Rejuvenating the blood and bone marrow to slow aging-associated cognitive decline and Alzheimer's disease

Seokjo Kang, V Alexandra Moser, Clive N Svendsen, Helen S Goodridge, Seokjo Kang, V Alexandra Moser, Clive N Svendsen, Helen S Goodridge

Abstract

Parabiosis, blood exchange and plasma transfer experiments have highlighted the rejuvenating properties of young blood. Our Communications Biology study demonstrated that young bone marrow transplantation attenuates cognitive decline in old mice, with preservation of hippocampal synapses and reduced microglial reactivity. We now discuss subsequent studies that shed additional light on how blood impacts cognitive function, and potential clinical applications, including ongoing clinical trials with young plasma and experimental strategies targeting the hematopoietic system to slow or reverse cognitive decline.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Potential peripheral interventions to prevent…
Fig. 1. Potential peripheral interventions to prevent or rescue aging-associated cognitive decline.
A greater understanding of the impacts of circulating factors on the hippocampus may inform strategies to replace youthful factors lost during aging (a) or neutralize pro-aging factors (b). Alternatively, it may be more effective to deliver young or rejuvenated HSCs (c) or differentiated hematopoietic cells (d), or to target the bone marrow niche to rejuvenate endogenous HSCs (e).

References

    1. Conboy IM, Rando TA. Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle. 2012;11:2260–2267. doi: 10.4161/cc.20437.
    1. Eggel A, Wyss-Coray T. A revival of parabiosis in biomedical research. Swiss Med. Wkly. 2014;144:w13914.
    1. Bouchard J, Villeda SA. Aging and brain rejuvenation as systemic events. J. Neurochem. 2015;132:5–19. doi: 10.1111/jnc.12969.
    1. Rebo J, et al. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat. Commun. 2016;7:13363. doi: 10.1038/ncomms13363.
    1. Castellano JM, et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature. 2017;544:488–492. doi: 10.1038/nature22067.
    1. Villeda SA, et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 2014;20:659–663. doi: 10.1038/nm.3569.
    1. Fan X, Wheatley EG, Villeda SA. Mechanisms of Hippocampal Aging and the Potential for Rejuvenation. Annu Rev. Neurosci. 2017;40:251–272. doi: 10.1146/annurev-neuro-072116-031357.
    1. Das MM, et al. Young bone marrow transplantation preserves learning and memory in old mice. Commun. Biol. 2019;2:73. doi: 10.1038/s42003-019-0298-5.
    1. Gan KJ, Sudhof TC. Specific factors in blood from young but not old mice directly promote synapse formation and NMDA-receptor recruitment. Proc. Natl Acad. Sci. USA. 2019;116:12524–12533. doi: 10.1073/pnas.1902672116.
    1. Yousef H, et al. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat. Med. 2019;25:988–1000. doi: 10.1038/s41591-019-0440-4.
    1. Xia E, et al. Young blood rescues the cognition of Alzheimer’s model mice by restoring the hippocampal cholinergic circuit. Neuroscience. 2019;417:57–69. doi: 10.1016/j.neuroscience.2019.08.010.
    1. Middeldorp J, et al. Preclinical assessment of young blood plasma for Alzheimer disease. JAMA Neurol. 2016;73:1325–1333. doi: 10.1001/jamaneurol.2016.3185.
    1. Sha SJ, et al. Safety, tolerability, and feasibility of young plasma infusion in the plasma for Alzheimer symptom amelioration study: a randomized clinical trial. JAMA Neurol. 2019;76:35–40. doi: 10.1001/jamaneurol.2018.3288.
    1. Katsimpardi L, et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science. 2014;344:630–634. doi: 10.1126/science.1251141.
    1. Ozek C, Krolewski RC, Buchanan SM, Rubin LL. Growth Differentiation Factor 11 treatment leads to neuronal and vascular improvements in the hippocampus of aged mice. Sci. Rep. 2018;8:17293. doi: 10.1038/s41598-018-35716-6.
    1. Jin K, et al. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc. Natl Acad. Sci. USA. 2002;99:11946–11950. doi: 10.1073/pnas.182296499.
    1. Maung KK, Horwitz ME. Current and future perspectives on allogeneic transplantation using ex vivo expansion or manipulation of umbilical cord blood cells. Int J. Hematol. 2019;110:50–58. doi: 10.1007/s12185-019-02670-6.
    1. Sugimura R, et al. Haematopoietic stem and progenitor cells from human pluripotent stem cells. Nature. 2017;545:432–438. doi: 10.1038/nature22370.
    1. Tan YT, et al. Respecifying human iPSC-derived blood cells into highly engraftable hematopoietic stem and progenitor cells with a single factor. Proc. Natl Acad. Sci. USA. 2018;115:2180–2185. doi: 10.1073/pnas.1718446115.
    1. Zeisig BB, So CWE. MLL-AF4, a double-edged sword for iPSC respecification into HSPCs. Proc. Natl Acad. Sci. USA. 2018;115:1964–1966. doi: 10.1073/pnas.1800622115.
    1. Koronyo Y, et al. Therapeutic effects of glatiramer acetate and grafted CD115(+) monocytes in a mouse model of Alzheimer’s disease. Brain. 2015;138:2399–2422. doi: 10.1093/brain/awv150.
    1. Ho YH, et al. Remodeling of bone marrow hematopoietic stem cell niches promotes myeloid cell expansion during premature or physiological aging. Cell Stem Cell. 2019;25:407–418 e406. doi: 10.1016/j.stem.2019.06.007.
    1. Maryanovich M, et al. Adrenergic nerve degeneration in bone marrow drives aging of the hematopoietic stem cell niche. Nat. Med. 2018;24:782–791. doi: 10.1038/s41591-018-0030-x.
    1. Guidi N, et al. Osteopontin attenuates aging-associated phenotypes of hematopoietic stem cells. EMBO J. 2017;36:840–853. doi: 10.15252/embj.201694969.

Source: PubMed

Подписаться