ArticlePDF Available

Healthy blood, healthy brain: a window into understanding and treating neurodegenerative diseases

Authors:

Abstract and Figures

Our limited understanding of complex neurodegenerative disorders has held us back on the development of efficient therapies. While several approaches are currently being considered, it is still unclear what will be most successful. Among the latest and more novel ideas, the concept of blood or plasma transfusion from young healthy donors to diseased patients is gaining momentum and attracting attention beyond the scientific arena. While young or healthy blood is enriched with protective and restorative components, blood from older subjects may accumulate neurotoxic agents or be impoverished of beneficial factors. In this commentary, we present an overview of the compelling evidence collected in various animal models of brain diseases (e.g., Alzheimer, Parkinson, Huntington) to the actual clinical trials that have been conducted to test the validity of blood-related treatments in neurodegenerative diseases and argue in favor of such approach.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
Journal of Neurology
https://doi.org/10.1007/s00415-024-12337-w
NEUROLOGICAL UPDATE
Healthy blood, healthy brain: awindow intounderstanding
andtreating neurodegenerative diseases
ThyagoR.Cardim‑Pires1· AuréliedeRusJacquet1,2· FrancescaCicchetti1,2
Received: 26 January 2024 / Revised: 17 March 2024 / Accepted: 18 March 2024
© Springer-Verlag GmbH Germany, part of Springer Nature 2024
Abstract
Our limited understanding of complex neurodegenerative disorders has held us back on the development of efficient therapies.
While several approaches are currently being considered, it is still unclear what will be most successful. Among the latest
and more novel ideas, the concept of blood or plasma transfusion from young healthy donors to diseased patients is gaining
momentum and attracting attention beyond the scientific arena. While young orhealthy blood is enriched with protective
and restorative components, blood from older subjects may accumulate neurotoxic agents or be impoverished of beneficial
factors. In this commentary, we present an overview of the compelling evidence collected in various animal models of brain
diseases (e.g., Alzheimer, Parkinson, Huntington) to the actual clinical trials that have been conducted to test the validity of
blood-related treatments in neurodegenerative diseases and argue in favor of such approach.
Keywords Neurodegenerative diseases· Aging· Plasma· Plasma infusion· Plasmapheresis· Rejuvenation
The benets ofblood: ahistorical
perspective
The seventeenth century was marked by a burgeoning of
discoveries in numerous fields including in anatomy and
physiology, which, despite several tribulations, paved the
way to modern medicine. Among them, the English physi-
cian William Harvey provided the first detailed description
of blood circulation and how the heart could pump this fluid
throughout the entire body, including to the brain. A con-
temporary of Harvey, Richard Lower, considered the blood
as a potential treatment for hemorrhage and in 1666, dem-
onstrated the feasibility of blood transfusion in animals. A
few years later, the French physician Jean-Baptiste Denys
performed the first sheep-to-human transfusion (Fig.1).
While the young patient who suffered fever survived the
xenotransfusion, a second subject died and Denys was tri-
aled for homicide, but ultimately acquitted. In the years that
followed, examples of failed attempts outweighed the fore-
seen potential of blood transfusion and led authorities to ban
such practices. It would take more than 150years for this
idea to be revisited and the first human blood transfusion to
be implemented in a patient with postpartum hemorrhage,
thus renewing interest in the use of blood as a therapeutic
means among scientists [1].
The tragedy of World War IIlater prompted increased
investments in science and technologies, in particular in
studies pertaining to blood transfusion. Charles Drew, a
renowned African American surgeon also made a game-
changing discovery by fractionating the blood into red
blood cells and plasma. Drew’s work showed that plasma
is more easily stored than blood, facilitating transportation
[2]. One significant outcome of this new wave of research
led to the development and subsequent use of pooled dried
blood plasma serum, saving the lives of thousands of sol-
diers [3]. Over decades of experimentations, the benefits of
blood became irrefutable, and it propelled the advancement
of approaches using blood from experimental paradigms
to concrete human-based therapies. The concept of blood
banks materialized and heavy campaigning for blood drives
was set in motion (Fig.1).
From an experimental standpoint, the work of the French
physiologist Paul Bert, in the 1800s, was also significant.
Bert developed a model that involved the surgical fusion of
* Francesca Cicchetti
francesca.cicchetti@crchudequebec.ulaval.ca
1 Centre de Recherche du CHU de Québec, Université Laval,
Axe Neurosciences, T2-07, 2705, Boulevard Laurier,
Québec, QCG1V4G2, Canada
2 Département de Psychiatrie & Neurosciences, Université
Laval, Québec, QCG1K0A6, Canada
Journal of Neurology
two living organisms, which he named parabiosis; a term
derived from the English word para (beside) and the Greek
word biosis (model of life) [4]. The ability of parabiosis
to demonstrate the involvement, or lack of involvement, of
circulating factors made it a popular technique to answer a
diverse array of research questions from elements influenc-
ing obesity, to the cause of dental cavities [5]. More recently,
Fig. 1 Chronological timeline of groundbreaking blood-related discoveries
Journal of Neurology
this approachrevealed the benefits of young1 blood in regen-
erating muscle tissue [6], reducing the expression of age-
associated kidney markers, slowing renal tissue damage [7],
and even improving fertility [8]. Exposing an aged animal
to young blood has also proven to promote vascular remod-
eling, neurogenesis, olfactory discrimination, and spatial
memory in older mice [9, 10]. The mechanistic basis for
these striking observations is an active field of investiga-
tion, as the molecular profile of the peripheral circulation
is highly dynamic and reflects general health. For exam-
ple, plasma protein signatures change across the life span
[11]. While aged plasma seemingly contains more “toxic”
elements, young plasma is composed of more protective/
restorative factors [1215].
While tales of the existence of the fountain of youth have
been recounted for thousands of years, blood, plasma and
their derivatives are now considered in modern medicine
as a way to achieve rejuvenation and have the advantage of
already being approved for use in human in different healthy
or pathological conditions. Several fields related to health,
in particular cosmetics and plastic surgery, are indeed using
plasma for tissue regeneration. If healthy plasma can repair
normal signs of aging, could disease processes, which are
recurrently associated with accelerated aging, also be ame-
liorated…? Would considering such blood-related therapy
to treat neurodegenerative diseases be a stretch of the
imagination?
Manipulation ofblood and/orplasma
totreat brain diseases
Before diving into this topic, it should be understood that
blood and/or plasma can be manipulated using various
methodologies. Plasmapheresis, for example, allows to
remove toxic components from the bloodstream. During
this procedure, the blood is fractionated to discard certain
components and subsequently reinjected into the patient.
Plasma exchange is a type of plasmapheresis which con-
sists of replacing aged or diseased blood with a saline solu-
tion, to which albumin can additionally be incorporated to
help reestablish osmotic pressure and restore volume lost
by blood removal [16]. Finally, healthy plasma can be trans-
fused to a diseased patient by plasma infusion.
To generate a tangible impact on the central nerv-
ous system to treat brain pathologies, favorable elements
of the blood would need to cross the blood–brain barrier
(BBB); an entity that tightly controls the traffic of plasma
components (e.g., proteins) between the periphery and the
brain. However, aging affects BBB permeability and older
mice are characterized by less efficient brain uptake than
younger mice [17]. Crossing of the BBB and internalization
of plasma constituents by various brain cells is a physiologi-
cal process [17] and plasma factors linked with youth have
been shown to prompt several desirable effects within the
central nervous system. Among these, it has now repeat-
edly been demonstrated that young plasma can trigger brain
rejuvenation in aged mice by, among various mechanisms,
reducing microglia and astrocyte reactivity, as well as induc-
ing neuronal remyelination [9, 10] and reversing cognitive
impairments induced by anesthesia and surgery [18].
The infusion of healthy plasma in the triple trans-
genicmouse model of Alzheimer's disease (AD), can
further rescue disease hallmarks such as brain accumula-
tion of Aβ plaque and neuroinflammation [19], and con-
currently improve cognitive impairments [20]. Parabiosis
experiments showed that healthy blood can ameliorate AD
pathology causing similar effects as periodical administra-
tion of healthy plasma [21]. The cognitive deficits classically
observed in AD models can be more significantly attenuated
if the healthy plasma source is from an age-matched con-
trol that is submitted to regular physical exercise (access to
spinning wheels) compared to sedentary controls (no access
to spinning wheels) [20]. While young plasma and young
plasma derived from a donor practicing physical exercise
can induce the same beneficial outcomes on the recipient
organism, the molecular mechanisms underlying the pro-
health benefits differ [22]. At the transcriptomic level, young
plasma transferred by parabiosis triggers a strong rejuvena-
tion effect on aged-neural progenitor cells by decreasing the
expression of aging-related genes (e.g., Bgp3, B2m, Rtp4,
Usp18, and others related to interferon-γ response). In con-
trast, exercised plasma does not extensively rejuvenate the
profile of neural progenitors but does upregulate essential
genes that tend to decrease with age (e.g., Cox17) [22].
Preclinical studies conducted in models of Huntington’s
disease (HD) have also demonstrated that by joining the
circulation of a wildtype (WT) and an HD mouse (i.e., the
knock-in zQ175 model), blood can serve as a vehicle topro-
vide factors that both improve or worsen disease [23]. In
the WT animal paired with an HD model, the presence of
mutant huntingtin (mHtt) was observed within blood cells
and plasma. The circulating mHtt within the WT parabiont
led to abnormal protein accumulation in all organs studied
including the kidneys, liver, muscles, and brain. In contrast,
healthy blood ameliorated several features in the diseased
mouse, while not affecting mHtt plasma levels. For exam-
ple, the HD parabiont receiving WT blood accumulated
fewer mHtt aggregates in the liver and kidney, and levels of
1 The United Nations defines young subjects, for statistical pur-
poses, as individuals between 15 and 24years of age. The majority
of studies cited in this commentary refer to “young individuals” as
being between 18 and 30years of age, and older individuals as being
above 60years of age. Most of the reports on mouse models consider
“young adults” between the ages of 2 and 6 months, while above
18months or older are considered “aged animals”.
Journal of Neurology
mitochondrial stress markers were lowered in these organs
when compared with a non-parabiont HD mouse. These
findings were not restricted to the periphery, as an improve-
ment in BBB leakage was observed in the HD mouse after
healthy plasma exposure. In a subsequent study, it was dem-
onstrated that the beneficial effects of parabiosis originated
from soluble molecules and not cellular components of
the blood since bone marrow irradiation of a parabiont did
not abrogate the beneficial and/or deleterious effects in the
paired animal [24].
What factors inyoung orhealthy blood are
benecial tothebrain?
By definition, factors in young blood refer to elements that
differ from an aged subject while healthy blood refers to
factors that differ from a diseased subject. In theory, both
young and healthy blood are enriched with protective fac-
tors when compared to older or ill individuals. To this day,
a number of both young and healthy plasma-derived fac-
tors have been identified as being potentially beneficial for
the brain. For example, low plasmatic levels of α-klotho are
observed during aging as well as in early onset AD [25]
and Parkinson’s disease (PD) patients [26]. Overexpress-
ing this factor in APP/PS1 AD mice enhanced Aβ clearance
and short-term memory [27], while peripheral administra-
tion improved spatial and working memory in both aged
and transgenic mice overexpressing human α-synuclein,
respectively [28]. The myokine FNDC5/Irisin mediates
the advantages of physical exercise on cognitive function
[29] and the overexpression of the recombinant protein
using an adenoviral vector can rescue synaptic plasticity in
an AD mouse model that expresses both human amyloid
precursor protein (APP) and presenilin 1 (PS1) [30]. The
administration of growth differentiation factor 11 to aged
WT mice restored the vascularization of the subventricular
zone and enhanced neurogenesis [31]. This factor does not
cross the BBB and may act on the brain indirectly through
mechanisms involving the regulation of brain endothelial
cells [31]. While these proteins have indeed been tested
in clinical trials for various illnesses (ClinicalTrials.gov
Identifier: NCT03532568, NCT05144672, NCT04133896,
NCT02856074), they have not been evaluated in the realm
of neurodegenerative diseases.
While most studies have looked into proteins to identify
beneficial blood factors, other components such as small
molecules associated with metabolism, exosomes and/or
miRNAs have all been considered [3234]. For example,
ketone bodies — small chain fatty acids mainly produced
by the liver — showed increased plasma levels in young
humans submitted to mixed aerobic and strength exercise
[35]. The metabolic impairments such as decreased glucose
uptake, defective mitochondrial metabolism, reduction of
oxygen consumption and ATP levels or imbalance in reac-
tive oxygen speciesobserved in aged or diseased brains (e.g.,
AD, HD, and Amyotrophic lateral sclerosis) can be attenu-
ated by a ketogenic diet in humans [3640]. In fact, some
clinical trials have already reported the beneficial effect of
ketone bodies in AD patients with significant improvements
in memory-related tests [32, 36].
Can old blood be shaped togenerate
benets tothebrain?
Plasma-derived from both older humans and mice is char-
acterized by the presence of more toxic factors [41, 42].
Accordingly, it can be speculated that the benefits gener-
ated by young orhealthy plasma infusion originate from
the “dilution or elimination” of deleterious components. A
number of studies have shown that in heterochronic parabio-
sis, where animals of different ages are paired together, the
young mouse suffers more damaging effects (i.e., decreased
neurogenesis and hepatogenesis, muscular atrophy, reduced
life expectancy, etc.) than the aged parabiont [6, 15, 43].
Similarly, substituting low amounts of aged plasma with
a saline-albumin solution by plasma exchange can trigger
beneficial effects (i.e., hippocampal neurogenesis) [4446].
Patients with Guillain–Barre syndrome, a demyelinat-
ing disease, are often treated with plasma exchange on the
grounds that this condition has an immune basis triggered
by an infection such as the Zika virus, the cytomegalovirus
or others [4749]. In elderly healthy humans, the removal
of plasma by plasma exchange is associated with decreased
expression of age-related markers (such as oxidized DNA)
within blood cells and the newly generated protein signature
resembles more closely the profile of that found in younger
subjects [45]. Hence, filtration/cleansing of plasma could be
considered a therapeutic approach to treat neurodegenerative
diseases that present with an accumulation of toxic media-
tors such as AD and the amyloid peptide [50].
The use ofblood‑derived products totreat
brain diseases: frombench tobedside
Considering the promising preclinical results obtained in
models of AD, AD-related studies are largely ahead of this
field of investigation and two distinct clinical trials have
already been conducted: the AMBAR and the PLASMA
study. The AMBAR (Alzheimer’s management by albumin
replacement) initiative was a multicentric (US and Spain),
randomized, blinded, and placebo-controlled trial that evalu-
ated the effects of therapeutic plasma exchange (using a 5%
albumin solution) in more than 300 mild-to-moderate AD
Journal of Neurology
patients [51]. In addition to being safe and well-tolerated,
replacing AD plasma with an albumin solution improved
clinical outcomes including memory and language functions
[52]. While patients with milder AD improved in language
fluency, moderate disease patients demonstrated ameliorated
short-term verbal memory [53]. Plasma exchange further
decreased the levels of specific AD markers such as Aβ1-42 in
both blood and cerebrospinal fluid [52]. It was suggested that
the capacity of albumin to bind to Aβ fragments could be
responsible for some of these benefits [51]. The PLASMA
(Plasma for Alzheimer symptom amelioration) study was
conducted by Stanford University and evaluated the effects
of plasma injection in mild-to-moderate AD patients. This
small-scale (n = 18) trial confirmed tolerability and fea-
sibility [54]. One specific plasma fraction, referred to as
GRF6019, was used in a phase II, double-blind, placebo-
controlled in AD patients with mild-to-moderate (n = 47)
[55] or severe dementia (n = 26) [56]. The plasma fraction
infusion proved to be safe and well-tolerated but with no
difference in the preliminary analysis of cognitive func-
tion in either case. A new trial (ExPlas) is set to evaluate
the beneficial effects in early-stage AD patients but using
plasma from exercised donors (ClinicalTrials.gov Identifier:
NCT05068830). The hypothesis is that, as shown in pre-
clinical studies, physical exercise will enrich the blood with
additional beneficial factors.
A second plasma fraction, GRF6021, was tested in PD
patients (n = 79) in a phase II, double-blind, placebo-con-
trolled study to assess safety and tolerability (Clinical-
Trials.gov Identifier: NCT03713957) but the results are
pending. This same fraction is currently being tested to
evaluate its effect on postoperative recovery for primary
hip or knee arthroplasty (ClinicalTrials.gov Identifier:
NCT03981419). In another PD trial (n = 15), the safety
of weekly plasma infusions during a 1-month period was
also evaluated [57] and highlighted, once more, the safety
of this approach. Improvements in phonemic fluency and
in the stigma subscore were measured immediately after
plasma infusion and lasted up to 4weeks. Another clini-
cal trial evaluating the role of young plasma (from 18 to
25years old donors) infused intravenously to PD patients
(n = 22) has been conducted (ClinicalTrials.gov Identifier:
NCT04202757) but results are pending. The infusion of
young plasma in patients with Progressive Supranuclear
Palsy, a primary tauopathy, was also performed (Clini-
calTrials.gov Identifier: NCT02460731). Although there
Fig. 2 Past and current clinical trials involving plasma treatment for neurodegenerative diseases and various conditions
Journal of Neurology
was no difference on disease progression between con-
trol and treated patients, the study revealed that plasma
infusions were safe and well-tolerated [58]. Another trial
was conducted in ALS (ClinicalTrials.gov Identifier:
NCT04454840) but the literature search failed to identify
any reported observations (see Fig.2).
Plasmapheresis, plasma exchange, and plasma infusion
all present with advantages and disadvantages. Plasma-
pheresis and plasma exchange do not require donors and
therefore are less likely to generate an allergic reaction
that would be associated with the procedure per se [59].
While plasma exchange may also lower blood pressure, the
main risk associated with this approach relates to the fact
that the methodology is not restricted to eliminating toxic
factors but also essential components such as electrolytes
and immunoglobulin. By removing such proteins, there is
an increased risk of opportunistic infections [60]. While
plasma infusion requires sample collection from another
individual that increases the risk of allergies and infec-
tions, it allows the transfusion of nutrients and protective
factors found in the young and/or healthy blood. A com-
bination of these approaches, by clearing toxic elements
and replenishing the blood with beneficial factors, may
ultimately be the best angle to tackle neurodegenerative
diseases.
Final consideration
Based on the compelling preclinical evidence reported in
the literature, which has rapidly translated into a number
of clinical trials, blood-related approaches to treat neu-
rodegenerative diseases are not a stretch of the imagina-
tion. The brain is the most vascularized organ of the entire
human body and a continuous source of healthy blood is
needed for its optimal function. While we still need to
interrogate the blood to zoom in on the key components
that can have beneficial or detrimental effects, perhaps
the twenty-first century will be mirrored by the pioneer-
ing work of Harvey, Lower, Denys, Drew and others, and
lead to the treatment of neurodegenerative diseases using
blood.
Acknowledgements FC is a recipient of a Researcher Chair from the
Fonds de Recherche du Québec en Santé (FRQS, 35059) providing sal-
ary support and operating funds, and receives funding from the Cana-
dian Institutes of Health Research (CIHR, PJT162164 and PJT168865)
to conduct her HD-related research. ADRJ is supported by a Launch
Award from the Parkinson’s Foundation and funds from the Fondation
CHU de Québec.
Author contributions TRCP reviewed the literature, conceptualized
figures and wrote the manuscript. ADRJ contributed to the literature
review and edited the manuscript. FC contributed to the literature
review, conceptualized figures and wrote the manuscript.
Declarations
Conflicts of interest The authors declare that they have no conflict of
interest.
References
1. Baskett TF (2002) James Blundell: the first transfusion of
human blood. Resuscitation 52:229–233. https:// doi. org/ 10.
1016/ S0300- 9572(02) 00013-8
2. Tan SY, Merritt C (2017) Charles Richard Drew (1904-1950):
father of blood banking. Singap Med J 58:593–594. https:// doi.
org/ 10. 11622/ SMEDJ. 20170 99
3. Schmidt PJ (2012) The plasma wars: a history. Transfusion
(Paris). https:// doi. org/ 10. 1111/J. 1537- 2995. 2012. 03689.X
4. Bert Paul (1864) Expériences et considérations sur la greffe
animale, pp 1–23
5. Kamrin BB (1954) The effects of a high carbohydrate diet on the
teeth of parabiosed albino rats. J Dent Res 33:175–180. https://
doi. org/ 10. 1177/ 00220 34554 03300 20301/ ASSET/ 00220 34554
03300 20301. FP. PNG_ V03
6. Rebo J, Mehdipour M, Gathwala R etal (2016) A single het-
erochronic blood exchange reveals rapid inhibition of multiple
tissues by old blood. Nat Commun 7:1–11. https:// doi. org/ 10.
1038/ ncomm s13363
7. Huang Q, Ning Y, Liu D etal (2018) A young blood environment
decreases aging of senile mice kidneys. J Gerontol Ser A Biol Sci
Med Sci 73:421–428. https:// doi. org/ 10. 1093/ gerona/ glx183
8. Erdogan K, Ceylani T, Teker HT etal (2023) Young plasma trans-
fer recovers decreased sperm counts and restores epigenetics in
aged testis. Exp Gerontol 172:112042. https:// doi. org/ 10. 1016/j.
exger. 2022. 112042
9. Katsimpardi L, Litterman NK, Schein PA etal (1979) (2014) Vas-
cular and neurogenic rejuvenation of the aging mouse brain by
young systemic factors. Science 344:630–634. https:// doi. org/ 10.
1126/ scien ce. 12511 41
10. Villeda SA, Plambeck KE, Middeldorp J etal (2014) Young blood
reverses age-related impairments in cognitive function and syn-
aptic plasticity in mice. Nat Med 20:659–663. https:// doi. org/ 10.
1038/ nm. 3569
11. Lehallier B, Gate D, Schaum N etal (2019) Undulating changes
in human plasma proteome profiles across the lifespan. Nat Med
25:1843–1850. https:// doi. org/ 10. 1038/ s41591- 019- 0673-2
12. Gan KJ, Südhof TC (2019) Specific factors in blood from young
but not old mice directly promote synapse formation and NMDA-
receptor recruitment. Proc Natl Acad Sci U S A 116:12524–
12533. https:// doi. org/ 10. 1073/ pnas. 19026 72116
13. Bieri G, Schroer AB, Villeda SA (2023) Blood-to-brain commu-
nication in aging and rejuvenation. Nat Neurosci Rev. https:// doi.
org/ 10. 1038/ s41593- 022- 01238-8
14. Villeda SA, Luo J, Mosher KI etal (2011) The ageing systemic
milieu negatively regulates neurogenesis and cognitive function.
Nature 477:90–96. https:// doi. org/ 10. 1038/ natur e10357
15. Jeon OH, Mehdipour M, Gil TH etal (2022) Systemic induc-
tion of senescence in young mice after single heterochronic
blood exchange. Nat Metab 4:995–1006. https:// doi. org/ 10. 1038/
s42255- 022- 00609-6
16. Caraceni P, Tufoni M, Bonavita ME (2013) Clinical use of albu-
min. Blood Transfus. DOI 10(2450/2013):005S
Journal of Neurology
17. Yang AC, Stevens MY, Chen MB et al (2020) Physiologi-
cal blood–brain transport is impaired with age by a shift in
transcytosis. Nature 583:425–430. https:// doi. org/ 10. 1038/
s41586- 020- 2453-z
18. Li Y, Zhang Q, Yan W etal (2022) Young plasma reverses anes-
thesia and surgery-induced cognitive impairment in aged rats by
modulating hippocampal synaptic plasticity. Front Aging Neuro-
sci. https:// doi. org/ 10. 3389/ fnagi. 2022. 996223
19. Zhao Y, Qian R, Zhang J etal (2020) Young blood plasma reduces
Alzheimer’s disease-like brain pathologies and ameliorates cogni-
tive impairment in 3×Tg-AD mice. Alzheimers Res Ther 12:1–13.
https:// doi. org/ 10. 1186/ s13195- 020- 00639-w
20. Kim TW, Park SS, Park JY, Park HS (2020) Infusion of plasma
from exercised mice ameliorates cognitive dysfunction by increas-
ing hippocampal neuroplasticity and mitochondrial functions in
3xtg-ad mice. Int J Mol Sci. https:// doi. org/ 10. 3390/ ijms2 10932
91
21. Middeldorp J, Lehallier B, Villeda SA etal (2016) Preclinical
assessment of young blood plasma for Alzheimer disease. JAMA
Neurol 73:1325–1333. https:// doi. org/ 10. 1001/ jaman eurol. 2016.
3185
22. Buckley MT, Sun ED, George BM etal (2023) Cell-type-specific
aging clocks to quantify aging and rejuvenation in neurogenic
regions of the brain. Nat Aging 3:121–137. https:// doi. org/ 10.
1038/ s43587- 022- 00335-4
23. Rieux M, Alpaugh M, Sciacca G etal (2021) Shedding a new light
on Huntington’s disease: how blood can both propagate and ame-
liorate disease pathology. Mol Psychiatry 26:5441–5463. https://
doi. org/ 10. 1038/ s41380- 020- 0787-4
24. Rieux M, Alpaugh M, Salem S etal (2023) Understanding the role
of the hematopoietic niche in Huntington’s disease’s phenotypic
expression: invivo evidence using a parabiosis model. Neurobiol
Dis. https:// doi. org/ 10. 1016/j. nbd. 2023. 106091
25. Fung TY, Iyaswamy A, Sreenivasmurthy SG etal (2022) Klotho
an autophagy stimulator as a potential therapeutic target for Alz-
heimer’s disease: a review. Biomedicines. https:// doi. org/ 10. 3390/
BIOME DICIN ES100 30705
26. Sancesario GM, Di Lazzaro G, Grillo P etal (2021) Biofluids pro-
file of α-Klotho in patients with Parkinson’s disease. Parkinsonism
Relat Disord 90:62–64. https:// doi. org/ 10. 1016/j. parkr eldis. 2021.
08. 004
27. Zhao Y, Zeng CY, Li XH etal (2020) Klotho overexpression
improves amyloid-β clearance and cognition in the APP/PS1
mouse model of Alzheimer’s disease. Aging Cell 19:e13239.
https:// doi. org/ 10. 1111/ ACEL. 13239
28. Leon J, Moreno AJ, Garay BI etal (2017) Peripheral elevation of a
klotho fragment enhances brain function and resilience in young,
aging, and α-synuclein transgenic mice. Cell Rep 20:1360–1371.
https:// doi. org/ 10. 1016/j. celrep. 2017. 07. 024
29. Islam MR, Valaris S, Young MF etal (2021) Exercise hormone
irisin is a critical regulator of cognitive function. Nat Metab
3:1058–1070. https:// doi. org/ 10. 1038/ s42255- 021- 00438-z
30. Lourenco MV, Frozza RL, de Freitas GB etal (2019) Exercise-
linked FNDC5/irisin rescues synaptic plasticity and memory
defects in Alzheimer’s models. Nat Med 25:165–175. https:// doi.
org/ 10. 1038/ s41591- 018- 0275-4
31. Ozek C, Krolewski RC, Buchanan SM, Rubin LL (2018) Growth
Differentiation Factor 11 treatment leads to neuronal and vascular
improvements in the hippocampus of aged mice. Sci Rep. https://
doi. org/ 10. 1038/ S41598- 018- 35716-6
32. Reger MA, Henderson ST, Hale C etal (2004) Effects of
β-hydroxybutyrate on cognition in memory-impaired adults.
Neurobiol Aging 25:311–314. https:// doi. org/ 10. 1016/ S0197-
4580(03) 00087-3
33. Xhuti D, Nilsson MI, Manta K etal (2023) Circulating exo-
some-like vesicle and skeletal muscle microRNAs are altered
with age and resistance training. J Physiol. https:// doi. org/ 10.
1113/ JP282 663
34. Yang P, Dong X, Zhang Y (2020) MicroRNA profiles in plasma
samples from young metabolically healthy obese patients and
miRNA-21 are associated with diastolic dysfunction via TGF-
β1/Smad pathway. J Clin Lab Anal. https:// doi. org/ 10. 1002/
JCLA. 23246
35. Koay YC, Stanton K, Kienzle V etal (2021) Effect of chronic
exercise in healthy young male adults: A metabolomic analysis.
Cardiovasc Res 117:613–622. https:// doi. org/ 10. 1093/ cvr/ cv aa0
51
36. Taylor MK, Sullivan DK, Mahnken JD etal (2018) Feasibility
and efficacy data from a ketogenic diet intervention in Alz-
heimer’s disease. Alzheimer’s Dement Transl Res Clin Interv
4:28–36. https:// doi. org/ 10. 1016/j. trci. 2017. 11. 002
37. Gómora-García JC, Montiel T, Hüttenrauch M etal (2023)
Effect of the ketone body, D-β-hydroxybutyrate, on sirtuin2-
mediated regulation of mitochondrial quality control and the
autophagy-lysosomal pathway. Cells. https:// doi. org/ 10. 3390/
CELLS 12030 486
38. Vandoorne T, De Bock K, Van Den Bosch L (2018)
Energy metabolism in ALS: an underappreciated opportu-
nity? Acta Neuropathol 135:489. https:// doi. org/ 10. 1007/
S00401- 018- 1835-X
39. Pagano G, Niccolini F, Politis M (2016) Current status of PET
imaging in Huntington’s disease. Eur J Nucl Med Mol Imaging
43:1171–1182. https:// doi. org/ 10. 1007/ S00259- 016- 3324-6/
TABLES/2
40. Cunnane S, Nugent S, Roy M etal (2011) Brain fuel metabo-
lism, aging, and Alzheimer’s disease. Nutrition 27:3–20. https://
doi. org/ 10. 1016/J. NUT. 2010. 07. 021
41. Sathyan S, Ayers E, Gao T etal (2020) Plasma proteomic pro-
file of age, health span, and all-cause mortality in older adults.
Aging Cell. https:// doi. org/ 10. 1111/ ACEL. 13250
42. Smith LK, He Y, Park JS etal (2015) β2-microglobulin is a
systemic pro-aging factor that impairs cognitive function and
neurogenesis. Nat Med 21:932–937. https:// doi. org/ 10. 1038/
NM. 3898
43. Yankova T, Dubiley T, Shytikov D, Pishel I (2022) Three month
heterochronic parabiosis has a deleterious effect on the lifespan
of young animals, without a positive effect for old animals.
Rejuvenation Res 25:191–199. https:// doi. org/ 10. 1089/ rej. 2022.
0029
44. Mehdipour M, Mehdipour T, Skinner CM etal (2021) Plasma
dilution improves cognition and attenuates neuroinflammation
in old mice. Geroscience 43:1–18. https:// doi. org/ 10. 1007/
s11357- 020- 00297-8
45. Kim D, Kiprov DD, Luellen C etal (2022) Old plasma dilution
reduces human biological age: a clinical study. Geroscience
44:2701–2720. https:// doi. org/ 10. 1007/ s11357- 022- 00645-w
46. Mehdipour M, Etienne J, Liu C etal (2021) Attenuation of age-
elevated blood factors by repositioning plasmapheresis: a novel
perspective and approach. Transfus Apheres Sci 60:103162.
https:// doi. org/ 10. 1016/j. trans ci. 2021. 103162
47. Chevret S, Hughes RAC, Annane D (2017) Plasma exchange for
Guillain-Barré syndrome. Cochrane Database Syst Rev. https://
doi. org/ 10. 1002/ 14651 858. CD001 798. PUB3
48. Tiwari A, Setya D, Tanna D etal (2023) Patient outcome in
antibody-positive systemic vasculitis treated with therapeutic
plasma exchange. Asian J Transfus Sci 17:79–84. https:// doi.
org/ 10. 4103/ AJTS. AJTS_ 50_ 21
49. Madden J, Spadaro A, Koyfman A, Long B (2024) High risk and
low prevalence diseases: Guillain-Barré syndrome. Am J Emerg
Med 75:90–97
Journal of Neurology
50. Bu XL, Xiang Y, Jin WS etal (2018) Blood-derived amyloid-β
protein induces Alzheimer’s disease pathologies. Mol Psychiatry
23:1948–1956. https:// doi. org/ 10. 1038/ mp. 2017. 204
51. Boada M, López O, Núñez L etal (2019) Plasma exchange for
Alzheimer’s disease Management by Albumin Replacement
(AMBAR) trial: Study design and progress. Alzheimer’s Dement
Transl Res Clin Interv 5:61–69. https:// doi. org/ 10. 1016/j. trci.
2019. 01. 001
52. Boada M, Anaya F, Ortiz P etal (2017) Efficacy and safety of
plasma exchange with 5% albumin to modify cerebrospinal fluid
and plasma amyloid-β concentrations and cognition outcomes in
Alzheimer’s disease patients: A multicenter, randomized, con-
trolled clinical trial. J Alzheimer’s Dis 56:129–143. https:// doi.
org/ 10. 3233/ JAD- 160565
53. Boada M, López OL, Olazarán J etal (2022) Neuropsychologi-
cal, neuropsychiatric, and quality-of-life assessments in Alzhei-
mer’s disease patients treated with plasma exchange with albumin
replacement from the randomized AMBAR study. Alzheimer’s
Dement 18:1314–1324. https:// doi. org/ 10. 1002/ alz. 12477
54. Sha SJ, Deutsch GK, Tian L etal (2019) Safety, tolerability, and
feasibility of young plasma infusion in the plasma for Alzheimer
symptom amelioration study: a randomized clinical trial. JAMA
Neurol 76:35–40. https:// doi. org/ 10. 1001/ jaman eurol. 2018. 3288
55. Hannestad J, Koborsi K, Klutzaritz V etal (2020) Safety and
tolerability of GRF6019 in mild-to-moderate Alzheimer’s disease
dementia. Alzheimer’s Dementia Transl Res Clin Interv 6:1–10.
https:// doi. org/ 10. 1002/ trc2. 12115
56. Hannestad J, Duclos T, Chao W etal (2021) Safety and toler-
ability of GRF6019 infusions in severe Alzheimer’s disease: a
phase II double-blind placebo-controlled trial. J Alzheimer’s Dis
81:1649–1662. https:// doi. org/ 10. 3233/ JAD- 210011
57. Parker JE, Martinez A, Deutsch GK etal (2020) Safety of plasma
infusions in Parkinson’s disease. Mov Disord 35:1905–1913.
https:// doi. org/ 10. 1002/ mds. 28198
58. VandeVrede L, Dale ML, Fields S etal (2020) Open-label phase
1 futility studies of salsalate and young plasma in progressive
supranuclear palsy. Mov Disord Clin Pract 7:440–447. https://
doi. org/ 10. 1002/ mdc3. 12940
59. Altobelli C, Anastasio P, Cerrone A etal (2023) Therapeutic
plasmapheresis: a revision of literature. Kidney Blood Press Res
48:66–78. https:// doi. org/ 10. 1159/ 00052 8556
60. Weinstein R (2023) Basic principles of therapeutic plasma
exchange. Transfus Apher Sci. https:// doi. org/ 10. 1016/J. TRANS
CI. 2023. 103675
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
BACKGROUND: Therapeutic plasma exchange (TPE) has been advocated as an adjunct to steroids and cytotoxic drugs in treating patients suffering from vasculitis and presenting with active disease, but we still have insufficient evidence on its effectiveness in improving the clinical response, especially in India. This study was planned to study the clinical outcome in severe vasculitic presentations treated with TPE as an adjunctive therapy. MATERIALS AND METHODS: A retrospective analysis of TPE procedures performed from July 2013 to July 2017 in the department of transfusion medicine at a large tertiary care hospital was done. All consecutive patients admitted with new diagnosis of systemic vasculitis presenting with active disease and severe presentations such as advanced renal failure or severe respiratory abnormalities or life-threatening vasculitis affecting the gastrointestinal tract, neurological and musculoskeletal system; who needed TPE for removal of preformed antibodies, were included in the study. RESULTS: There were a total of 31 patients in whom TPE was performed for severe systemic vasculitis; 26 adults and five pediatric. Six patients tested positive for perinuclear fluorescence, 13 for cytoplasmic fluorescence (cANCA), two for atypical antineutrophil cytoplasmic autoantibody, seven for anti-glomerular basement membrane antibodies, two for antinuclear antibodies (ANA), and one patient tested positive for ANA as well as cANCA before the augmentation of TPE. Out of 31, seven patients showed no clinical improvement and succumbed to the disease. At the end of desired number of procedures, 19 tested negative and five tested weak positive for their respective antibodies. CONCLUSION: Favorable clinical outcomes were observed with TPE in patients with antibody-positive systemic vasculitis.
Article
Full-text available
Mitochondrial activity and quality control are essential for neuronal homeostasis as neurons rely on glucose oxidative metabolism. The ketone body, D-β-hydroxybutyrate (D-BHB), is metabolized to acetyl-CoA in brain mitochondria and used as an energy fuel alternative to glucose. We have previously reported that D-BHB sustains ATP production and stimulates the autophagic flux under glucose deprivation in neurons; however, the effects of D-BHB on mitochondrial turnover under physiological conditions are still unknown. Sirtuins (SIRTs) are NAD+-activated protein deacetylases involved in the regulation of mitochondrial biogenesis and mitophagy through the activation of transcription factors FOXO1, FOXO3a, TFEB and PGC1α coactivator. Here, we aimed to investigate the effect of D-BHB on mitochondrial turnover in cultured neurons and the mechanisms involved. Results show that D-BHB increased mitochondrial membrane potential and regulated the NAD+/NADH ratio. D-BHB enhanced FOXO1, FOXO3a and PGC1α nuclear levels in an SIRT2-dependent manner and stimulated autophagy, mitophagy and mitochondrial biogenesis. These effects increased neuronal resistance to energy stress. D-BHB also stimulated the autophagic–lysosomal pathway through AMPK activation and TFEB-mediated lysosomal biogenesis. Upregulation of SIRT2, FOXOs, PGC1α and TFEB was confirmed in the brain of ketogenic diet (KD)-treated mice. Altogether, the results identify SIRT2, for the first time, as a target of D-BHB in neurons, which is involved in the regulation of autophagy/mitophagy and mitochondrial quality control.
Article
Full-text available
The age‐related loss of skeletal muscle mass and functionality, known as sarcopenia, is a critical risk factor for morbidity and all‐cause mortality. Resistance exercise training (RET) is the primary countermeasure to fight sarcopenia and ageing. Altered intercellular communication is a hallmark of ageing, which is not well elucidated. Circulating extracellular vesicles (EVs), including exosomes, contribute to intercellular communication by delivering microRNAs (miRNAs), which modulate post‐translational modifications, and have been shown to be released following exercise. There is little evidence regarding how EVs or EV‐miRNAs are altered with age or RET. Therefore, we sought to characterize circulating EVs in young and older individuals, prior to and following a 12‐week resistance exercise programme. Plasma EVs were isolated using size exclusion chromatography and ultracentrifugation. We found that ageing reduced circulating expression markers of CD9, and CD81. Using late‐passage human myotubes as a model for ageing in vitro, we show significantly lower secreted exosome‐like vesicles (ELVs). Further, levels of circulating ELV‐miRNAs associated with muscle health were lower in older individuals at baseline but increased following RET to levels comparable to young. Muscle biopsies show similar age‐related reductions in miRNA expressions, with largely no effect of training. This is reflected in vitro, where aged myotubes show significantly reduced expression of endogenous and secreted muscle‐specific miRNAs (myomiRs). Lastly, proteins associated with ELV and miRNA biogenesis were significantly higher in both older skeletal muscle tissues and aged human myotubes. Together we show that ageing significantly affects ELV and miRNA cargo biogenesis, and release. RET can partially normalize this altered intercellular communication. image Key points We show that ageing reduces circulating expression of exosome‐like vesicle (ELV) markers, CD9 and CD81. Using late‐passage human skeletal myotubes as a model of ageing, we show that secreted ELV markers are significantly reduced in vitro. We find circulating ELV miRNAs associated with skeletal muscle health are lower in older individuals but can increase following resistance exercise training (RET). In skeletal muscle, we find altered expression of miRNAs in older individuals, with no effect of RET. Late‐passage myotubes also appear to have aberrant production of endogenous myomiRs with lower abundance than youthful counterparts In older skeletal muscle and late‐passage myotubes, proteins involved with ELV‐ and miRNA biogenesis are upregulated
Article
Full-text available
The diversity of cell types is a challenge for quantifying aging and its reversal. Here we develop ‘aging clocks’ based on single-cell transcriptomics to characterize cell-type-specific aging and rejuvenation. We generated single-cell transcriptomes from the subventricular zone neurogenic region of 28 mice, tiling ages from young to old. We trained single-cell-based regression models to predict chronological age and biological age (neural stem cell proliferation capacity). These aging clocks are generalizable to independent cohorts of mice, other regions of the brains, and other species. To determine if these aging clocks could quantify transcriptomic rejuvenation, we generated single-cell transcriptomic datasets of neurogenic regions for two interventions—heterochronic parabiosis and exercise. Aging clocks revealed that heterochronic parabiosis and exercise reverse transcriptomic aging in neurogenic regions, but in different ways. This study represents the first development of high-resolution aging clocks from single-cell transcriptomic data and demonstrates their application to quantify transcriptomic rejuvenation.
Article
Full-text available
Background: Therapeutic Plasmapheresis (TP) is an extracorporeal therapy that allows the removal of pathogens from plasma. The role of TP in immuno-mediated diseases and toxic conditions has been of interest for decades. Summary: We reviewed the recent literature on the application and the optimal choice of TP technique ranging from Plasma Exchange, Double Filtration Plasmapheresis, Rheopheresis, Immunoadsorptions and Lipidoapheresis. In addition, we report our experience in the application of TP for various diseases ranging in different medical specialties, following the American Society for Apheresis (ASFA) recommendations. Key messages: Overall patients receiving TP showed an improvement in clinical and laboratory parameters. Our review and single center experience suggest a benefit of the application of TP in multiple clinical disciplines.
Article
In a previous study, we have shown that parabiotic coupling of a knock-in mouse model (zQ175) of Huntington's disease (HD) to wild-type (WT) littermates resulted in a worsening of the normal phenotype as seen by detection of mutant huntingtin protein (mHTT) aggregates within peripheral organs and the cerebral cortex as well as vascular abnormalities in WT mice. In contrast, parabiosis improved disease features in the zQ175 mice such as reduction of mHTT aggregate number in the liver and cortex, decrease in blood-brain barrier (BBB) permeability and attenuation of mitochondrial impairments. While the shared circulation mediated these effects, no specific factor was identified. To better understand which blood elements were involved in the aforementioned changes, WT and zQ175 mice underwent parabiotic surgery prior to exposing one of the paired animals to irradiation. The irradiation procedure successfully eliminated the hematopoietic niche followed by repopulation with cells originating from the non-irradiated parabiont, as measured by the quantification of mHTT levels in peripheral blood mononuclear cells. Although irradiation of the WT parabiont, causing the loss of healthy hematopoietic cells, did lead to a few alterations in mitochondrial functions in the muscle (TOM40 levels), and increased neuroinflammation in the striatum (GFAP levels), most of the changes observed were likely attributable to the irradiation procedure itself (e.g. mHTT aggregates in cortex and liver; cellular stress in peripheral organs). However, factors such as mHTT aggregation in the brain and periphery, and BBB leakage, which were improved in zQ175 mice when paired to WT littermate in the previous parabiosis experiment, were unaffected by perturbation of the hematopoietic niche. It would therefore appear that cells of the hematopoietic stem cell niche are largely uninvolved in the beneficial effects of parabiosis.
Article
BACKGROUND: Therapeutic plasma exchange (TPE) has been advocated as an adjunct to steroids and cytotoxic drugs in treating patients suffering from vasculitis and presenting with active disease, but we still have insufficient evidence on its effectiveness in improving the clinical response, especially in India. This study was planned to study the clinical outcome in severe vasculitic presentations treated with TPE as an adjunctive therapy.
Article
Therapeutic plasma exchange is a method of treatment for clinical conditions that represent diverse fields of medicine. The rationale for this mode of therapy is based on sound mathematical modeling of the synthesis and removal of large molecules, usually proteins, from the circulation. The basic assumptions underlying therapeutic plasma exchange are that a clinical illness is caused by, or related to, a pathogenic substance in the plasma, and that removing that substance from the plasma will alleviate the patient's illness. This approach has proven applicable to a wide variety of clinical conditions. Therapeutic plasma exchange is largely a safe procedure in experienced hands. The principal adverse effect, the hypocalcemic reaction, is readily ameliorated or prevented.
Article
Aging induces molecular, cellular and functional changes in the adult brain that drive cognitive decline and increase vulnerability to dementia-related neurodegenerative diseases. Leveraging systemic and lifestyle interventions, such as heterochronic parabiosis, administration of ‘young blood’, exercise and caloric restriction, has challenged prevalent views of brain aging as a rigid process and has demonstrated that aging-associated cognitive and cellular impairments can be restored to more youthful levels. Technological advances in proteomic and transcriptomic analyses have further facilitated investigations into the functional impact of intertissue communication on brain aging and have led to the identification of a growing number of pro-aging and pro-youthful factors in blood. In this review, we discuss blood-to-brain communication from a systems physiology perspective with an emphasis on blood-derived signals as potent drivers of both age-related brain dysfunction and brain rejuvenation. In this Review, Villeda and colleagues describe blood-to-brain communication from a systems physiology perspective, with an emphasis on blood-derived signals as potent drivers of both age-related brain dysfunction and brain rejuvenation.