., Ramsey, M., & Carter, C. S. (2005). Growth hormone and insulin-like growth factor-1 (IGF-1) and their influence on cognitive aging. Ageing Research Reviews, 4(2), 195–212. doi:10.1016/j.arr.2005.02.001
  • Stamatovic, S. M., Martinez-Revollar, G., Hu, A., Choi, J., Keep, R. F., & Andjelkovic, A. V. (2019). Decline in Sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiology of Disease, 126, 105–116. doi:10.1016/J.NBD.2018.09.006
  • Suzuki, Y., Nagai, N., & Umemura, K. (2016). A review of the mechanisms of blood-brain barrier permeability by tissue-type plasminogen activator treatment for cerebral ischemia. Frontiers in Cellular Neuroscience, 10(2). doi:10.3389/fncel.2016.00002
  • Tang, Z., Arjunan, P., Lee, C., Li, Y., Kumar, A., Hou, X., … Li, X. (2010). Survival effect of PDGF-CC rescues neurons from apoptosis in both brain and retina by regulating GSK3beta phosphorylation. The Journal of Experimental Medicine, 207(4), 867–880. doi:10.1084/jem.20091704
  • Van Dam, D., Lenders, G., & De Deyn, P. P. (2006). Effect of Morris water maze diameter on visual-spatial learning in different mouse strains. Neurobiology of Learning and Memory, 85(2), 164–172. doi:10.1016/J.NLM.2005.09.006
  • Wasserman, A., Matthewson, G., & MacDonald, P. (2018). Platelet-Rich Plasma and the Knee—applications in orthopedic surgery. Current Reviews in Musculoskeletal Medicine, 11(4), 607–615. doi:10.1007/s12178-018-9521-0
  • Yurek, D. M., Hipkens, S. B., Hebert, M. A., Gash, D. M., & Gerhardt, G. A. (1998). Age-related decline in striatal dopamine release and motoric function in Brown Norway/Fischer 344 hybrid rats. Brain Research, 791(1–2), 246–256. doi:10.1016/S0006-8993(98)00110-3
  • Zachrisson, O., Zhao, M., Andersson, A., Dannaeus, K., Häggblad, J., Isacson, R., … Haegerstrand, A. (2011). Restorative effects of platelet derived growth factor-BB in rodent models of Parkinson’s disease. Journal of Parkinson’s Disease, 1(1), 49–63. doi:10.3233/JPD-2011-0003

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.