Novel therapeutic approaches to target neurodegenerationRunning title: Tackling pathways contributing to neurodegeneration
Alerie G. de la Fuente1,2,3, *, Silvia
Pelucchi4,5, *, Jerome Mertens5,
Monica Di Luca4, Daniela Mauceri6,7,#, Elena Marcello4,#1 Instituto de Investigación Sanitaria y Biomédica de
Alicante (ISABIAL), Avenida Pintor Baeza 12, 03010 Alicante, Spain2 Instituto de Neurociencias CSIC-UMH, Avenida Ramón y
Cajal, 03550, San Juan de Alicante, Spain3 Wellcome-Wolfson Institute for Experimental
Medicine, Queen’s University Belfast, 97 Lisburn Road, BT9 7BL, Belfast,
UK4 Department of Pharmacological and Biomolecular
Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy.5 Neural Aging Laboratory, Institute of Molecular
Biology, Leopold-Franzens-University; Innsbruck, Austria6 Institute of Anatomy and Cell Biology, Dept.
Molecular and Cellular Neuroscience, University of Marburg,
Robert-Koch-Str. 8, 35032 Marburg, Germany.7 Department of Neurobiology, Interdisciplinary Centre
for Neurosciences (IZN), Heidelberg University, INF 366, 69120
Heidelberg, Germany.
* These authors are the co-first authors
# These authors are the co-senior authors
Corresponding author:
Elena Marcello
Dept. Pharmacological and Biomolecular Sciences - University of Milan
Via Balzaretti 9 – 20133 Milan
Email: elena.marcello@unimi.it
Word count: 9524AcknowledgementsThe authors would like to apologize to colleagues whose work has not
been mentioned or discussed here due to constraints of space and scope.
We have attempted to provide a balanced representation of different
topics and mechanistic aspects but also acknowledge that this work will
not be comprehensive and will not realistically cover the vast body of
pertaining literature.
We acknowledge funding from the Italian Ministry of University and
Research (PRIN 202039WMFP to E.M. and 20202THZAW to M.D.L.), from
Fondazione Cariplo (Grant no. 2018-0511 to E.M.), from Alzheimer
Forschung Initiative (Grant no. 21019) and the Chica and Heinz Schaller
Foundation to D.M., from Spanish Health Institute Carlos III (CP21/0032
to A.G.F.) and Spanish Science State Agency (PID2021-124465OA-I00 to
A.G.F.), from the BrightFocus Foundation (grant A2019562S to J.M.); from
the European Union (grants ERC-STG-2019-852086 and
H2020-MSCA-IF-2017-797205 to J.M.); from the Austrian Science Fund
(grant FWF-I5057 to J.M.), Clene Nanomedicine to J.M., the U.S. National
Academy of Medicine (NAM) to J.M., the Michael J. Fox Foundation (MJFF)
to J.M., and the NIH National Institute on Aging (grant AG056306 to JM).
Figure was created with BioRender.com.Conflict of interestAll the authors have no conflict of interest.AbstractAgeing is the main risk factor common to most primary neurodegenerative
disorders. Indeed, age-related brain alterations have been long
considered to predispose to neurodegeneration. Although protein
misfolding and the accumulation of toxic protein aggregates have been
contemplated as causative events in neurodegeneration, several
biological pathways affected by brain ageing are also contributing to
pathogenesis.
Here, we discuss the evidence showing the involvement of the mechanisms
controlling neuronal structure, gene expression, autophagy, cell
metabolism, and neuroinflammation in the onset and progression of
neurodegenerative disorders. Furthermore, we review the therapeutic
strategies currently under development or as future approaches designed
to normalize these pathways, which may then boost brain resilience to
cope with toxic protein species. Therefore, in addition to therapies
targeting the insoluble protein aggregates specifically associated with
each neurodegenerative disorder, these novel pharmacological approaches
may be part of combined therapies designed to rescue brain function.Keywords: neurodegenerative disorders, synapse, dendrite,
autophagy, metabolism, neuroinflammation, gene expression.LIST OF ABBREVIATIONS
- AD: Alzheimer’s disease
- ADAM10: a disintegrin and metalloproteinase 10
- ALS: amyotrophic lateral sclerosis
- AMPK: AMP activated protein kinase
- APP: amyloid-β precursor protein
- AUTEN-67: mTOR-dependent modulator autophagy enhancer-67
- Aβ: Amyloid-β
- CNS: central nervous system
- CREB: cAMP response element-binding protein
- DDQ: methylphosphonate
- FDA: Food and Drug Administration
- FTD: frontotemporal dementia
- HAT: histone acetyltransferase
- HD: Huntington Disease
- HDACs: histone deacetylases
- iNs: induced neurons
- MCU: mitochondrial calcium uniporter
- Mdivi-1: mitochondrial division inhibitor 1
- Mdivi-1: mitochondrial division inhibitor 1
- Mfn1: mitofusin-1
- Mfn2: mitofusin 2
- MS: Multiple sclerosis
- mTOR: mammalian target of rapamycin
- mTORC1: mTOR enzymatic complex1
- NF-κB: nuclear factor-κB
- NMDAR: NMDA receptors
- OMM: outer mitochondrial membrane
- OPA1: atrophy type 1
- OPC: oligodendrocyte progenitor cells
- OPTN: optineurin
- OXPHOS: oxidative phosphorylation
- PD: Parkinson’s disease
- PINK1: PTEN-induced protein kinase 1
- PKC: protein kinase C
- PKM: pyruvate kinase M
- RNF10: RING Finger protein 10
- ROS: reactive oxygen species
- SASP: senescent associated secretory phenotype
- SQSTM/p62: sequestrosome-1
- TARDBP: (TAR)-DNA-binding protein
Ageing and neurodegenerative diseases
In the last century, advances in medical care and the creation of
healthier environments contributed to an increase in life expectancy .
Given that advanced age is the main risk factor for neurodegenerative
diseases , elderly population growth leads to a significant increase in
the number of patients affected by age-related primary neurodegenerative
diseases, such as Alzheimer’s disease (AD) , Parkinson’s disease (PD) or
amyotrophic lateral sclerosis (ALS) . Therefore, considering the current
demographic changes, primary neurodegenerative diseases will have
substantial socioeconomic implications for healthcare systems, due to
their large costs and severe impact on the quality of life of affected
individuals and caregivers, posing a critical social emergency. Hence,
addressing these large burdens for the society requires an intensified
research and novel approaches and solutions .
In the last 20 years, substantial advances in the comprehension of
neurodegenerative disorder pathogenesis have been made . Much of this
progress is the result of biochemical and histochemical characterization
of proteins that accumulate within various inclusions in the diseased
brain and genetic linkage studies identifying mutations in genes that
cause neurodegenerative diseases.
The identification of specific, disease-segregating mutations in
previously unknown genes directed the attention to proteins and pathways
that are now considered crucial in the pathogenesis of neurodegenerative
diseases. For instance, certain pathogenic mutations in the gene coding
for the amyloid-β precursor protein (APP) cause AD, in the α-synuclein
gene are related to PD, in huntingtin to Huntington Disease (HD), or in
microtubule-associated protein tau are associated to frontotemporal
dementia (FTD) with parkinsonism . The accumulation of species derived
from these proteins in the brain of patients often represents a
histological hallmark for each specific neurodegenerative disorder
(Table 1).
Despite the presence of these inherited cases, most neurodegenerative
disorders develop sporadically in the absence of any known genetic
etiology. The onset of these sporadic forms is significantly influenced
by risk factors , ageing being the one with the highest impact . Hence,
age-associated brain modifications are considered key contributors to
the pathogenesis of neurodegenerative disorders . However, mechanistic
interface between brain ageing and neurodegeneration have not been fully
elucidated. During the 2000s the ageing research field has grown
considerably ; as understanding how exactly ageing increases the risk to
develop neurodegenerative diseases can provide important clues for the
development of new therapeutic strategies for neurodegeneration
treatment.
Even though protein misfolding and the accumulation and formation of
toxic species of proteins, due to inadequate folding, have been seen as
causative events in neurodegenerative disorders, in this review we
carefully examine the role of other, different, biological pathways that
are altered during ageing and implicated in the pathogenesis and
progression of neurodegenerative disorders. We focused on the mechanisms
controlling neuronal structure, gene expression, autophagy system, cell
metabolism, and, finally, neuroinflammation (Figure 1). Further, we
summarize the therapeutic approaches developed to restore these pathways
that may elevate the resilience of the brain to cope with toxic species
for each neurodegenerative disorder (Table 2).
The structural disintegration: how to reshape neurons and
synapses
The synapse is the biological locus responsible for the transmission of
information between neurons. Neuron-to-neuron synapses are composed of a
pre-synaptic and post-synaptic compartment, each with unique proteins
and structures to facilitate excitatory and inhibitory
neurotransmission.
The majority of synapses are found on dendrites, branch-like extensions
of a neuron that receive information from other neurons and carry it to
the neuron soma. The excitatory postsynaptic machinery is localized in
dendritic spines, small protrusions from the dendrite shaft. Dendrites
can support information processing at multiple spatial scales to
integrate synaptic signals finally transformed into action potentials .
Neuronal synaptic structures are not static but highly dynamic. The
capability of neurons to modify the efficacy of synaptic transmission
and the synaptic structure in response to different stimuli is called
synaptic plasticity . Synaptic plasticity has been proposed to play a
central role in the brain’s capacity to incorporate transient
experiences into persistent memory traces.
Astrocytes and microglia can transmit information and modulate synaptic
activity . Astrocyte processes encapsulate the synaptic cleft and ensure
recycling of released neurotransmitters, release co-factors important
for physiological neuronal transmission, and maintain tissue ion
homeostasis. Furthermore, astrocytes are connected via
gap-junction-coupled networks that synchronize neuronal activity within
brain regions . Microglia, the brain resident immune cells, on the other
hand phagocytose inactive synapses and release co-factors that are
important for the induction and maintenance of synaptic plasticity .
In addition to microglia and astrocytes, myelin, a passive insulating
layer formed by oligodendrocytes that ensures fast-saltatory conduction
of action potentials, is also essential for neuroprotection providing
physical axonal protection and trophic support. Despite being considered
for long a static component of the central nervous system (CNS), it has
now been demonstrated that myelin has a plastic nature, and that myelin
plasticity is required for motor learning and fear memory and
conditioning .
The impact of ageing and neurodegenerative disorders on the
structure of brain cells
Several studies documented changes in the molecular signature, the
morphology and function of brain cells with ageing. The principal
age-related neuronal structural alteration involves a reduction in
dendrites length and number, with a loss of various dendritic spines .
On the contrary, astrocytes undergo an increased expression of
cytoskeletal proteins, cell body hypertrophy, and a reduction in the
number of long, slender processes with ageing . Furthermore, aged
microglial cells show a gradual decrease in function, most notably in
chemotactic and phagocytic capacity. In particular, mouse studies have
documented impairments in the ability of ageing microglia to phagocytose
Amyloid-β (Aβ) fibrils and myelin debris
In addition, recent studies have revealed that myelin remodelling
persists throughout the lifespan . However, white matter and thus myelin
volume is shown to decline after 13 months in mice, and in humans, after
the age of 44-47 with myelin alterations that contribute to age-linked
functional decline being detected prior to neuronal loss. These
alterations include widespread and diffuse myelin breakdown,
degeneration, and reduced myelin renewal , decreased myelin stability
associated with lipid peroxidation formation of splits containing
cytoplasm and myelin balloons or spheroids, accumulation of myelin
debris, like multilamellar myelin fragments .
The age-dependent altered function of glial cells reduces their ability
to homeostatically nurture, protect and regenerate neurons, generating a
more inflammatory microenvironment that consequently promotes neuron and
synapse loss and, thereby, neurodegeneration. Most of these alterations
are mild in healthy ageing, but exaggerate in a range of
neurodegenerative diseases, such as AD, PD, ALS, HD, where they
contribute or accelerate neurodegeneration, facilitate protein
aggregates deposition, impair cognition and motor function by disrupting
connective pathways
Neurodegenerative diseases are characterized by abnormalities in
dendritic structure and synapse loss in different brain regions
depending on the neurodegenerative diseases . In HD, for example,
synapse loss is mainly detected in the striatal brain region, which is
linked with progressive movement discoordination . There is growing
evidence from ALS patients, FTD patients, and animal models that suggest
synaptic dysfunction and alterations in dendritic branching begins very
early in the disease before symptom onset and motor neuron death .
In AD neurons, the dendritic tree undergoes fast decline with a decrease
in the number of dendritic shafts while the few remaining show fewer and
shorter branches . Furthermore, synaptic loss in the hippocampus and
neocortex is known to be an early process in AD and the main structural
correlate with AD cognitive dysfunction.
In AD and PD, the oligomeric forms of Aβ and α-synuclein are the most
toxic species for synapses. The oligomers of α-synuclein but not its
fibrils contribute significantly to dopaminergic loss and neuronal cell
death . Similarly, it has been shown that the Aβ oligomers cause
synaptic loss and impair the mechanisms of synaptic plasticity .
Interestingly, APP and the enzymes implicated in the amyloid cascade are
synaptic elements located both, at pre and post synaptic side, and play
a critical role in regulating synaptic function . For instance, a
disintegrin and metalloproteinase 10 (ADAM10), the metalloprotease that
prevents Aβ generation, is a shedding enzyme that cleaves adhesion
molecules, such as N-Cadherin, and shapes spine morphology .
Furthermore, ADAM10 synaptic localization and activity are finely tuned
by synaptic plasticity phenomena and its synaptic abundance and activity
towards APP are affected in the hippocampus of AD patients . This is the
result of an impairment of ADAM10 local forward trafficking, that
depends on the PKC-regulated association with SAP97 and is also related
to an increase in ADAM10 endocytosis . Notably, alterations in ADAM10
have been described also in HD, with increased levels of the mature form
of ADAM10 in the brain areas that predominantly degenerate in HD have
been reported in mouse models of HD and in human HD brain samples .
Accumulation of active ADAM10 at the postsynaptic compartment leads to
increased proteolysis of N-Cadherin, likely promoting synaptic
instability in HD. These data confirm that synaptic failure is a common
trait of neurodegenerative disorders but highlight the importance of
investigating the molecular mechanisms underlying synaptic dysfunction
in each disorder. Therefore, given that synapses are the most vulnerable
regions of neurons, differences among synapse structure, metabolism and
signaling mechanisms might be determinants of neuronal vulnerability in
the different neurodegenerative disorders.
Therapeutic approaches to tackle synaptic and neuronal
dysfunction
Although we currently lack therapeutic approaches aimed at enhancing
myelination in the clinic, recent advances and high-throughput screening
approaches have provided us with potential pro-myelinating compounds.
Clemastine fumarate, a muscarinic receptor antagonist that was
identified in a pro-remyelinating drug screening, has met clinically
defined efficacy endpoints in a clinical trial in patients with Multiple
sclerosis (MS) . Recent evidence has also shown that Clemastine prevents
age-related myelin loss, neurodegeneration, and cognitive decline in
healthy ageing as well as in a mouse model of AD . Additionally, other
drugs that have been shown to promote myelin repair in the context of
MS, such as metformin or LY294002 , could also be beneficial to prevent
myelin breakdown and degeneration with age or in other neurodegenerative
diseases, but their efficacy in this context is yet to be investigated.
As neuronal connections represent the hardware for proper cognitive
abilities, therapeutic strategies aimed at preserving dendritic and
synaptic connections could conceivably be useful in neurodegenerations.
The process of neurite repair to replenish the degenerated dendrites
would involve regrowth and rewiring of the new connections within the
network. However, the local molecular and cellular milieu in the CNS
opposes neurite growth and thus renders this approach particularly
challenging . A better option could be to use strategies aimed at
upholding neuronal dendritic integrity rather than promoting its
regrowth. In this view, promising findings showed that specifically
preserving dendritic architecture in mouse models of acute
neurodegeneration (i.e. stroke) counteracted the loss of neurons which
is typically accelerated by disrupted connectivity and, ultimately,
resulted beneficial also at a functional level .
Regarding synaptic dysfunction, the 2022 drug pipeline for AD showed
that synaptic plasticity/ neuroprotective agents for which phase 2 and
phase 3 clinical trials are currently ongoing are 17% and 19% of all
disease-modifying therapies respectively, indicating that significant
efforts are being made for targeting these mechanisms .
A systematic review analysed the efficacy profile of 12 published
results of clinical trials investigating the safety and efficacy of
disease-modifying drugs targeting synaptic plasticity in dementia .
However, only three molecules (Levetiracetam, Bryostatin 1 and
Masitinib) show promising results.
Levetiracetam is a second-generation antiepileptic drug approved as an
adjunct therapy for partial seizures. The mechanism of action seems to
involve neuronal binding to the synaptic vesicle protein 2 A, inhibiting
the release of calcium from intraneuronal deposits, opposing the
activity of negative modulators of GABA and glycine-dependent currents,
and inhibiting excessive synchronized activity between neurons . An
important role of this molecule also appears to be linked to synaptic
plasticity. The administration of Levetiracetam to AD mice was reported
to reverse synaptic dysfunction . The initial clinical trials with
Levetiracetam showed limited results because of the high clinical
heterogeneity of the enrolled cohort. On the other hand, a recent
clinical trial found that Levetiracetam was able to significantly
improve cognitive status only in patients with cortical
hyperexcitability. This suggests that preselection of AD patients
presenting symptoms ranging from subclinical epileptiform activity to
seizures and network hyperexcitability could improve the capacity to
identify therapeutic effects of Levetiracetam.
Bryostatin 1 is a macrocyclic lactone that activates protein kinase C
(PKC) and, thereby, regulates neurogenesis, axonal transport, and
synaptic plasticity . The administration of Bryostatin 1 in AD mice
restored the number of dendritic spines in the hippocampal CA1 area .
The results of phase 1 and 2 trials were conflicting, while a recent
pooled analyses from 2 randomized clinical trials confirmed a
significant cognitive restoration elicited by Bryostatin 1 in the
absence of a Memantine treatment .
Masitinib, a tyrosine kinase inhibitor, is usually used in the treatment
of mast cell tumours in animals. Studies in animal models showed that
Masitinib administration has a protective effect on synapses because of
mast cell inhibition and reduction of the secretion of specific
mediators potentially toxic to synapses . Initial studies in patients
reported that participants treated with Masitinib showed significant
improvement in cognitive function . Thus, Masitinib is being currently
investigated in a multicentre phase 3 trial for patients with
mild-to-moderate AD (NCT01872598) and showed some beneficial effects in
ALS .
Pharmacological treatments of ALS patients aim at counteracting
glutamate excitotoxicity. Riluzole, an inhibitor of glutamate release,
was approved by the United States Food and Drug Administration (FDA) in
1995. Despite being associated with a short survival benefit of 2–3
months , the subsequent adoption of riluzole as a treatment for ALS was
perhaps reflective of the need for therapeutic options in the face of
this devastatingly progressive disease . Remarkably, findings from
several open-label non-randomised trials have suggested that the
greatest benefit occurs at earlier disease stages . Therefore, is
possible that riluzole’s therapeutic benefit is likely to affect or
activate different cellular pathways depending on the disease stage .
Another molecule investigated to counteract excitoxicity is the
antibiotic Ceftriaxone that causes the upregulation of glutamate
transporter and decreases glutamate-induced toxicity . Phase-3 trial of
ceftriaxone indicated an overall increase in survival of patients with
ALS.
Overall, these strategies tackle synaptic failure and dysfunction as
common molecular mechanism across neurodegenerative disorders. Next
challenge for drug discovery research will be to design tools and
molecules specifically targeting the mechanisms underlying synaptic
dysfunction or dendrite degeneration in each of the different
neurodegenerative disorders. An example is the control of the
trafficking of ADAM10. As mentioned above, alterations in ADAM10
synaptic localization and activity are implicated both in AD and in HD.
In AD however, ADAM10 levels are reduced in hippocampal synapses while
active ADAM10 is overexpressed in HD striatal synapses. Therefore, the
strategy to counteract synaptic failure in AD takes advantage of the
administration of a cell permeable peptide that blocks ADAM10
endocytosis, upregulates its activity, restores synaptic function
without affecting plaque deposition and rescues cognitive defects in AD
mice . On the other hand, the use of a peptide designed to inhibit
SAP97-mediated ADAM10 trafficking to the synapse , normalizes ADAM10
activity and rescues cognitive deficits in HD mice .
Therefore, understanding the mechanisms implicated in synaptic failure
in the different neurodegenerative disorders is required to design
disease-tailored therapeutic strategies.
The main players controlling gene expression during ageing and
in neurodegenerative disorders
A key aspect sustaining and enabling various forms of plasticity is the
regulation of gene expression. Basal transcription ensures the mere
survival of neurons as cells; to be functional computational units,
neurons further need to adapt their transcriptional responses to
physiological and pathological cues. To do so, neurons employ an array
or regulatory elements, epigenetic mechanisms, and transcription
factors, all of which are modulated by diverse stimuli. One of the most
prominent stimuli that neurons are specialized to adapt to is synaptic
activity and calcium signals. Among the many epigenetic mechanisms
employed by neurons, the ones influencing chromatin accessibility -DNA
methylation and post translational modifications of histone proteins -
received the most attention and are the best characterized. However,
more recently, non-coding RNAs have received increasing attention.
Given its key role in ensuring neural functions, it comes as no surprise
that a long list of alterations in the transcriptional landscape or
dysfunctions of molecular players have been associated with
neurodegeneration. Mechanistically, the role of transcription factors or
epigenetic regulators in the pathogenesis of neurodegenerative
conditions is explained by their capability to affect genes directly
involved in the pathology or in mediating co-morbidities. Here, we
highlight some of the better studied transcription factors and
epigenetic regulators and their involvement in neurodegeneration.
cAMP response element-binding protein (CREB) is a ubiquitous
transcription factor playing multiple roles in the CNS. Several
signaling cascade converge on CREB which acts in cooperation with
cofactors such as CBP/p300. CREB is fundamental for memory and learning
but also essential to neuronal survival and protection . Due to its
broad expression, being downstream of different signaling events and
driving expression of a myriad of critical neuronal genes, alterations
of CREB activity and/or expression CREB has been indeed reported for
neurodegenerative conditions such as AD or PD both in animal models and
humans .
Not all transcription factors are always localized to the nucleus, a
considerable number of them is instead regulate their expression by
subcellular localization. Thus, failures in the nucleo-cytoplasmic
shuttling of transcription factors represent another possibility leading
to transcriptional dysfunction in neurodegenerative conditions. The
first transcription factor for which movement between the cytosol and
the nucleus was described was nuclear factor-κB (NF-κB). Activity of
NF-κB is prevented when sequestered in the cytosol. Alterations of NF-κB
localization have been observed in the proximity of plaques in
post-mortem samples from AD patients but also in dopaminergic neurons of
PD patients and are tightly linked to inflammatory states.
Furthermore, synapse to nucleus shuttling of proteins with the capacity
to modulate gene transcription is a prominent way in which inputs
received at the synapse are transferred to the nucleus to implement
long-term changes. Synapses contain several nuclear localization
signals-containing cargo proteins and different components of the
nuclear import machinery, like importin-α and importin-β, which have
been shown to translocate to the nucleus in an activity-dependent manner
. In the last decade, few synaptonuclear protein messengers (i.e. Abi-
1, AIDA-1D, Jacob, RNF10) have been identified, and shown to play key
roles in plasticity and synapse function . Notably the activation of
NMDA receptors (NMDAR) can regulate gene transcription , thus affecting
global protein synthesis and, thereby memory formation. Such effect of
NMDAR activation requires the long-distance trafficking of
synaptonuclear proteins. The synaptonuclear messengers can associate
with different classes of receptors and specifically translate their
activation in gene expression changes. For instance, the RING Finger
protein 10 (RNF10) operates as a mobile hub that docks GluN2A-containing
NMDAR derived signalosomes to nuclear target sites , while protein
messenger such as Jacob can encode the synaptic and extrasynaptic origin
of NMDAR signals, following long-distance transport and nuclear import .
Remarkably, alterations in synaptonuclear messengers have been reported
in neurodegenerative disorders such as AD .
From the epigenetic perspective, reduced global methylation levels have
been found in the blood of AD and PD patients in comparison to healthy
controls, probably due to the decrease in expression of DNA
methyltransferase 3a , a key mediator of this epigenetic mark in the
context of signal-regulated neuroepigenetics . The scenario is however
unclear as studies derived from post-mortem samples of AD patients in
some cases reported a reduction of global methylation levels in cortex
and hippocampus , while in other studies lack changes or even an
increase in global DNA methylation have been observed. Studies focusing
on DNA methylation changes at specific genomic regions, functional
elements, and individual gene loci have yielded a more comprehensive
view in the context of AD, but also revealed that epigenetic changes are
complex, and often contradictory results are reported from different
models (reviewed in ). Similarly, studies have indeed reported an
association between PD disease progression and global methylation levels
detected in brain or blood or changes in the methylation pattern of
specific genes .
Given the prominent role that histone deacetylases (HDACs) play in the
modulation of several neuronal functions, it comes as no surprise that
they have also been implicated in neurodegeneration. These heterogenous
group of proteins, which are classified based on their activity,
structure and co-factors, are responsible for the removal of the acetyl
group from histone as well as non-histone proteins. An example of their
involvement in neuropathologies comes from HDAC2, HDAC6 or HDAC5, whose
levels are elevated in brain areas of post-mortem AD patients .
Interestingly, an increased association between HDAC1 and CREB, possibly
facilitating CREB pathological dephosphorylation, was observed in
neuronal samples of PD patients . Expression levels of HDAC4, which has
been implicated in many forms of pathologies of the nervous systems in
animal models , was shown to be strongly associated with rapid
progression of ALS in patients . Alteration in the expression level of
HDAC11 and HDAC2 were also reported in in post-mortem brain and spinal
cord tissue of ALS patients . Beside their action as transcriptional
regulators, HDACs act also on cytosolic proteins of which the most
prominent is tubulin. For example, HDAC6 acts on tubulin and has been
associated to deficits in axonal transport in ALS patients-derived motor
neurons.
Therapeutic approaches to modify gene expression in
neurodegenerative disorders
Targeting transcriptional regulators might be beneficial against
neurodegenerative conditions as this would result in a widespread
modulation of many affected downstream processes. However, this aspect
also represents a potential caveat as affecting the transcription of
several genes might be detrimental as not all of them necessarily
participate in the pathogenesis. Nevertheless, efforts have been made in
exploring, designing, and testing of therapeutical approaches aiming at
modulation of transcription-related processes.
One of the most sought-after targets in the treatment of
neurodegenerative disorders is CREB due to the copious evidence showing
a reduction in its functionality or expression in many diseases. At
present, however, there are no available drugs specifically boosting
CREB-dependent signaling. Multiple strategies could be followed, from
acting on its upstream signaling regulators to using molecular biology
or genetic approaches to restore its expression levels.
The complexity and multifactorial nature of many neurodegenerative
disorders is likely to push the development of drugs towards targets
different than the prototypical players associated with a certain
disorder. A case for this is NF-κB in the treatment of AD. Memantine, an
FDA-approved molecule for the treatment of AD seems to contrast the
pro-inflammatory activity of NF-κB. Additional anti-inflammatory drugs
are under preclinical or clinical evaluation for their capacity to
interfere with NF-κB (see for review .
Great efforts have been spent in the exploration of HDACs inhibitors as
useful drugs in neurodegeneration. HDACs inhibitors may broadly
facilitate gene expression via rendering the chromatin more permissive
for transcription. In animal models, non-selective HDAC inhibitors
successfully restored memory function and neuronal structural
aberrations . However, care should be taken to follow the path of
non-selective inhibition due to its potential side effects . The design
and development of specific HDAC inhibitors has been hindered by
different problems including the fact that all HDACs share considerable
structural similarities and are expressed across different organs and
cell types. Nevertheless, with the advent of better technological
opportunities, is it quite likely that specific HDAC inhibition may be a
viable therapeutic avenue to pursue . An encouraging example comes from
inhibition of HDAC6 via Tubastatin A, ACY-1215, MPT0G211, or
5-Aroylindoles which showed promising results in AD animal models and
are currently under clinical trial evaluation for diseases other than
neurodegeneration.
Autophagy: when the clearance system fails and drives
neurodegeneration
Cellular homeostasis is the process that controls different cellular
activities. It is a fundamental condition that allow the cell to
maintain the physiological balance in term of cellular identity,
resilience, and survival. Defects in protein homeostasis contribute to
the lack of intracellular proteins and organelles, which progressively
accumulate in the cytoplasm. Therefore, such alterations can be
implicated in the onset and progression of neurodegenerative disorders .
Cellular component degradation within the cell can follow two different
pathways: autophagy and the ubiquitin system, specific for protein
degradation. Both processes require a concerted action of different
proteins that are recruited and specifically recognize the damaged
material, driving protein degradation as the last step to maintain
cellular homeostasis. Under stress conditions, such as ageing or
disease, this tuned process fails, resulting in an engulfment in cell
physiology.
In mammalian cells there are three types of autophagy: macroautophagy,
microautophagy and chaperone-mediated autophagy. Macroautophagy, simply
known as autophagy, plays the major role in maintaining the cellular
homeostasis because it helps in the removal of bulky protein aggregates
and bigger cytoplasmic bodies. It begins with the phagophore formation
that entraps the misfolded proteins and, after different steps, finally
fuses with lysosomes to generate the autophagolysosome . Autophagy is
tightly controlled by mammalian target of rapamycin (mTOR) and AMP
activated protein kinase (AMPK). Autophagy is promoted by AMPK, which is
a key energy sensor and regulates cellular metabolism to maintain energy
homeostasis . Conversely, autophagy is inhibited by mTOR, a central
cell-growth regulator that integrates growth factor and nutrient signals
.
Given its crucial role in cell homeostasis, a defect in autophagy is
associated with neuronal loss and cognitive decline, both in
physiological conditions, such as ageing, but also in neurodegenerative
disease . Interestingly, defect in the autophagy machinery have also
been linked to axonal and dendritic degeneration and might thereby
further promote the dysfunction of neural network dysfunctions .
Several neurodegenerative diseases are characterized by defects in the
degradation process of misfolded proteins and thus, abnormal protein
aggregation. Autophagy failure can be attributed to the impairment in
the clearance of pathological protein such as α-synuclein, Aβ and tau .
For instance, Beclin1 is a protein involved in the regulation of
autophagy and has been shown to be reduced in AD patients . Furthermore,
it has been shown that the downregulation of Beclin-1 in mice resulted
in reduced neuronal autophagy and Aβ accumulation .
Moreover, the most common autosomal-dominant form of PD and a familial
variant that closely resembles sporadic PD is associated to LRRK2
mutation . The altered function of mutated LRRK2 has been linked to
defects in endosomal-lysosomal trafficking and chaperone-mediated
autophagy . In dopaminergic neurons, the lysosome number has been
reported to be depleted in a mouse model of PD .
In addition to autophagy, cells take advantage of mitophagy, a specific
process responsible for the selective degradation of dysfunctional
mitochondria in PD . The PTEN-induced protein kinase 1 (PINK1),
localized on the external mitochondrial membrane, phosphorylates
mitofusin 2 (Mfn2) and ubiquitin triggering the recruitment of the
Parkin protein. This event activates several ubiquitin-binding proteins
such as optineurin (OPTN) and sequestrosome-1 (SQSTM/p62), that initiate
mitochondria to the pathway of mitophagy. Defects in the mitophagy
machinery are also a pathological pathway in PD and the accumulation of
damaged mitochondria represents one of the main pathogenetic alterations
. The PD-associated familial autosomal recessive mutations in the genes
PINK1 and Parkin have been discovered to have a key role in
mitochondrial quality control . The phosphorylation of PINK1 at the
outer mitochondrial membrane (OMM) leads to the recruitment of parkin.
Once active, parkin allows the synthesis of ubiquitin chains on OMM
proteins leading to ubiquitin chain assembly. The mutations in the PINK1
(PARK6) and parkin (PARK2) genes are linked to autosomal recessive
early-onset PD and a pathological accumulation on the OMM that triggers
an abnormal mitophagy . In PD α-synuclein, the main component of those
pathological Lewy bodies, has been found to bind mitochondria components
, inhibiting the protein import and leading to an impaired cellular
respiration. In particular, α-synuclein interacts with and disrupts
mitochondrial proteins such as TOM20, VDAC, and F1Fo-ATP
synthase, causing mitochondrial metabolic impairment .
Targeting autophagy: how to promote protein and organelle
clearance
In the last two decades, several studies have reported the
neuroprotective activity of rapamycin, one of the most powerful
pro-autophagy agents, in both cellular and animal models of
neurodegenerative diseases and, with some limitations, in human .
Rapamycin acts blocking the kinase activity of mTOR enzymatic complex1
(mTORC1), removing its autophagy suppressor activity observed under
physiological conditions. The strategy of the rapamycin treatment is to
activate the autophagic flux negatively controlled by mTORC1.
Interestingly, rapamycin has been proposed as an anti-ageing drug in
mice, since it was reported to increase the lifespan of animal models
treated .
However, rapamycin treatment has been also tested in animal models of
AD. It has been shown that rapamycin administration reduced the
accumulation of Aβ aggregates and prevented tau phosphorylation in the
brain of AD transgenic mice, showing a global impact in maintaining
cognition .
Regarding the use of rapamycin in humans, some encouraging results have
been reported in the ALS field where a recent, randomized,
placebo-controlled, phase II clinical trial was started to evaluate the
efficacy of rapamycin in patients affected by ALS .
Another approach to modulate the mTOR signaling level is possible using
mTOR-dependent modulator autophagy enhancer-67 (AUTEN-67), a small
molecule identified as a potent candidate with anti-ageing and
neuroprotective effects, by significantly increasing autophagic flux in
neurons and protecting them from undergoing stress-induced cell death .
Other agents that can indirectly trigger AMPK-dependent mTOR
inactivation are metformin and resveratrol. Metformin activates AMPK
which in turn promotes autophagy blocking mTORC1 activity through direct
inhibitions of regulatory associated protein of mTOR. Moreover, the
activation of AMPK directly promotes the activation of
phagophore-forming enzymatic complex unc-51-like kinase (Ulk1)1/2, which
is considered the initiator of the autophagic cascade.
Furthermore, PROteolysis TArgeting Chimeras (PROTAC) technology has
emerged as one of the most promising approaches to remove specific
disease-associated proteins using the autophagy machinery of the cells.
In the last few years, the PROTAC approach has been extensively used,
with several PROTACs molecules currently in clinical trials in the
cancer field . A PROTAC molecule targeting tau protein has also been
developed. It is a chimera construct made of a tau-binding peptide, a
linker, a VHL-binding peptide, and a cell-penetrating peptide.
Interestingly, this molecule leads to a significant degradation of tau
and reduced neurotoxicity of Aβ, highlighting the therapeutic potential
of this approach .
Metabolic failure and energy crisis of brain cells
Among the different brain resident cell types, neurons are extremely
energy demanding. Neurons rely almost exclusively on the mitochondrial
oxidative phosphorylation (OXPHOS) system to fulfill their energy needs
through ATP. The OXPHOS-mediated mitochondrial functions are diverse,
ranging from the cell-intrinsic energy production to the regulation of
intracellular calcium homeostasis, synaptic plasticity, and
neurotransmitter synthesis . On the other hand, this important energy
production is accompanied by the formation of reactive oxygen species
(ROS), which in excess are detrimental for cells.
ROS derive mainly from the process of the OXPHOS, which reduces
O2 into H2O using the electrons flux
deriving from the respiratory chain and leading to the formation of
superoxide anion radicals of the oxygen. Even if cell-intrinsic
antioxidant defense systems can buffer ROS, when this buffering systems
is overloaded and cell-homeostasis altered, ROS become toxic. Given the
fundamental role of mitochondria in neuronal energy supply, their
dysfunction leads to an impairment of basal neuronal energy source,
impacting on several aspects of brain physiology. In addition to their
metabolic activity, mitochondria play a key role in cellular calcium
homeostasis. Cellular calcium concentration is strictly regulated as it
sustains vital aspects of the neurons such as secretion, motility,
metabolic regulation, synaptic plasticity, proliferation, gene
expression, and apoptosis . Therefore, mitochondrial calcium
dysregulation contributes to neurodegeneration since it is the major
mechanism by which increased excitatory neurotransmission triggers
mitochondrial depletion from and retraction of dendritic structures .
The function of mitochondria is strictly related to their structure and
dynamics. Thus, the mitochondrial efficiency is measured through their
capacity to undergo continue fusion and fission cycles. Hence,
mitochondrial dynamism is important for their morphology and function,
and therefore, relevant for neuronal viability and synaptic activity.
The equilibrium between fission and fusion is key for adequate
mitochondrial function and is compromised in different neurodegenerative
disorders. Drp1, the GTPase that controls the process of fission, is
altered in AD, leading to an excessive mitochondria fragmentation and,
thereby, altering their function . This peculiar phenotype has been
shown in neuronal cultures upon Aβ exposure as well as in several
neurodegenerative disease animal models . Defects in the dynamin-related
GTPase proteins mitofusin-1 (Mfn1) and Mfn2, and atrophy type 1 (OPA1)
protein cause mitochondrial fusion alterations and have been reported in
various neurodegenerative disorders, including AD .
Failure in mitochondria trafficking, function and positioning in
dendrites and synapses have been also observed in ALS and FTD and may
contribute to early synaptic loss in disease .
In FTD, several pathways controlling mitochondrial trafficking,
dynamics, and consequently activity are altered . In the genetic FTD
caused by MAPT mutations, a decrease in mitochondria-Tau interactions in
iPSC-derived neurons has been observed. The authors reported that
neurons expressing the Tau protein carrying the V337M mutation were
characterized by alterations in mitochondria bioenergetics affecting
efficiency in maintaining ATP levels under prolonged energetic stress.
Also, Tau P301L mutation, which is known to cause Tau
hyperphosphorylation, decreases mitochondrial respiration and ATP
production, leading to a global mitochondrial and oxidative impairment .
Mutations in transactive response (TAR)-DNA-binding protein (TARDBP),
coding for the TDP-43 protein, are also associated with FTD.
Interestingly, TDP-43 inclusions aggregate outside of the nuclear
compartment and can directly affect the dynamics and trafficking of
mitochondria, both at the axonal and dendritic compartment, leading to
functional impairment. The overexpression or reduction of TDP-43, in
different animal model systems, leads to a mitochondria dysfunction
In general, an altered cellular metabolism is considered a hallmark of
ageing . During cellular senescence, which is distinct from, but
associated with biological aging, the mitochondria exhibit numerous
changes in their structure, dynamics, and function. Therefore, in
senescent cells a decrease in mitochondrial membrane potential, an
increase in proton leakage and a lowered oxidative capacity has been
observed. The consequence of modified processes resides in an altered
aged metabolic homeostasis, with a significant increase in ROS
generation, diminished antioxidant defense and a decrease in ATP
production. All these phenomena have a great impact on neurons,
post-mitotic cells that are particularly sensitive to stress and to the
accumulation of a senescent profile typical of ageing . In addition, the
pool of healthy mitochondria tends to decrease with ageing.
In addition to the role played by mitochondria in neurons themselves,
the demanding energy request by neurons is also sustained by glial
cells, which are extremely flexible and respond to environmental changes
providing neurons the required energy . Every time that neurons need
energy to perform the highly energy consuming neuronal synaptic burst,
the so-called astrocyte-neuron lactate shuttle responds to this energy
demand by creating an energy bridge by which astrocyte produced lactate
is received by the neuronal part . The astrocytic-neuronal metabolic
bridge is supported by the capacity of astrocytes to convert GABA and
glutamate, removed from the synaptic cleft, into glutamine, which is
used as a precursor for refill synaptic vesicles or for phosphorylation
via the TCA cycle
Neuronal metabolism pathways are comparatively inflexible and believed
to be strictly regulated. However, metabolic changes including state
shifts and alteration is individual metabolites can have great impacts
on the neuronal epigenome. This enables the cells to adapt to
environmental changes, but also poses a risk for, as energetic
challenges may lead to highly consequential epigenetic alterations .
Indeed, the cell fate specification and the consequent cell identity are
established by a highly specific epigenetic control which must be also
plastic to allow the cell to adapt to the environment. For instance,
high glucose levels produce high acetylCoA:CoA ratio, which regulates
histone acetyltransferase (HAT) activity, and contributes to increased
chromatin accessibility and gene activation . Both the early TCA cycle
intermediate α-ketoglutarate and oxygen are co-substrates for
demethylases, affecting DNA and histone methylation and, thereby,
changing transcription scenario . Moreover, metabolic enzymes can
translocate directly to the nucleus in a splicing- and
signaling-dependent manner, and act directly on histones triggering
changes in transcription. This is the case of pyruvate kinase M (PKM)
which translocates to the nucleus where it phosphorylates histone 3 and
leads to a de-repression of cell-cycle and glycolytic genes . In
directly converted induced neurons (iNs) from AD-patient-derived
fibroblasts, a cancer-like metabolic switch from neuronal OXPHOS to
aerobic glycolysis in AD iNs is associated with a higher level of the
PKM2 nuclear isoform compared to the physiological PKM1. PKM2 prevalence
is associated with metabolic and transcriptional changes in AD iNs,
contributing to AD-related-neuronal defects. Overall, all these new
findings are suggestive of the presence of a metabolic reprogramming
towards an aerobic glycolytic profile in AD, throughout a Warburg-effect
.
The epigenetic modulation of metabolism, influenced by the combination
of pathology and ageing, could be primary or secondary to the
mitochondrial impairment . In general, in the neurodegenerative context,
there is an accumulation of macromolecular damage and metabolic
reprogramming that leads to the damage of organelles, including
mitochondria, and eventually to tissue dysfunction .
Targeting mitochondria as therapeutic strategy for treating
neurodegeneration
To reduce and buffer mitochondrial dysfunction the most used indirect
therapies rely on the use of antioxidants that mitigate the
mitochondrial ROS production. Some of those compounds include the
lipophilic MitoQ, CoQ10, MitoVitE, MitoTEMPOL , and resveratrol, which
indirectly activates PGC-1α and induces mitochondrial biogenesis . There
are also compounds that modify mitochondrial dynamics, such as
mitochondrial division inhibitor 1 (Mdivi-1), and methylphosphonate
(DDQ) . In particular, Mdivi-1, an inhibitor of Dpr1, showed activity
against Aβ-induced excessive mitochondrial fragmentation . A novel
approach, recently proposed, suggest combining the antioxidant effect
with epigenetic modulation. Shikonin, an anti-cancer PKM2 modulator,
would act on the metabolic shift caused by neuronal PKM2 in AD, acting
as an apoptotic brake on mature .
Given the pathogenic role for excitatory mitochondrial calcium
dysregulation in mediating sublethal dendritic atrophy observed in
chronic neurodegenerative diseases, inhibiting calcium uptake has been
reported to be neuroprotective. The major protein complex involved in
mitochondrial calcium uptake is the mitochondrial calcium uniporter
(MCU) and, thereby, MCU inhibitors are neuroprotective in different
genetic models of chronic neurodegenerative diseases
Neuroinflammation
In recent years, it has become clear that despite having many different
primary causes, all neurodegenerative diseases share a common hallmark:
neuroinflammation. Inflammation is the first line in host pathogen
defence and essential to the body’s healing processes. However, chronic
or prolonged inflammation, like that observed in ageing and further
exacerbated in neurological diseases, is detrimental for tissue
homeostasis. Neuroinflammation can be triggered by CNS resident immune
and glial cells (e.g. microglia, astrocytes, oligodendrocyte lineage
cells), cells from the peripheral innate or adaptive immune system (e.g.
T cells, B cells, macrophages), meningeal inflammation, or
autoantibodies directed to the CNS.
Neuroinflammation in ageing and neurodegeneration
There is growing evidence showing that both, innate and adaptive immune
cells are present in the healthy CNS, where they have key roles in
maintaining homeostasis and immunosurveillance, being associated with
neurogenesis , learning and memory and synaptic pruning , among other
functions. However, this tightly regulated immune-CNS interaction is
distorted with ageing, and even more abruptly in neurodegenerative
disorders, leading to pathological neuroinflammation and subsequent
neurodegeneration . Even though present at low levels in the healthy
young CNS , an increase in adaptive immune cell infiltration, mainly
CD8+ T cells and to a lesser extent
CD4+ T cells, has been observed in the neurogenic
niches, the white matter, and the optic nerve with age. Enhanced T cell
infiltration alters CNS resident cell function increasing the expression
of interferon responsive genes in CNS stem and glial cells (neural stem
cells, microglia, and oligodendrocytes) and contributes to age-related
myelin degeneration, impaired neurogenesis, and axonal degeneration .
Increased CNS infiltration of peripheral immune cells with ageing, may
result from blood-brain-barrier alterations, increased permeability and
a decreased CNS perfusion and lymphatic drainage .
Similar changes in T cell infiltration have been also observed in a
range of neurodegenerative disorders not considered primary autoimmune
disorders such as AD, PD and ALS. Evidence has shown increased in T cell
numbers, especially CD8+ T cells, in the post-mortem
CNS tissue of AD, ALS and PD patients , and altered T cell levels or
subsets in the cerebrospinal fluid and peripheral blood AD, PD and ALS
patients . The role of adaptive immune cell-mediated inflammation in AD
remains controversial. Even if T cell depletion has rendered beneficial
roles reversing cognitive decline, increasing Aβ clearance and promoting
neuronal survival , other studies have described detrimental roles for T
cells in AD pathology . In PD models on the other hand, mice lacking
mature lymphocytes show attenuated dopaminergic cell , while in an ALS
mouse model CD8+ T cells infiltrate in the CNS are
associated with motor neuron loss . Thus, the role of T cell-mediated
neuroinflammation in neurodegenerative diseases may be subset and
context specific, highlighting the complexity of the CNS-immune
crosstalk and the role of neuroinflammation in neurodegeneration.
Prolonged CNS immune infiltration together with the enhanced production
of pro-inflammatory cytokines (e.g: IFN-γ, TNF-α, IL-6 or IL-1β)
described in both, ageing and neurodegenerative diseases, also
contribute to neurodegeneration indirectly, by perpetuating inflammation
through the priming of CNS glial cells. Single cell sequencing analysis
of CNS resident cells such as microglia, astrocytes and oligodendrocyte
lineage cells in different neurodegenerative diseases contexts and
ageing has unveiled disease-specific phenotypes characterized by the
expression of inflammatory and neurotoxic markers such as Clec7a,C3, Lgals3, Trem2 in microglia , Serpina3n, Lcn2, Ifitm3,
Timp1, Chi3l1 in astrocytes and Serpina3n, C4b, or Klk6 in
oligodendrocyte lineage cells . Beyond this disease-specific phenotype,
an elevated expression of interferon responsive/stimulatory genes (e.g:Irf1, Irf7, Irf8, Isg15, Ifit3) has also been described across
glial cells in ageing and neuroinflammation . Additionally,
neuroinflammation enhances the expression of antigen presenting genes
(e.g: Cd74, B2m, Cd9, H2-K1, H2-D1) as well as immune cell
chemoattractant cues such as Icam-1, Ccl2, Cxcl12 or Ccl3 by all
the main glial cells, which in turn further activate CNS infiltrating T
cells, contributing to a positive feedback loop that perpetuates
neuroinflammation and thus, neurodegeneration
In addition to immune cell infiltration and CNS resident cell-driven
pro-inflammatory reactions, another source of neuroinflammation is
linked to the accumulation of senescent cells with ageing and in CNS
pathology. Mounting evidence has demonstrated the accumulation of
senescence markers such as P16, P21, YH2A.X, lipofuscin, GATA4 and
high-mobility group box protein 1 in microglia, oligodendrocyte
progenitor cells, oligodendrocytes, astrocytes, and neurons with ageing
and in pathology such as AD . Senescent cells accumulate in aged tissues
and contribute to the pathogenesis of a range of neurodegenerative
diseases, at least in part, through their pro-inflammatory senescent
associated secretory phenotype (SASP) , which can propagate senescence
to neighbouring cells in a paracrine manner and contributes to immune
cell recruitment to eliminate senescent cells . Hence, neuroinflammation
and senescent cells establish an additional positive feedback loop
exacerbating ageing and disease pathogenesis: chronic inflammation like
that observed in ageing and neurodegenerative disorders enhances the
appearance of senescent cells, which in turn further contribute to
neuroinflammation by the secretion of pro-inflammatory molecules.
Therefore, it is plausible that either by eliminating senescent cells or
by modulating SASP, we can limit neuroinflammation and prevent
neurodegeneration.
Therapeutic approaches to counteract neuroinflammation
Several drugs that limit immune cell infiltration in the CNS have been
developed in the context of MS, like Siponimod, Fingolimod or
Natalizumab However, whether these drugs are beneficial in other primary
neurodegenerative diseases such as AD or ALS is still under debate.
Natalizumab is a monoclonal antibody that blocks the extravasation of
immune cells in the CNS and has been proven successful in mouse models
of ALS such as SOD1G93A and
TDP43A315T, where it has diminished astrocyte and
microglia priming, increasing motor neuron number and survival .
Natalizumab has also shown beneficial effects in pre-clinical models of
AD such as APP/PS1 and 3xTg mice. In APP/PS1 mice Natalizumab reduced of
proinflammatory cytokines in the spleen, CD4 immunoreactivity and
general inflammation in the CNS . Similarly, in 3xTg-AD mice,
Natalizumab has improved memory and reduced microgliosis, Aβ load and
tau hyperphosphorylation . Fingolimod on the other hand, is a structural
sphingosine analogue and sphingosine-1p-phosphate (S1P) receptor
modulator which blocks immune cell migration outside primary lymphoid
organs and thus, reduces T and B cell number in circulation. Fingolimod
has shown to promote survival and improve the phenotype in
SOD1G93A mice and is well tolerated by patients with
ALS, although its efficacy in disease progression is yet to be evaluated
. In AD on the other hand, fingolimod has shown to ameliorate Aβ
neurotoxicity in neuronal cultures while reducing Aβ and neuronal loss
and astrocyte and microglial activation and improving memory and
learning deficits in 5xFAD mice . Furthermore, in PD mouse models such
as the MPTP mouse model and a model performed by intracerebral
injections of 6-hydroxydopamine, fingolimod attenuated
neuroinflammation, neuronal loss and motor deficits . Moreover, low-dose
of Fingolimod improved motor function and reduced brain atrophy, leading
to the extended survival of R6/2 mice, a mouse model of HD . Thus, even
though drugs that limit immune cell trafficking to the CNS appear to
have a beneficial effect and help preventing neurodegeneration in
different mouse models, further pre-clinical investigations followed up
by clinical trials are needed before clearly establishing their benefits
for patients in other neurodegenerative diseases beyond MS.
An alternative approach to limit not only immune cell mediated
inflammation, but also neuroinflammation mediated by CNS resident cells,
involves the use of other less specific anti-inflammatory drugs such as
minocycline or non-steroidal anti-inflammatory compounds. Minocycline is
a broad-spectrum antibiotic with important anti-inflammatory properties
and as such, it has been studied for several years now in mouse models
of neurodegeneration. Even if minocycline has been proven successful in
limiting neuroinflammation and in some cases neurodegeneration in mouse
models of AD , PD and ALS , its beneficial effect in subsequent clinical
trials has been less robust, with no clear neurocognitive improvement
observed in AD or HD, and disease worsening was detected in ALS trials .
Therefore, although most of the pre-clinical data using minocycline
reported positive results, its current negative outcomes or even the
symptom worsening observed in some clinical trials, questions its
effectiveness as a therapy for neurodegenerative diseases . Other
anti-inflammatory therapies considered for neurodegeneration include
non-steroidal anti-inflammatory drugs (NSAIDs), however, the potential
beneficial effects observed in some animal models, have not been
reproduced in clinical trials and thus their use as potential therapy
for neurodegeneration dropped .
Considering the negative impact of sustained inflammation, mammals have
developed their own endogenous anti-inflammatory break, which is
mediated by regulatory T cells (Tregs), a subset of immune cells with
high immune suppressive capacity. Tregs are known to be either depleted
or functionally impaired in multiple neurodegenerative disorders . Thus,
systemic Treg expansion or Treg adoptive transfer have been considered
as a potential therapeutic approach to tackle neuroinflammation and
prevent neurodegeneration. Treg expansion through peripheral
interleukin-2/interleukin-2 monoclonal antibody complexes or adoptive
transfer upon ex vivo activation has rendered positive results in
mouse models of ALS and AD, such as SOD1G93A mice ,
5xFAD -Rag2Ko mice , 3-Tg-AD mice and APP/PS1 mice . Tregs have been
shown to protect motor neurons, supress astrocytic and microglial
immunoreactivity, reduce amyloid burden and restore cognitive
dysfunction. Moreover, an inverse correlation was observed between Treg
numbers and disease progression upon Treg expansion in ALS patients,
suggesting a neuroprotective effect also in humans . Despite the ample
evidence of a beneficial role for Treg having in neurodegenerative
diseases, systemic Treg expansion has not been widely considered for
clinical trials, as it can lead to systemic immune suppression in
patients of advanced aged and already vulnerable to infections, limiting
its therapeutic use. Recent investigations have developed a gene
delivery approach to locally expand Treg in the CNS by overexpressing
interleukin-2 in astrocytes, and thus, avoiding systemic immune
suppression. This adenoviral based gene delivery approach has rendered
positive results in mouse models of MS, stroke, and traumatic brain
injury . Even though still to be tested in primary neurodegenerative
disorders, such as ALS, PD or AD, this approach opens novel therapeutic
venue to harness Treg immunosuppressive capacity to limit CNS
neuroinflammation and neurodegeneration.
Last, we review the potential use of senolytics to limit
neuroinflammation and prevent neurodegeneration. The fact that mice
genetically engineered to remove p16INK4a+ senescent
cells show a decrease in age-related pathologies in several tissues,
together with an extended lifespan and health span , boosted the
interest in the development of senolytics, such as dasatinib, digoxin or
quercetin, as potential therapeutic approaches for neurodegeneration .
Senescent oligodendrocyte progenitor cells (OPC) have been found around
Aβ plaques in post-mortem tissue of patients with mild-cognitive
impairment or AD and APP/PS1 mice. The elimination of senescent OPCs by
the administration of the senolytic cocktail formed by dasatinib and
quercetin, decreased microglial activation, Aβ load, and the
concentration of inflammatory cytokines IL-6, IL-1β and TNF-α and
improved cognitive performance . Additionally, elimination of senescent
astrocyte and microglia via the administration of senolytic AP20187 in
MAPTP301SPS19 tau-pathology mouse model prevented
gliosis, hyperphosphorylation of tau and neurodegeneration and preserved
cognitive function . Similarly, removal of senescent microglia via
AP20187 or dasatinib and quercetin administration prevented age-related
cognitive decline and neuroinflammation . Thus, senolytics prevent
neurodegeneration in ageing and pre-clinical models of AD, supporting
their use in clinical trials with older adults suffering from MCI or
early-stage AD (NCT04685590. SToMP-AD and NCT04785300, ALSENLITE) ,
However, their beneficial effect in other neurodegenerative diseases
such as ALS or PD is yet to be investigated. Even though the
pre-clinical results in AD and ageing look promising, the use of
senolytic approaches to eliminate senescent cells should be considered
cautiously, due to the lack of knowledge regarding the role of senescent
cells in neurodegeneration. One alternative to avoid the potential
negative effects of eliminating senescent cells is to focus on
developing therapies aiming at reducing or eliminating SASP to limit
neuroinflammation . Senomorphics or SASP inhibitors can limit senescent
cell SASP production by inhibiting NF-kB, JAK-STAT, mTOR or
mitochondrial complex I and IV related targets. However, since
senomorphics do not eliminate senescent cells, continuous treatment with
SASP inhibitor would be required to obtain long-lasting effects, which
could also increase off-target effects associated with the suppression
of cytokine secretion by other cells. Thus, a better understanding of
the role of SASP and senescent cells in the CNS diseases and ageing is
essential to successfully develop senotherapeutic interventions to limit
neuroinflammation and target neurodegeneration .
Conclusions and future perspectives
We have attempted to briefly review the enormous field of the biological
pathways that are affected during ageing and implicated in the
pathogenesis of neurodegenerative disorders.
Even though a common feature of neurodegenerative diseases is the
abnormal deposition and mis-localization of insoluble protein
aggregates, different cellular pathways contribute to neuronal loss.
Furthermore, these pathways are all affected by ageing, which represents
the main common risk factor for most neurodegenerative disorders. Cells
in all regions of the CNS are affected by ageing, as indicated by the
decline of sensory, motor, and cognitive functions with time . However,
how cellular and molecular changes that occur during normal ageing
render neurons vulnerable to degeneration has not yet been fully
elucidated.
Currently, most efforts to treat neurodegenerative disorders focus on
strategies that target the insoluble aggregates of proteins specifically
associated with each neurodegenerative disorder. So far, most of the
clinical trial results have been disappointing because cognitive
function is not restored even when protein aggregates are removed. These
results point towards the importance of studying the cellular pathways
that contribute to neuronal dysfunction, in order to provide combined
therapies to the patients.
Neuronal function requires an efficient network of pathways that are
strictly connected and interdependent. For instance, synaptic function
is affected by the inflammatory microenvironment generated by the ageing
glial cells. Furthermore, synaptic transmission requires energy and
perturbed mitochondrial function has been associated with ageing and
neurodegeneration as the quality control of the cellular components is
regulated by energy sensors. Additionally, the lysosomal-dependent
self-digestive process of damaged proteins and organelles, called
autophagy, is important to generate nutrients and energy to maintain
essential cellular activities. Defects in autophagy results in
intracellular protein accumulation, contributing to the formation of the
insoluble aggregates of protein specifically associated to
neurodegenerative disorders. Finally, gene expression translates
synaptic activity and alterations in metabolic function into changes in
gene expression that can profoundly modify neuronal structure and
function.
Different therapeutic strategies have been developed to target these
cellular pathways and the drugs have been evaluated for the treatment of
different neurodegenerative disorders. However, the future challenges in
drug discovery for neurodegenerative disorders are (i) the detection of
the earliest events in the neurodegenerative cascade and (ii) the
identification of the pathways responsible for the specific
vulnerability of cellular populations in each neurodegenerative disease.
The understanding of these mechanism is critical to develop
disease-modifying therapies and to design tailored therapies that can be
administered to specific patients’ populations.ReferencesFigure legendFigure. 1 Scheme of the biological pathways contributing to
neurodegeneration