Alzheimer’s Disease: Models and Molecular Mechanisms Informing Disease and Treatments
Kaden Nystuen,
No information about this author
Shannon M. McNamee,
No information about this author
Monica Akula
No information about this author
et al.
Bioengineering,
Journal Year:
2024,
Volume and Issue:
11(1), P. 45 - 45
Published: Jan. 1, 2024
Alzheimer’s
Disease
(AD)
is
a
complex
neurodegenerative
disease
resulting
in
progressive
loss
of
memory,
language
and
motor
abilities
caused
by
cortical
hippocampal
degeneration.
This
review
captures
the
landscape
understanding
AD
pathology,
diagnostics,
current
therapies.
Two
major
mechanisms
direct
pathology:
(1)
accumulation
amyloid
β
(Aβ)
plaque
(2)
tau-derived
neurofibrillary
tangles
(NFT).
The
most
common
variants
Aβ
pathway
APP,
PSEN1,
PSEN2
are
largely
responsible
for
early-onset
(EOAD),
while
MAPT,
APOE,
TREM2
ABCA7
have
modifying
effect
on
late-onset
(LOAD).
More
recent
studies
implicate
chaperone
proteins
degrading
AD.
Several
tests,
such
as
cognitive
function,
brain
imaging,
cerebral
spinal
fluid
(CSF)
blood
used
diagnosis.
Additionally,
several
biomarkers
seem
to
unique
specific
combination
expression
could
potentially
be
improved,
less
invasive
diagnostics.
In
addition
genetic
perturbations,
environmental
influences,
altered
gut
microbiome
signatures,
affect
Effective
treatments
been
challenging
develop.
Currently,
there
FDA
approved
drugs
(cholinesterase
inhibitors,
Aß-targeting
antibodies
an
NMDA
antagonist)
that
mitigate
rate
decline
symptoms
distress.
Language: Английский
Advancements in APOE and dementia research: Highlights from the 2023 AAIC Advancements: APOE conference
Alzheimer s & Dementia,
Journal Year:
2024,
Volume and Issue:
20(9), P. 6590 - 6605
Published: July 19, 2024
Abstract
INTRODUCTION
The
apolipoprotein
E
gene
(
APOE
)
is
an
established
central
player
in
the
pathogenesis
of
Alzheimer's
disease
(AD),
with
distinct
apoE
isoforms
exerting
diverse
effects.
influences
not
only
amyloid‐beta
and
tau
pathologies
but
also
lipid
energy
metabolism,
neuroinflammation,
cerebral
vascular
health,
sex‐dependent
manifestations.
Furthermore,
ancestral
background
may
significantly
impact
link
between
AD,
underscoring
need
for
more
inclusive
research.
METHODS
In
2023,
Association
convened
multidisciplinary
researchers
at
“AAIC
Advancements:
APOE”
conference
to
discuss
various
topics,
including
their
roles
AD
pathogenesis,
progress
apoE‐targeted
therapeutic
strategies,
updates
on
models
interventions
that
modulate
expression
function.
RESULTS
This
manuscript
presents
highlights
from
provides
overview
opportunities
further
research
field.
DISCUSSION
Understanding
apoE's
multifaceted
will
help
develop
targeted
advance
field
precision
medicine.
Highlights
a
disease.
exerts
numerous
effects
throughout
brain
amyloid‐beta,
tau,
other
pathways.
AAIC
encouraged
discussions
collaborations
understanding
role
APOE.
Language: Английский
Single-nucleus multi-omics of Parkinson’s disease reveals a glutamatergic neuronal subtype susceptible to gene dysregulation via alteration of transcriptional networks
E. Keats Shwab,
No information about this author
Daniel Gingerich,
No information about this author
Zhaohui Man
No information about this author
et al.
Acta Neuropathologica Communications,
Journal Year:
2024,
Volume and Issue:
12(1)
Published: July 2, 2024
Abstract
The
genetic
architecture
of
Parkinson’s
disease
(PD)
is
complex
and
multiple
brain
cell
subtypes
are
involved
in
the
neuropathological
progression
disease.
Here
we
aimed
to
advance
our
understanding
PD
complexity
at
a
subtype
precision
level.
Using
parallel
single-nucleus
(sn)RNA-seq
snATAC-seq
analyses
simultaneously
profiled
transcriptomic
chromatin
accessibility
landscapes
temporal
cortex
tissues
from
12
compared
control
subjects
granular
single
resolution.
An
integrative
bioinformatic
pipeline
was
developed
applied
for
these
snMulti-omics
datasets.
results
identified
subpopulation
cortical
glutamatergic
excitatory
neurons
with
remarkably
altered
gene
expression
PD,
including
differentially-expressed
genes
within
risk
loci
genome-wide
association
studies
(GWAS).
This
only
neuronal
showing
significant
robust
overexpression
SNCA
.
Further
characterization
this
neuronal-subpopulation
showed
upregulation
specific
pathways
related
axon
guidance,
neurite
outgrowth
post-synaptic
structure,
downregulated
presynaptic
organization
calcium
response.
Additionally,
characterized
roles
three
molecular
mechanisms
governing
PD-associated
subtype-specific
dysregulation
expression:
(1)
changes
cis-regulatory
element
transcriptional
machinery;
(2)
abundance
master
regulators,
YY1,
SP3,
KLF16;
(3)
candidate
regulatory
variants
high
linkage
disequilibrium
PD-GWAS
genomic
impacting
transcription
factor
binding
affinities.
To
knowledge,
study
first
most
comprehensive
interrogation
multi-omics
landscape
cell-subtype
Our
findings
provide
new
insights
into
precise
subtype,
causal
genes,
non-coding
underlying
paving
way
development
cell-
gene-targeted
therapeutics
halt
as
well
biomarkers
early
preclinical
diagnosis.
Language: Английский
Chromatin accessibility provides a window into the genetic etiology of human brain disease
Trends in Genetics,
Journal Year:
2025,
Volume and Issue:
unknown
Published: Jan. 1, 2025
Language: Английский
Integrative Epigenomic Landscape of Alzheimer's Disease Brains Reveals Oligodendrocyte Molecular Perturbations Associated with Tau
Stephanie R. Oatman,
No information about this author
Joseph S. Reddy,
No information about this author
Amin Atashgaran
No information about this author
et al.
bioRxiv (Cold Spring Harbor Laboratory),
Journal Year:
2025,
Volume and Issue:
unknown
Published: Feb. 17, 2025
Abstract
Alzheimer’s
disease
(AD)
brains
are
characterized
by
neuropathologic
and
biochemical
changes
that
highly
variable
across
individuals.
Capturing
epigenetic
factors
associate
with
this
variability
can
reveal
novel
biological
insights
into
AD
pathophysiology.
We
conducted
an
epigenome-wide
association
study
of
DNA
methylation
(DNAm)
in
472
measures
(Braak
stage,
Thal
phase,
cerebral
amyloid
angiopathy
score)
brain
levels
five
proteins
(APOE,
amyloid-β
(Aβ)40,
Aβ42,
tau,
p-tau)
core
to
pathogenesis.
Using
a
regional
(rCpGm)
approach,
we
identified
5,478
significant
associations,
99.7%
which
were
tau
measures.
Of
the
tau-associated
rCpGms,
93
had
concordant
associations
external
datasets
comprising
1,337
samples.
Integrative
transcriptome-methylome
analyses
uncovered
535
gene
expression
for
these
rCpGms.
Genes
concurrent
perturbations
enriched
oligodendrocyte
marker
genes,
including
known
risk
genes
such
as
BIN1
,
myelination
MYRF,
MBP
MAG
previously
implicated
AD,
well
like
LDB3
.
further
annotated
top
additional
6
single
cell
2
bulk
transcriptome
from
two
other
tauopathies,
Pick’s
progressive
supranuclear
palsy
(PSP).
Our
findings
support
consistent
rCpGm
tauopathies
tau-related
phenotypes
both
tissue
clusters.
In
summary,
uncover
integrative
epigenomic
landscape
demonstrate
common
potential
pathomechanism
different
tauopathies.
Language: Английский
Personalized Single-cell Transcriptomics Reveals Molecular Diversity in Alzheimer's Disease
medRxiv (Cold Spring Harbor Laboratory),
Journal Year:
2024,
Volume and Issue:
unknown
Published: Nov. 4, 2024
Abstract
Precision
medicine
for
brain
diseases
faces
many
challenges,
including
understanding
the
heterogeneity
of
disease
phenotypes.
Such
can
be
attributed
to
variations
in
cellular
and
molecular
mechanisms
across
individuals.
However,
personalized
remain
elusive,
especially
at
single-cell
level.
To
address
this,
PsychAD
project
generated
population-level
single-nucleus
RNA-seq
data
1,494
human
brains
with
over
6.3
million
nuclei
covering
diverse
clinical
phenotypes
neuropsychiatric
symptoms
(NPSs)
Alzheimer’s
(AD).
Leveraging
this
data,
we
analyzed
functional
genomics
involving
cell
type
interactions
gene
regulatory
networks.
In
particular,
developed
a
knowledge-guided
graph
neural
network
model
learn
latent
representations
(embeddings)
quantify
importance
scores
types,
genes,
their
each
individual.
Our
embeddings
improved
phenotype
classifications
revealed
potentially
novel
subtypes
population
trajectories
AD
progression,
cognitive
impairment,
NPSs.
prioritized
genomic
information
showed
significant
differences
level
various
also
allowed
us
further
identify
subpopulation-level
biological
pathways,
ancestry
AD.
Finally,
associated
genetic
variants
type-gene
changes
individuals,
i.e.,
QTLs
(grQTLs),
providing
insights
compared
existing
QTLs.
We
validated
our
results
using
external
cohorts.
analyses
are
available
through
iBrainMap,
an
open-source
computational
framework,
as
atlas
Disease.
Language: Английский
Application of computational algorithms for single-cell RNA-seq and ATAC-seq in neurodegenerative diseases
Hwisoo Choi,
No information about this author
Hyeonkyu Kim,
No information about this author
Hyunhee Chung
No information about this author
et al.
Briefings in Functional Genomics,
Journal Year:
2024,
Volume and Issue:
24
Published: Nov. 5, 2024
Recent
advancements
in
single-cell
technologies,
including
RNA
sequencing
(scRNA-seq)
and
Assay
for
Transposase-Accessible
Chromatin
using
(scATAC-seq),
have
greatly
improved
our
insight
into
the
epigenomic
landscapes
across
various
biological
contexts
diseases.
This
paper
reviews
key
computational
tools
machine
learning
approaches
that
integrate
scRNA-seq
scATAC-seq
data
to
facilitate
alignment
of
transcriptomic
with
chromatin
accessibility
profiles.
Applying
these
integrated
technologies
neurodegenerative
diseases,
such
as
Alzheimer's
disease
Parkinson's
disease,
reveals
how
changes
gene
expression
can
illuminate
pathogenic
mechanisms
identify
potential
therapeutic
targets.
Despite
facing
challenges
like
sparsity
demands,
ongoing
enhancements
along
better
analytical
methods,
continue
expand
their
applications.
These
promise
revolutionize
approach
medical
research
clinical
diagnostics,
offering
a
comprehensive
view
cellular
function
pathology.
Language: Английский
Comparative mapping of single-cell transcriptomic landscapes in neurodegenerative diseases
E. Keats Shwab,
No information about this author
Zhaohui Man,
No information about this author
Daniel Oliveros Gingerich
No information about this author
et al.
Published: Dec. 18, 2024
ABSTRACT
INTRODUCTION
Alzheimer’s
disease
(AD),
Dementia
with
Lewy
bodies
(DLB),
and
Parkinson’s
(PD)
represent
a
spectrum
of
neurodegenerative
disorders
(NDDs).
Here,
we
performed
the
first
direct
comparison
their
transcriptomic
landscapes.
METHODS
We
profiled
whole
transcriptomes
NDD
cortical
tissue
by
snRNA-seq.
used
computational
analyses
to
identify
common
distinct
differentially
expressed
genes
(DEGs),
biological
pathways,
vulnerable
disease-driver
cell
subtypes,
alteration
in
cell-to-cell
interactions.
RESULTS
The
same
inhibitory
neuron
subtype
was
depleted
both
AD
DLB.
Potentially
disease-driving
neuronal
subtypes
were
present
PD
Cell-cell
communication
predicted
be
increased
but
decreased
DLB
PD.
DEGs
most
commonly
shared
across
NDDs
within
subtypes.
Overall,
observed
greatest
divergence
between
PD,
while
exhibited
an
intermediate
signature.
DISCUSSION
These
results
help
explain
clinicopathological
this
group
provide
unique
insights
into
molecular
mechanisms
underlying
pathogenesis
NDDs.
Language: Английский