ArticlePDF AvailableLiterature Review

Involvement of MicroRNA in Microglia-Mediated Immune Response

Authors:

Abstract and Figures

MicroRNAs (miRNAs) are an abundant class of small noncoding RNA molecules that play an important role in the regulation of gene expression at the posttranscriptional level. Due to their ability to simultaneously modulate the fate of different genes, these molecules are particularly well suited to act as key regulators during immune cell differentiation and activation, and their dysfunction can contribute to pathological conditions associated with neuroinflammation. Recent studies have addressed the role of miRNAs in the differentiation of progenitor cells into microglia and in the activation process, aiming at clarifying the origin of adult microglia cells and the contribution of the central nervous system (CNS) environment to microglia phenotype, in health and disease. Altered expression of several miRNAs has been associated with Alzheimer's disease, multiple sclerosis, and ischemic injury, hence strongly advocating the use of these small molecules as disease markers and new therapeutic targets. This review summarizes the recent advances in the field of miRNA-mediated regulation of microglia development and activation. We discuss the role of specific miRNAs in the maintenance and switching of microglia activation states and illustrate the potential of this class of nucleic acids both as biomarkers of inflammation and new therapeutic tools for the modulation of microglia behavior in the CNS.
This content is subject to copyright. Terms and conditions apply.
Hindawi Publishing Corporation
Clinical and Developmental Immunology
Volume , Article ID ,  pages
http://dx.doi.org/.//
Review Article
Involvement of MicroRNA in Microglia-Mediated
Immune Response
J. Guedes,1,2 A. L. C. Cardoso,3andM.C.PedrosodeLima
3,4
1PhD Programme in Experimental Biology and Biomedicine (PDBEB), CNC - Center for Neuroscience and Cell Biology,
University of Coimbra, 3004-517 Coimbra, Portugal
2Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789 Coimbra, Portugal
3CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
4Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3001-401 Coimbra, Portugal
Correspondence should be addressed to M. C. Pedroso de Lima; mdelima@ci.uc.pt
Received  March ; Accepted  May 
Academic Editor: Anirban Basu
Copyright ©  J. Guedes et al. is is an open access article distributed under the Creative Commons Attribution License, which
permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
MicroRNAs (miRNAs) are an abundant class of small noncoding RNA molecules that play an important role in the regulation
of gene expression at the posttranscriptional level. Due to their ability to simultaneously modulate the fate of dierent genes,
these molecules are particularly well suited to act as key regulators during immune cell dierentiation and activation, and their
dysfunction can contribute to pathological conditions associated with neuroinammation. Recent studies have addressed the role
of miRNAs in the dierentiation of progenitor cells into microglia and in the activation process, aiming at clarifying the origin
of adult microglia cells and the contribution of the central nervous system (CNS) environment to microglia phenotype, in health
and disease. Altered expression of several miRNAs has been associated with Alzheimer’s disease, multiple sclerosis, and ischemic
injury, hence strongly advocating the use of these small molecules as disease markers and new therapeutic targets. is review
summarizes the recent advances in the eld of miRNA-mediated regulation of microglia development and activation. We discuss
the role of specic miRNAs in the maintenance and switching of microglia activation states and illustrate the potential of this class
of nucleic acids both as biomarkers of inammation and new therapeutic tools for the modulation of microglia behavior in the
CNS.
1. Introduction
Microglia cells are crucial for the development and main-
tenance of the central nervous system (CNS). In addition
to acting as sensors of environmental changes that precede
pathological events, these cells have been shown to support
neuronal function by monitoring synaptic activity, control-
ling synaptogenesis, and promoting neuronal apoptosis dur-
ing development []. Although they are considered one of
the four major cellular types of the CNS, they do not originate
from the same precursor cells as astrocytes, oligodendrocytes,
or neurons. Instead, they derive from myeloid progenitor
cells and share several markers with peripheral monocytes,
macrophages, and dendritic cells, such as CDb, F/, and
CD []. e rst resident parenchymal microglia cells are
believed to originate from yolk sac immature macrophages
in early stages of fetal development. In humans, microglia
precursor cells arrive at the brain in two waves during the
rst and second trimester of gestation, while in rodents this
migration occurs shortly before and aer birth.
e sudden increase in CDb+and F/+cells observed
in the early postnatal period in rodents was until recently
attributed to the recruitment of bone marrow derived cells,
suggesting that myeloid precursors could also contribute to
the initial pool of microglia cells in the CNS. However, most
of the studies supporting these ndings used irradiation of
the recipient animals to allow bone marrow engrament
of genetically-labeled cells [], which was later found to
strongly inuence the observed results []. In , Ginhoux
andcolleaguesshedlightontheoriginofmicroglia.e
authors performed in vivo lineage tracing studies using Cre
recombinase activity, which was induced into pregnant mice
between days  and  of fetal development, when embryonic
hematopoiesis is restricted to the yolk sac. e results from
Clinical and Developmental Immunology
this study clearly demonstrated that postnatal hematopoietic
progenitors do not contribute signicantly to microglia post-
natal numbers and that the cellular expansion observed in
this period is mainly dependent on the proliferation of the
resident yolk sac-derived microglia population [].
e question remains whether this is also true in the
adultbrain,especiallyfollowinganeurologicalinsultorin
the case of a neurodegenerative disease, wherein the integrity
of the blood-brain barrier (BBB) may be compromised.
Several studies have shown that the inltration of bone
marrow-derived cells into the brain is possible under those
circumstances and may even play a central role in disease
modulation. Nevertheless, the exact nature of the contri-
bution of parenchymal and blood-derived microglia to the
neuroimmune response, in the context of neuronal disease,
remainstobeclaried[].
Following their migration to the neuronal tissue, micro-
glia cells assume a surveying phenotype, usually referred as
“resting microglia,” characterized by a small and static cell
body,alargenumberofhighlymotileramications,andlow
expression of macrophage-related surface markers, such as
the major histocompatibility complex II (MHC II) and CD
[]. e low levels of these markers distinguish parenchymal
“resting” microglia from peripheral macrophages. However,
following a neuronal insult, such as ischemia, infection,
and trauma or in the presence of inammatory mediators
(IFN-𝛾), microglia cells assume an amoeboid form, losing
their ramications, and overexpressing the above-mentioned
markers. is process is referred to as microglia “activation
and is known to induce profound phenotypical changes,
making parenchymal microglia to become almost indistin-
guishable from peripheral macrophages [].
Similarly to what has been described in macrophages,
microglia activation can also originate dierent subsets of
cells, depending on the nature of the activating stimulus
and surrounding environment. ese dierent activation
phenotypes express distinct molecular markers and exert
dierent functions in the neuronal tissue []. e denition
of the dierent activation states of macrophages was initially
based on the expression of proinammatory receptors and
cytokines (M phenotype—classical activation) or on the
expression of anti-inammatory receptors and cytokines (M
phenotype—alternative activation). Further studies revealed
the existence of several intermediate activation states, which
led to the conclusion that these two basic phenotypes can
overlap and that macrophages are able to assume a broad
spectrum of phenotypes that cannot be oversimplied and
separatedintodiscretecategories.
Contrarily to macrophages, the mechanisms responsible
for microglia phenotype regulation in the CNS are poorly
understood. M activation, which is characterized by an
increase in the production of IL-𝛽,IL-,TNF-𝛼,andnitric
oxide (NO), has been identied following acute brain injury
caused by trauma or stroke [,]. However, in the context of
neurodegenerative disorders, the distinction between the M
and M phenotypes has proven to be more challenging. While
several studies have identied the presence of M markers,
such as TGF-𝛽and IL-, in the brain of Alzheimer’s disease
(AD) animal models, as well as an increase in the expression
of M genes AG (arginase ) and CHIL/CHIL (chitinase
-like /) in AD patients [], inammation in the human
AD brain has also been associated with upregulation of IL-
, IL-𝛽,andTNF-𝛼,allmarkersoftheMstate,inthe
vicinity of amyloid deposits [,]. Most authors believe that
the M-like phenotypes are less aggressive to the neuronal
tissue, promoting tissue repair and phagocytosis of protein
aggregatesandcelldebris,whiletheM-likephenotypes
are more prone to induce neuronal toxicity by themselves,
due to the high expression of inammatory mediators and
NO.WhetherhumanmicrogliacanswitchfromanMto
an M phenotype with a detrimental eect to the brain is
still not clear. However, several studies have pointed to the
possibility of microglia “priming,” a phenomenon associated
with age and chronic inammation, in which exposure to
low levels of systemic signaling molecules can exacerbate
microglia response to a second local stimulus, such as the
presence of A𝛽aggregates, potentiating the development
of tissue damaging phenotypes []. Although some of the
molecular intervenients and exogenous stressors underlying
microglia activation in vitro have been identied [],
in more complex environments, such as the diseased brain,
there is still a lack of answers concerning the molecular
mechanisms responsible for microglia phenotypic changes
[].isledseveralscientiststoproposearoleforcertain
key transcription factors and microRNAs (miRNAs) in these
processes [].
2. MiRNA Biogenesis and Activity
MiRNAs are transcribed from intragenic or intergenic
regions by RNA polymerase II or RNA polymerase III,
originating large stem-loop hairpin structures, designated
pri-miRNAs []. ese structures, which are asymmetrically
cleaved by an enzymatic complex containing Drosha, a
RNAse III endonuclease, originate hairpin-structured pre-
cursors designated pre-miRNA [,]. Alternatively, non-
canonical pathways for pre-miRNA biogenesis can occur,
such as the production of mirtrons, which consist in pre-
miRNAs generated through the direct splicing of introns [].
Pre-miRNAs are exported to the cytoplasm by the com-
plex Exportin-/RanGTP, which recognizes and binds the
characteristic overhangs of pre-miRNAs. ese precursors
arethenincorporatedintoaprocessingcomplexcontaining
another RNAse III endonuclease (Dicer), which is responsi-
ble for removing the stem-loop of the precursor, originating a
double-stranded miRNA molecule with 󸀠overhangs (mature
miRNA). is duplex molecule is then incorporated into
the RNA-induced silencing complex (RISC) or the microri-
bonucleoprotein complex (miRNP), where the strand with
the least thermodynamically stable 󸀠endisusedtoactivate
the complex and guide it to complementary mRNA targets
[]. During miRNA-mediated posttranscriptional gene reg-
ulation, the RISC complex, containing the single stranded
miRNA template, binds mainly to the 󸀠untranslated region
(󸀠UTR) of the target mRNA, in one or more locations. e
complementarity between the mRNA and the nucleotides –
onthe
󸀠region of the miRNA (seed region) is responsible
for this binding and allows the recognition of multiple mRNA
Clinical and Developmental Immunology
targets by a single miRNA molecule []. MiRNA-mediated
regulation of gene expression is achieved either by translation
repression or degradation of the mRNA target molecule []
and has been associated with many important biological
processes, including inammation, apoptosis, angiogenesis,
development, proliferation, patterning, and dierentiation.
3. MiRNA Role in Microglia Development
e mechanism of miRNA-mediated posttranscriptional reg-
ulation is very well suited to the control of fate-changing
cellular events, such as dierentiation and activation. Since
these processes usually involve changes in several proteins
and dierent signaling pathways, such control can be easily
achieved through the expression of a single miRNA or a set
of specic miRNAs. In addition, miRNAs can directly target
transcription factors, the master regulators of most cellular
events. A number of studies have revealed that miRNAs
may provide a genetic switch to inactivate a set of target
genes through the regulation of a specic transcription factor,
while miRNA expression can be regulated by transcription
factors that bind upstream of pre-miRNA genes [,].
erefore, these two classes of regulators can work together
to orchestrate complex cellular events, such as monocyte-
macrophage dierentiation and microglia development from
primitive macrophages in the yolk sac or bone marrow
precursors.
Two transcription factors, namely, CEBP𝛼and PU.,
have been shown to be critical for monocyte/macrophage
dierentiation and microglia development, CEBP𝛼being
considered the master regulator of hematopoietic stem cell
dierentiation. Several miRNAs are controlled directly by
CEBP𝛼, including miR-, which is also regulated by the
transcription factor NFI-A. However, while CEBP𝛼promotes
miR- expression, NFI-A inhibits the expression of this
miRNA [], which represses NFI-A through a feedback loop.
erefore, when CEBP𝛼levels are high, miR- expression
is enhanced, and NFI-A levels decrease, promoting granulo-
cyte dierentiation, but when NFI-A levels are relatively high
and miR- levels are low, other pathways are favored, such
as monocyte dierentiation.
On the other hand, PU. is required to promote the skew-
ing of granulocyte-macrophage progenitors to the monocyte
lineage. PU. levels, although directly controlled by CEBP𝛼,
have to be relatively high when compared to CEBP𝛼lev-
els, to avoid favoring the granulocyte lineage. MiR- is
upregulatedbyPU.and,throughthetargetingofNFI-A,
upregulates the expression of the M-CSF receptor, skewing
dierentiation towards the monocyte/macrophage lineage in
thebonemarrow[]. Forrest and coworkers have demon-
strated that, in addition to miR-, miR-, miR-, and
miR- play an important role in monocyte dierentiation
through combinatorial regulation []. ese authors have
shown that when overexpressed, these miRNAs are able to
cause cell-cycle arrest and partial dierentiation in THP-
 cells (leukemia model). MiR- and miR- caused G
arrest and apoptosis, while miR- and miR- caused G
arrest and the downregulation of miR-, an antidierentiative
miRNA, by targeting a site in its primary transcript.
In another recent study, the oncogenic miR-- cluster
(which carries six miRNAs: , a, a, a, b-
, and a) has been directly associated with the process
of monocyte to macrophage dierentiation, in which PU.
and Egr are also involved []. Upon dierentiation into
macrophages, the transcription factor PU. was found to
induce the secondary determinant Egr which, in turn,
directly represses miR-- expression by promoting histone
H demethylation within the CpG island of the miR--
promoter. Conversely, Egr itself is targeted by miR--,
indicating the existence of a mutual regulatory relationship
between miR-- and Egr []. Given the similarities
betweenmacrophagesandmicroglia,itisreasonableto
assume that some of these miRNAs and transcription factors
also play an important role in microglia dierentiation in
the brain, and it would be interesting to ascertain whether
the same regulatory loops might exist in yolk sac-derived
microglia.
So far, the only study addressing miRNA contribution to
microglia development was performed by Ponomarev and
colleagues [,].eseauthorsshowedthatmiR-,one
ofthemostabundantmiRNAsinthebrain,isrequiredto
maintain the quiescent state of microglia in the brain. By
targeting the transcription factor CEBP𝛼and the cyclins
CDKandCDK,miR-isabletoreducetheexpressionof
PU. and its downstream target, the M-CSF receptor, restrict-
ing cellular proliferation and potentiating the dierentiation
of primitive macrophages to adult microglia in the brain
[]. While during the rst two weeks aer birth, microglia
isolated from the brain presented low levels of miR- and
aCD
high/MHC class IIhigh phenotype, characteristic of
active and proliferating cells, adult microglia presented the
opposite phenotype, CDlow/MHC class IIlow/miR-high
[,]. e authors hypothesized that the high levels of miR-
 observed in adult microglia are a specic consequence of
the CNS environment. is idea was based on their observa-
tion that sublethally irradiated mice, transplanted with bone
marrow GFP+progenitor cells exhibiting a CDhigh/MHC
class IIhigh/miR-low phenotype, presented GFP+CDb+
positive cells in the brain with a CDlow/MHC class
IIlow/miR-high phenotype. To conrm this hypothesis,
Ponomarev and colleagues cocultured bone-marrow-derived
macrophages with astroglial or neuronal cell lines, and they
observed, in both cases, a downregulation of MHC class
IIandCDlevelsaswellasanupregulationofmiR-
expression. Several suggestions were made concerning the
mechanisms underlying miR- upregulation in microglia,
including the direct transfer of miR- from neuronal cells
to microglia through exosomal shuttle vesicles, direct cell-to-
cell contact between these two cell types and the release of
anti-inammatory factors, such as CXCL and TGF-𝛽,by
neuronal cells [,].
4. MiRNA Role in Microglia Activation
In addition to being involved in the regulation of the dieren-
tiation process of microglia, several studies suggest a role for
miRNAs as modulators of M and M polarization in both
Clinical and Developmental Immunology
microglia and macrophages. MiR-, broadly considered a
proinammatory miRNA, was one of the rst miRNAs to be
directly linked to the M phenotype (Figure ). is miRNA
was shown to be upregulated in macrophages, monocytes,
and microglia in response to several proinammatory stim-
uli, such as LPS, IFN-𝛾,andTNF-𝛼[,,]. In this
regard, we have recently shown that miR- targets anti-
inammatory proteins in microglia, such as the suppressor of
cytokine signaling  (SOCS-), leading to the upregulation of
several inammatory mediators characteristic of the M phe-
notype, including the inducible nitrogen synthase (iNOS), IL-
, and TNF-𝛼[], as described in Figure . In addition, miR-
 upregulation increases the expression of IFN-𝛽,which
is probably related with a feedback mechanism to control
theimmuneresponse,sinceIFN-𝛽isknowntoupregulate
the expression of SOCS- and IL-, two important anti-
inammatory mediators [,]. Our results conrm that
miR- upregulation contributes to a microglia-mediated
neurotoxic response, which has been largely associated with
the M phenotype. Furthermore, recent studies have shown
that miR- is able to target M-associated genes, such as
that encoding SMAD, a protein involved in the TGF-𝛽
pathway [], and CEBP𝛽,atranscriptionfactorimportant
fortheexpressionofIL-,arginase,andCD[], further
supporting the hypothesis that miR- is required to skew
microglia activation to M-like phenotypes.
Several other miRNAs have been directly related to
the M phenotype, including miR- and miR-b. Zhu
and coworkers observed an upregulation in miR- levels
in response to several TLR ligands in macrophages. e
overexpression of this miRNA resulted in the downreg-
ulation of MAPK phosphatase  (MPK-), promoting the
activation of MAPK and the expression of M-associated
proinammatory cytokines, such as IL-, TNF-𝛼,andIL-
. On the other hand, miR- inhibition enhanced MPK-
 expression and decreased p and JNK activation [].
Regarding miR-b, Chaudhuri and colleagues reported an
increase in M macrophage activation, with upregulation of
MHC class II, CD, CD, and CD, upon overexpression
of this miRNA. e authors related these eects to miR-
 targeting of interferon regulatory factor  (IRF) and
also observed that, upon forced expression of miR-b,
macrophages adopted an M cytotoxic phenotype, presented
elevated responsiveness to IFN-𝛾, and were more eective in
killing EL T-lymphoma tumor cells in vitro and in vivo [].
In contrast to miR- and miR-b, miR-a was
recentlyshowntobedownregulatedinresponsetothe
activation of dierent TLRs, this decrease being neces-
sary to enhance TLR-triggered production of inammatory
cytokines in macrophages []. On the other hand, miR-
a overexpression inhibited the activation of the JNK/C-
JUN pathway through the targeting of mitogen-activated
protein kinase kinase  (MKK), which activates JNK/stress-
activated protein kinase and a direct target of miR-a.
erefore, miR-a downregulation can be considered as an
additional requirement for M macrophage activation [].
Finally, as illustrated in Figure ,miR-hasbeen
reported to contribute to the M phenotype of macrophages
and microglia, since its forced overexpression led to the
downregulation of M-associated markers, such as IL-,
TNF-𝛼,andiNOS,andtoanincreaseofproteinsassociated
with the M phenotype, as is the case of TGF-𝛽, arginase-,
and FIZZ [].Itwasconcludedthat,inmicroglia,aninverse
correlation exists between the expression of M activation
markers and that of miR- (Figure ). Indeed, the highest
levels of miR- were detected in CDlow/MHC class
IIlow nonactivated resident microglia, but miR- expression
decreased dramatically upon cell treatment with IFN-𝛾and
GM-CSF, two stimulus known to upregulate CD and MHC
class II expression and to potentiate the M phenotype [].
Table lists several miRNAs involved in microglia activation,
indicating the phenotype (M or M) favored by their up- or
downregulation.
Taken together, the above-mentioned results highlight
the prominent role of miRNAs in the regulation of the
activation of both microglia and macrophages and open new
possibilities in the eld of anti-inammatory therapies. Using
appropriate gene therapy tools, miRNA modulation could
be an interesting and promising strategy to ne-tune the
immune response, skewing cell activation to the M or M
phenotypes according to the specic requirements of each
disease setting.
5. MiRNA Dysregulation in
Neurodegeneration and Neuroinflammation
Neurodegeneration is characterized by neuronal loss of spe-
cic neuronal circuits associated with cognitive and motor
functions. In this context, neuronal death is considered both
cause and consequence of neuroinammation, a process
involving microglia and astrocyte proliferation and activa-
tion. e excessive production of inammatory mediators
by these cells propagates inammation in the brain and
contributes to the triggering of a local and systemic immune
response, which is characterized by changes in miRNA levels
in the nervous tissue and in the periphery. Although the
trac of peripheral mononuclear cells from the blood stream
to the CNS is tightly regulated at the level of the BBB, it is
knownthattheintegrityofthisbarrierishighlycompromised
in severe brain diseases, such as stroke, brain trauma [],
and AD [,,]. e local disruption of the BBB in
a disease context facilitates the passage of blood-derived
cells to the nervous tissue. erefore, both microglia and
peripheral mononuclear phagocytes are able to impact the
pathoetiology of neurodegenerative diseases, and miRNAs
presenting altered expression levels in both of these cell
types can be used as new potential biomarkers and targets of
neurodegeneration.
rough the cellular crosstalk between the brain and
the periphery, the nervous system can inuence peripheral
immune functions, and, conversely, the immune system can
aect neuronal activity []. A specic subset of miRNAs
is able to regulate both cognitive and immune features, but
the neuroimmune impact of these molecules is far from
being fully understood. However, the role of miRNAs in
neuroimmune pathologies has recently started to be unveiled.
In the past few years, a large number of studies have emerged
Clinical and Developmental Immunology
M2 phenotype
Resting microglia
M1 phenotype
CD45 MHC II CD206
miR-124
miR-155
miR-124
miR-155
miR-155
miR-124
TGF-𝛽
IL-10
IFN-𝛾
GM-CSF
LPS
iNOS FIZZ-1
A𝛽??
IL-6, TNF-𝛼and IFN-𝛽
SOCS-1, CEBP𝛽and SMAD2
IL-10, Arg-1 and TGF-𝛽
c-Jun, NF-𝜅B
F : MiRNA contribution to microglia activation phenotypes. Resting microglia cells are characterized by low expression levels of
miR- and a relatively high expression of miR-. In the presence of strong inammatory stimuli, such as IFN-𝛾,GM-CSF,andLPS,
microglia assume a classical activation phenotype (M), characterized by the upregulation of both CD and MHC II, the expression of several
inammatory mediators, such as iNOS, the inammatory cytokines IL- and TNF-𝛼, the type I interferon IFN-𝛽, and the downregulation
of miR-. MiR- upregulation is thought to be crucial for the establishment of this phenotype, since this miRNA directly targets several
anti-inammatory molecules, including SOCS-. Alternatively, in the presence of TGF-𝛽or the anti-inammatory cytokine IL-, a dierent
activation phenotype is observed (M). In this case, CD is upregulated at the cell surface, and anti-inammatory molecules involved
in tissue repair and angiogenesis are expressed. Moreover, most proinammatory pathways, including those regulated by the transcription
factors c-Jun and NF-𝜅B, are inactivated, and miR- upregulation is not observed. Certain endogenous danger signals associated with
neurodegenerative disorders, such as A𝛽brils present in senile plaques of AD patients, can also cause microglia activation, although the
exact nature of the observed phenotypic changes is yet to be fully characterized.
T : MiRNAs in microglia and macrophage activation.
Role Phenotype Ref
miR-
Tar g e t s S O CS -  c ausing of iNOS, TNF-𝛼,IL-,
and IFN-𝛽
Tar g e t s S M A D  a n d C E B P 𝛽M [,,]
miR- Lead to IL-, iNOS, and TNF-𝛼and TGF-𝛽,
arginase-, and FIZZ M [,]
miR- MPK- promoting MAPK activation and IL-,
TNF-𝛼, and IL- expression M []
miR-b MHC class II, CD, CD, and CD through
targeting of IFR
Elevates responsiveness to IFN-𝛾M []
miR-a
Inhibited activation of JNK/c-Jun through
targeting of MKK
Its downregulation is necessary to promote the
M phenotype
M []
reporting the disruption of miRNA expression during neu-
roinammation and neurodegeneration processes []. For
example, miR-, which has been directly related with AD
[] and epilepsy [], has also been shown to be highly
overexpressed in A𝛽and TNF-𝛼stressed human microglia
cells, and this eect was inversely correlated with the levels of
inammation-related proteins, such as CFH and IRAK- [].
Another brain enriched miRNA, miR-, which has been
directly related with the maintenance of the quiescent state of
microglia [], has also been shown to inhibit the neuronal
transcription regulator complex REST, which is involved in
Rett syndrome [] and neuronal changes during chronic
cocaine intake [].
MiRNA dysregulation in the context of neurodegenera-
tion can be a consequence of genetic or sporadic anomalies.
In addition, the miRNA-related machinery can be impaired
Clinical and Developmental Immunology
in neurological disorders []. Accordingly, altered miRNA
proles have been identied in several neurodegenerative
diseases. Striatal miR-, miR-c, miR-, miR-, miR-
, miR-, miR-, miR-, and miR--p, as well as
the cellular levels of Drosha were shown to be downregulated
in both YAC and R/ transgenic mouse models of
Huntington’s disease (HD) []. In the frontal cortex and
striatum of human HD brains, several miRNAs were found
to be altered with respect to the brains of healthy subjects,
andchangesintheprimarynucleotidestructureofthe
󸀠
terminus of certain miRNAs were also reported []. MiRNA
proling studies in Parkinson’s disease (PD) revealed an early
downregulation of miR-b/c in human brain areas, with
variable neuropathological eects at clinical (motor) stages
[],whileinALSmousemodelsdeciencyinmiR-
accelerated disease progression [].
A large number of studies using cellular and mouse mod-
els, human hippocampus, human cortex, and whole brain
samples have revealed altered miRNA expression proles
in AD. MiR-b was found to be aberrantly expressed in
the APPswe/PSΔE AD mouse model, and its levels were
correlated with the dysregulated expression of the TGF-𝛽
type II receptor []. Wang and coworkers have shown that
the exposure of SH-SYY cells to A𝛽1−42 oligomers leads
to the increase of miR-b expression and to the conse-
quent impairment in TGF-𝛽signaling, providing a possible
explanation for the observed neurodegeneration []. As
mentioned before, TGF-𝛽signaling is a marker of the M
microglia phenotype, and, as such, miR-b overexpression
could be considered an additional cause of microglia skewing
to the M phenotype following A𝛽exposure.
Interestingly, Schonrock and collaborators have shown
that half of the tested miRNAs in their study were downregu-
lated in hippocampal neuronal cultures in response to A𝛽1−42
peptides and that the dysregulated miRNAs were likely to
aect target genes belonging to signaling pathways known
to be disrupted in AD. ese results were further validated
in the hippocampus of APP mice and human AD brains
[]. Curiously, many of those miRNAs, as miR-, miR-
a, let-i, and miR-b, had already been associated with
inammation.
Although the majority of miRNA proling studies in
neurodegenerative diseases has been performed in brain
samples, miRNA dysregulation has been found in other
biological sources, such as plasma, peripheral blood, and
cerebral spinal uid (CSF). For example, miR-a was shown
to be signicantly increased in plasma from HD gene carriers
prior to symptom onset, suggesting that plasma miRNAs
canbeusedasbiomarkersinHD[]. In another study,
eighteen miRNAs were found to be dierentially expressed in
peripheral blood mononuclear cells of nineteen PD patients
with respect to miRNA levels in thirteen control subjects
[]. In blood serum from AD patients, miR-, miR-c,
miR-, and miR-a/b were found to be downregulated,
and although the ability of these miRNAs to conclusively
diagnose AD is currently unknown, these blood-circulating
miRNAs have potential to be used as additional biomarkers
of the disease []. Although more dicult to obtain, CSF
has also been considered a source of biomarkers for many
neurological diseases, since this uid constantly exchanges
material with the brain parenchyma and is less prone to the
inuence of peripheral organs. Alexandrov and collaborators
analyzed miRNA abundance in the CSF of AD patients and
observed a signicant increase in the levels of miR-, miR-
b, miR-a, and miR-, with respect to age-matched
controls. Interestingly, these miRNAs are NF-𝜅B-sensitive
proinammatory miRNAs, also known to be upregulated
in AD brains and have been associated with progressive
spreading of neuroinammation []. Taken together, these
results indicate that the eective application of miRNAs as
biomarkers for neurodegenerative diseases should include
miRNAproling,notonlyinthebloodbutalsoinserum,
plasma, and dierent cellular subtypes, as well as the parallel
correlation of the obtained results with brain morphology
overtime, in order to overcome clinical issues related with
disease staging and progression.
MiRNA deregulation has also been associated with viral-
induced neuroinammation and neurodegenerative pro-
cesses. Mishra and coworkers showed that human microglia
cells treated with HIV- Tat-C protein, a molecule involved
in neuroinammation in HIV-infected patients, present
increased miR- expression with consequent changes in
the levels of the downstream target TRAF, which, in turn,
was found to control IRF and IRF expression []. It was
also demonstrated that miR-a is upregulated in human
microglia cells under HIV infection, regulating the inam-
matory response by targeting the CCL [] chemokine.
erefore, it seems that miRNA expression is altered aer
inammation in immune and glial cells in order to support
the ne-tuning of immune functions essential to main-
tainbrainhomeostasis.AninterestingstudybyDaveand
Khalilishowedthatmorphine-inducedinammationand
oxidative stress in human monocyte-derived macrophages
contribute to the expansion of the HIV- viral reservoir in
the CNS and HIV-associated dementia []. e authors
provided evidence that this process is regulated by dier-
entially expressed miRNAs, including miR-b and miR-
b, both linked to several targets in the proinammatory
pathways.
MiRNAsarealsobelievedtomodulatemicrogliainam-
mation aer hypoxia/ischemia, which may contribute to
neuronal damage. Hypoxia causes upregulation of the Fas
ligand (FasL) and simultaneously downregulation of miR-
 in microglia, inuencing neuronal apoptosis. Interest-
ingly, according to the work of Zhang and colleagues, the
ectopic expression of miR- partially protects neurons from
cell death caused by hypoxia-activated microglia []. e
same authors reported a potential role for miR-c in
the regulation of TNF-𝛼expression aer hypoxia/ischemia
and microglia-mediated neuronal injury. ey showed that
oxygen-glucose deprivation (OGD) induces microglia acti-
vation in vitro and in the hippocampus of Wistar rats
(four-vessel occlusion—-VO—rat model of ischemia), as
concluded by observation of the overproduction of TNF-𝛼,
IL-𝛽, and NO and the downregulation of miR-c. On the
other hand, the heterologous expression of this miRNA was
found to protect neurons from cell death caused by OGD-
activated microglia [].
Clinical and Developmental Immunology
Regarding neurodegenerative pathologies with a known
inammatory component, such as multiple sclerosis (MS),
miRNAshavealsobeenshowntoplayacentralrolein
theregulationofmicroglia-mediatedimmuneresponses.
Ponomarev and coworkers have demonstrated that miR-
 is able to switch microglia from an inammatory to a
quiescent state, and this phenomena was considered essential
to successfully inhibit the onset of EAE []. Also in MS, miR-
−/− knockdown mice were shown to present signicantly
reduced numbers of encephalogenic CD+ cells, an
inammatoryT-cellsubsetwithanimportantroleinthis
disease []. In a very recent study, Butovsky et al. showed
that the modulation of the cytokine prole of inammatory
monocytes using an anti-LyC mAb reduced monocyte
recruitment to the spinal cord, decreased neuronal loss, and
extended survival in a mouse model of ALS. Interestingly, in
humans with ALS, the analogous CD+CDmonocytes
were shown to exhibit an ALS-specic miRNA inammatory
signature, which can be used as a biomarker of this disease
and reveal novel therapeutic targets [].
Another interesting eect associated with miRNAs
derives from their capacity to activate receptors by them-
selves, aer being secreted by cells within the CNS, which
allows them to function as signaling molecules. For example,
let- is known to induce neurodegeneration by binding to
TLR in neurons and microglia. Let- is increased in the CSF
of AD patients, and accordingly, the injection of this miRNA
in the CSF of wild-type animals caused neurodegeneration,
which did not occur in mice lacking TLR []. Also in the
context of AD, the NF-𝜅B-dependent miR-a was reported
to be upregulated in the brain of AD patients and to enhance
inammation by targeting the complement factor H (CFH)
[]. MiR-a was also found to be overexpressed in prion-
infected mouse brain tissues, concurrent with the onset of
prion deposition and microglia activation, which suggests
that this miRNA plays a role in the proinammatory response
of microglia to prion replication [].
e above-mentioned studies stress the role of miRNAs
as modulators of both neuroinammation and neurodegen-
eration and illustrate their potential as biomarkers and novel
therapeutic targets in CNS diseases.
6. MiRNA-Based Therapeutic
Applications in Neuroimmune and
Neurodegenerative Diseases
Over the last few years, there has been a signicant progress
in the development of strategies to modulate the levels of
certain miRNAs and miRNA clusters, aiming at adjusting
cellular functions dysregulated in several pathologies. Mod-
ulation of mature miRNAs can be accomplished by oligonu-
cleotides complementary to miRNA sequences (miRNA
inhibitors), causing miRNA knockdown or, alternatively,
by miRNA precursor oligonucleotides (miRNA mimics),
which cause miRNA overexpression []. Usually, these
oligonucleotides present chemical modications, such as the
incorporation of locked nucleic acids (LNA) nucleotides,
which have a methylene bridge between the 󸀠-oxygen and
the 󸀠-carbon of the ribose moiety or the incorporation of
󸀠-O-methyl modied nucleotides (antagomirs) in certain
positions of the oligonucleotide sequence. e purpose of
these modications is to increase the chemical stability and
resistance of the miRNA inhibitors or mimics to serum nucle-
ases, thus potentiating their therapeutic potential. However,
in order to achieve a successful clinical application of miRNA-
based therapies it is crucial that these oligonucleotides are
properly delivered by vehicles that not only reliably and
eectively overcome cellular and physiological barriers but
are also highly target specic. e presence of the BBB, which
restricts entry of therapeutic molecules into the brain, as
well as the possible degradation of nucleic acids by nucleases
present in the blood, constitutes major obstacles associated
with nucleic acid delivery in vivo. Nonviral vectors have been
developed to ensure protection and improvement of nucleic
acid delivery into target cells and have been employed in
miRNA-based therapeutic approaches to modulate neuroin-
ammatory signaling pathways [].
Inourownwork,wehaveshownthattheuseofa
nonviral strategy to promote miR- silencing in microglia,
prior to their activation, is able to reduce the release of
NO and other inammatory mediators, such as the major
inammatory cytokines TNF-𝛼and IL- to the extracellular
environment, through an increase in the levels of SOCS-, a
direct target of miR- (Figure ). e modulation of this
miRNA in microglia cells, prior to their activation with LPS,
proved to be enough to improve cell viability in neuronal
cultures incubated with microglia conditioned medium [].
ese results stress the neuroprotective potential of miR-
silencing in the context of neuroinammation.
Other miRNAs have been related with the regulation of
the neuroimmune response. As discussed before, miR- is
one of the main miRNAs responsible for the maintenance of
the quiescent state of microglia/macrophages in the brain and
spinal cord, and therefore, miR- may constitute an impor-
tant target for the development of therapeutic approaches
towards the control of neuroinammation. Recently, Wille-
men and coworkers investigated the contribution of miR-
to the regulation of hyperalgesia and microglia/macrophage
activation in LysM-GRK+/− mice, in which the expression of
the G protein-coupled receptor kinase (GRK)  is decreased
to about % in microglia/macrophages, with respect to wild-
type animals. ese mice develop inammatory hyperalgesia
caused by activation of microglia/macrophages in the spinal
cord. e authors showed that the pathological transition
from acute to persistent hyperalgesia is associated with
reduced levels of miR- in spinal cord microglia and
with a microglia M phenotype switch, leading to increased
production of proinammatory cytokines. e intrathecal
administration of miR- mimics prevented completely the
transition to persistent pain in LysM-GRK+/− mice and
normalized the expression of microglial M/M markers,
suggesting that miR- administration may be a promising
approach to prevent and treat persistent inammatory and
neuropathic pain [].
Another relevant study on miRNA therapeutics in the
context of neuroinammation was reported by Selvamani
Clinical and Developmental Immunology
SOCS-1 mRNA (fold change)
Control LPS
2
3
4
5
6
1.5
1
0.5
0
∗∗
Oligo scramble
+ LPS
Anti-miR-155
+ LPS
(a)
Cytokine levels (fold change)
70
60
50
40
30
20
10
3
2
1
0
TNF-𝛼IL-1 𝛽IL-6
Control
LPS
Anti-miRl55 + LPS
∗∗∗
∗∗∗
(b)
F : MiR- inhibition in microglia cells increases SOCS- levels and decreases the release of inammatory cytokines to the extracellular
medium. N mouse microglia cells were transfected with anti-miR- oligonucleotides (anti-miR) or with nontargeting oligonucleotides
(oligo scramble) complexed with cationic liposomes for h. Twenty-four hours aer transfection, cells were incubated with LPS at .𝜇g/mL
for  h. e cell culture medium was then collected to determine cytokine protein levels, and total RNA was extracted from the cultured
cells. (a) SOCS- mRNA levels were quantied by qRT-PCR. (b) e levels of TNF-𝛼,IL-𝛽, and IL- secreted to the cultured medium were
determined by ELISA. Results are expressed as fold change of mRNA or protein levels with respect to control (untransfected and untreated
cells). ∗∗𝑃< 0.01 compared to cells transfected with the scramble oligonucleotide and ∗∗∗𝑃< 0.01 compared to LPS-treated cells in the
absence of transfection. Results in (a) and (b) are representative of  independent experiments performed in triplicate.
andcoworkers.eauthorshypothesizedthatmiRNAs
able to target proteins from the insulin-like growth factor-
 (IGF-) signaling family could be suppressed to promote
the neuroprotection provided by IGF- following stroke.
Using middle-age female rats in which the treatment with
estrogen is known to be neurotoxic, the authors administered
stereotactically LNA antisense oligonucleotides against miR-
 or let-f,  h aer stroke, and observed that miRNA
inhibition extended the neuroprotection aorded by IGF-.
Interestingly, although let-f is a proinammatory miRNA
preferentially expressed in microglia from the ischemic hemi-
sphere, where the IGF- expression is detectable, the levels
of IGF- increased even further in microglia aer anti-let-f
treatment []. Finally, a study from Lukiw and coworkers
showed that the expression of miR-a is increased in AD
brains, which was correlated with a decrease in the CFH
expression, a protein responsible for the repression of the
inammatory response in the brain. e inhibition of miR-
, achieved through delivery of a specic LNA-antisense
oligonucleotide in human neural cells subjected to oxidative
stress, was found to restore CFH expression levels, which
weredecreasedfollowingoxidativedamage[].
Taken together, these reports illustrate the vast neuropro-
tective potential of miRNA-based therapeutic strategies using
anti-miRNA oligonucleotides targeting neuroinammation
and conrm the important role of miRNAs in modulating
neuroimmune responses to acute and chronic brain damage.
Although less explored, the application of miRNA mimics,
in order to restore miRNA expression and decrease the
levels of potentially neurotoxic proteins, is also an interesting
possibility with high therapeutic potential. Nevertheless, it is
important to mention that, due to incomplete pairing, each
miRNA has the ability to target multiple genes simultane-
ously, which signicantly increases the risks of unspecic
eects on miRNA-based therapeutic approaches. To avoid
this drawback, it will be relevant to consider not only the
strength of the binding between a certain miRNA and all
its available targets in a specic tissue but also the relative
amounts of these targets and the thermodynamic stability of
each miRNA : mRNA duplex [].
7. Conclusion
Although the exact nature of the contribution of microglia
and peripheral mononuclear phagocytes to neurodegenera-
tion remains to be fully elucidated, several benets have been
suggested for the use of these immune cells in therapeutic
strategies designed to curb amyloidosis, decrease EAE pro-
gression, ght CNS viral infection, or assist in the reduction
of neuroinammation associated with neurodegenerative
diseases. On the other hand, several groups have identied
signs of chronic activation and functional impairment in
microglia cells isolated from dierent brain disease mod-
els. Given their important role in the regulation of gene
expression, we believe that miRNA-based therapies could
constitute an interesting and attractive strategy to improve
Clinical and Developmental Immunology
microglia activity, modulating signaling pathways linked with
neuroinammation. In addition, the compromised activity
of the BBB in most neurodegenerative disorders, the lower
activation threshold of peripheral mononuclear phagocytes
compared to brain-derived microglia, and their easy access in
a clinical context make these cells another excellent tool for
future gene therapy approaches for brain disorders. Overall,
understanding the contribution of brain- and blood-derived
immune cells to microglia pools in the CNS in a disease
context will be of great importance to improve the existing
immunotherapies and generate new and eective therapeutic
strategies for these diseases.
e ne-tuning activity of miRNAs has been proven
crucial in the regulation of dierentiation of microglia allow-
ing the maintenance of brain homeostasis. Since a single
miRNA has the capacity to target more than one protein
involved in the same signaling pathway, their modulation can
signicantly change cell phenotypes that depend on the levels
and activation of specic proteins. Such capacity reects a
molecular paradigm suitable for therapeutic intervention.
Due to the lack of minimally invasive diagnosis tools and
eective therapeutic options for most CNS diseases, we
believethattheuseofmiRNAs,bothasdiseasebiomarkers
andtherapeutictargetsassociatedwithcellsoftheimmune
lineage, although yet poorly explored, will tend to grow in the
near future.
Acknowledgments
e present work was supported by Grants from the
Portuguese Foundation for Science and Technology and
FEDER/COMPETE (PTDC/BIM-MEC// and Pest-
C/SAU/LA/). Joana Guedes and Ana Lu´
ısa Cardoso
are recipients of fellowships from the Portuguese Founda-
tion for Science and Technology (SFRH/BD// and
SFRH/BPD//, resp.).
References
[] J. L. Mar´
ın-Teva, I. Dusart, C. Colin, A. Ger vais, N. Van Rooijen,
and M. Mallat, “Microglia promote the death of developing
Purkinje cells,Neuron,vol.,no.,pp.,.
[] A. Roumier, C. B´
echade, J. C. Poncer et al., “Impaired synaptic
function in the microglial KARAP/DAP-decient mouse,
Journal of Neuroscience, vol. , no. , pp. –, .
[] M.E.Tremblay,R.L.Lowery,andA.K.Majewska,“Microglial
interactions with synapses are modulated by visual experience,
PLoS Biology,vol.,no.,ArticleIDe,.
[] M.Prinz,J.Priller,S.S.Sisodia,andR.M.Ransoho,“Hetero-
geneity of CNS myeloid cells and their roles in neurodegenera-
tion,Nature Neuroscience,vol.,no.,pp.,.
[] T. M. Malm, J. Magga, G. F. Kuh, T. Vatanen, M. Koistinaho, and
J. Koistinaho, “Minocycline reduces engrament and activation
of bone marrow-derived cells but sustains their phagocytic
activity in a mouse model of Alzheimer’s disease,Glia,vol.,
no. , pp. –, .
[] A. Mildner, H. Schmidt, M. Nitsche et al., “Microg lia in the adult
brain arise from Ly-Cℎ𝑖CCR+monocytes only under dened
host conditions,Nature Neuroscience,vol.,no.,pp.
, .
[] F. Ginhoux, M. Greter, M. Leboeuf et al., “Fate mapping
analysis reveals that adult microglia derive from primitive
macrophages,Science,vol.,no.,pp.,.
[] M. Opydo-Chanek and Z. Dabrowski, “Response of astrocytes
and microglia/macrophages to brain injury aer bone marrow
stromal cell transplantation: a quantitative study,Neuroscience
Letters,vol.,no.,pp.,.
[] J.Priller,A.Fl
¨
ugel, T. Wehner et al., “Targeting gene-modied
hematopoietic cells to the central nervous system: use of green
uorescent protein uncovers microglial engrament,Nature
Medicine,vol.,no.,pp.,.
[] A. R. Simard and S. Rivest, “Bone marrow stem cells have the
ability to populate the entire central nervous system into fully
dierentiated parenchymal microglia,FASE B Journal,vol.,
no.,pp.,.
[] M. A. Lynch, “e multifaceted prole of activated microglia,
Molecular Neurobiology, vol. , no. , pp. –, .
[] D. Boche, V. H. Perry, and J. A. Nicoll, “Review: activation
patterns of microglia and their identication in the human
brain,Neuropathology and Applied Neurobiology,vol.,no.,
pp. –, .
[] S. M. Gentleman, P. D. Leclercq, L. Moyes et al., “Long-
term intracerebral inammatory response aer traumatic brain
injury,Forensic Science International,vol.,no.-,pp.
, .
[] Z.S.Vexler,X.N.Tang,andM.A.Yenari,“Inammationin
adult and neonatal stroke,Clinical Neuroscience Research,vol.
, no. , pp. –, .
[] C.A.Colton,R.T.Mott,H.Sharpe,Q.Xu,W.E.VanNostrand,
and M. P. Vitek, “Expression proles for macrophage alternative
activation genes in AD and in mouse models of AD,Journal of
Neuroinammation,vol.,article,.
[] D.H.Cribbs,N.C.Berchtold,V.Perreauetal.,“Extensiveinnate
immune gene activation accompanies brain aging, increasing
vulnerability to cognitive decline and neurodegeneration: a
microarray study,Journal of Neuroinammation,vol.,p.,
.
[] W. S. T. Grin, Jin Gen Sheng, G. W. Roberts, and R. E.
Mrak, “Interleukin- expression in dierent plaque types in
Alzheimer’s disease: signicance in plaque evolution,Journal
of Neuropathology and Experimental Neurology,vol.,no.,
pp.,.
[] A. L. Cardoso, J. R. Guedes, L. Pereira de Almeida, and M.
C. Pedroso de Lima, “miR- modulates microglia-mediated
immune response by down-regulating SOCS- and promoting
cytokine and nitric oxide pro duction,Immunology,vol.,no.
, pp. –, .
[] K. Dutta, D. Ghosh, A. Nazmi, K. lal Kumawat, and A. Basu, “A
common carcinogen benzo[a]pyrene causes neuronal death in
mouse via microglial activation,PLoS ONE,vol.,no.,Article
ID e, .
[]D.Ghosh,M.K.Mishra,S.Das,D.K.Kaushik,andA.
Basu, “Tobacco carcinogen induces microglial activation and
subsequent neuronal damage,Journal of Neurochemistry,vol.
, no. , pp. –, .
[] C. Cunningham, “Microglia and neurodegeneration: the role of
systemic inammation,Glia,vol.,no.,pp.,.
[] S.Arora,R.Rana,A.Chhabra,A.Jaiswal,andV.Rani,“miRNA-
transcription factor interactions: a combinatorial regulation of
 Clinical and Developmental Immunology
gene expression,Molecular Genetics and Genomics,vol.,no.
-, pp. –, .
[] W. Filipowicz, S. N. Bhattacharyya, and N. Sonenberg, “Mecha-
nisms of post-transcriptional regulation by microRNAs: are the
answers in sight? ” Nature Reviews Genetics,vol.,no.,pp.
, .
[] D. P. Bartel, “MicroRNAs: target recognition and regulatory
functions,Cell,vol.,no.,pp.,.
[] M. P. Perron and P. Provost, “Protein interactions and com-
plexes in human microRNA biogenesis and function,Frontiers
in Bioscience,vol.,no.,pp.,.
[] A. M. Denli, B. B. J. Tops, R. H. A. Plasterk, R. F. Ketting,
and G. J. Hannon, “Processing of primary microRNAs by the
microprocessor complex,Nature,vol.,no.,pp.
, .
[]H.Y.Hu,Z.Yan,Y.Xuetal.,“Sequencefeaturesassociated
with microRNA strand selection in humans and ies,BMC
Genomics,vol.,article,.
[] L. A. MacFarlane and P. R. Murphy, “MicroRNA: biogenesis,
function and role in cancer,Current Genomics, vol. , no. ,
pp.,.
[] C. Y. Chen, S. T. Chen, C. S. Fuh, H. F. Juan, and H. C.
Huang, “Coregulation of transcription factors and microRNAs
in human transcriptional regulatory network,BMC Bioinfor-
matics,vol.,no.,articleS,.
[] F. Fazi, A. Rosa, A. Fatica et al., “A minicircuitry comprised of
microRNA- and transcription factors NFI-A and C/EBP𝛼
regulates human granulopoiesis,” Cell,vol.,no.,pp.
, .
[] A. R. R. Forrest, M. Kanamori-Katayama, Y. Tomaru et al.,
“Induction of microRNAs, mir-, mir-, mir- and mir-
, promotes monocytic dierentiation through combinatorial
regulation,Leukemia, vol. , no. , pp. –, .
[] V. Pospisil, K. Vargova, J. Kokavec et al., “Epigenetic silenc-
ing of the oncogenic miR-- cluster during PU. -directed
macrophage dierentiation,e EMBO Journal,vol.,no.,
pp.,.
[] E. D. Ponomarev, T. Veremeyko, N. Barteneva, A. M.
Krichevsky, and H. L. Weiner, “MicroRNA- promotes
microglia quiescence and suppresses EAE by deactivating
macrophages via the C/EBP-𝛼-PU. pathway,” Nature Medicine,
vol.,no.,pp.,.
[] E. D. Ponomarev, T. Veremeyko, and H. L. Weiner, “MicroRNAs
are universal regulators of dierentiation, activation, and polar-
ization of microglia and macrophages in normal and diseased
CNS,Glia,vol.,no.,pp.,.
[] X. Zhou, B. Spittau, and K. Krieglstein, “TGFbeta signalling
plays an important role in IL-induced alternative activation of
microglia,Journal of Neuroinammation,vol.,p.,.
[] S. Bala, M. Marcos, K. Kodys et al., “Up-regulation of
microRNA- in macrophages contributes to increased Tumor
Necrosis Factor 𝛼(TNF𝛼) production via increased mRNA
half-life in alcoholic liver disease,Journal of Biological Chem-
istry,vol.,no.,pp.,.
[] P.Wang,J.Hou,L.Linetal.,“InduciblemicroRNA-feedback
promotes type I IFN signaling in antiviral innate immunity
by targeting suppressor of cytokine signaling ,Journal of
Immunology,vol.,no.,pp.,.
[] E. N. Benveniste and H. Qin, “Type I interferons as anti-
inammatory mediators,Science’s STKE,vol.,no.,
article pe, .
[] A. Dalpke, K. Heeg, H. Bartz, and A. Baetz, “Regulation of
innate immunity by suppressor of cytokine signaling (SOCS)
proteins,Immunobiology,vol.,no.-,pp.,.
[] F. Loua, R. T. Martinez-Nunez, and T. Sanchez-Elsner,
“MicroRNA- targets SMAD and modulates the response
of macrophages to transforming growth factor-𝛽,” Journal of
Biological Chemistry, vol. , no. , pp. –, .
[] D. Ruell, F. Mourkioti, A. Gambardella et al., “A CREB-
C/EBP𝛽cascade induces M macrophage-specic gene expres-
sion and promotes muscle injury repair,Proceedings of the
National Academy of Sciences of the United States of America,
vol. , no. , pp. – ,  .
[] Q. Y. Zhu, L. Qin, C. Jian-Xia, L. Ke, and G. Bao-Xue,
“MicroRNA- targets MAPK phosphatase- to regulate the
activation of MAPKs in macrophages,Journal of Immunology,
vol. , no. , pp. –, .
[] A. A. Chaudhuri, A. Y. So, N. Sinha et al., “MicroRNA-b
potentiates macrophage activation,e Journal of Immunology,
vol. , no. , pp. –, .
[] L. Lai, Y. Song, Y. Liu et al., “MicroRNA-a negatively regu-
lates TLR-triggered inammatory response in macrophages by
targeting MKK,Journal of Biological Chemistry, vol. , no.
, pp. –, .
[] M. Fiala, Q. N. Liu, J. Sayre et al., “Cyclooxygenase--positive
macrophages inltrate the Alzheimer’s disease brain and dam-
age the blood-brain barrier,European Journal of Clinical
Investigation,vol.,no.,pp.,.
[]A.R.Simard,D.Soulet,G.Gowing,J.P.Julien,andS.
Rivest, “Bone marrow-derived microglia play a critical role
in restricting senile plaque formation in Alzheimer’s disease,”
Neuron,vol.,no.,pp.,.
[] K. J. Tracey, “Understanding immunity requires more than
immunology,Nature Immunology,vol.,no.,pp.,
.
[] H. Soreq and Y. Wolf, “NeurimmiRs: microRNAs in the neu-
roimmune interface,Trends in Molecular Medicine,vol.,no.
, pp. –, .
[]W.J.Lukiw,Y.Zhao,andG.C.Jian,“AnNF-𝜅B-sensitive
micro RNA-a-mediated inammatory circuit in alzheimer
disease and in stressed human brain cells,Journal of Biological
Chemistry, vol. , no. , pp. –, .
[] E. Aronica, K. Fluiter, A. Iyer et al., “Expression pattern of miR-
a, an inammation-associated microRNA, in experimental
and human temporal lobe epilepsy,European Journal of Neuro-
science,vol.,no.,pp.,.
[] Y.Y.Li,J.G.Cui,P.Dua,A.I.Pogue,S.Bhattacharjee,and
W. J. Lukiw, “Dierential expression of miRNA-a-regulated
inammatory genes in human primary neural, astroglial and
microglial cells,Neuroscience Letters,vol.,no.,pp.
, .
[] J. Visvanathan, S. Lee, B. Lee, J. W. Lee, and S. K. Lee, “e
microRNA miR- antagonizes the anti-neural REST/SCP
pathway during embryonic CNS development,Genes and
Development,vol.,no.,pp.,.
[] V. Chandrasekar and J. L. Dreyer, “microRNAs miR-, let-
d and miR-a regulate cocaine-induced plasticity,Molecular
and Cellular Neuroscience,vol.,no.,pp.,.
[] K. F´
enelon,J.Mukai,B.Xuetal.,“DeciencyofDgcr,a
gene disrupted by the q. microdeletion, results in altered
short-term plasticity in the prefrontal cortex,” Proceedings of the
National Academy ofSciences of the United States of America,vol.
, no. , pp. –, .
Clinical and Developmental Immunology 
[] S. T. Lee, K. Chu, W. S. Im et al., “Altered microRNA regulation
in Huntington’s disease models,Experimental Neurology,vol.
,no.,pp.,.
[] E. Mart´
ı, L. Pantano, M. Ba˜
nez-Coroneletal.,“Amyriadof
miRNA variants in control and Huntington’s disease brain
regions detected by massively parallel sequencing,Nucleic
Acids Research, vol. , no. , pp. –, .
[] E. Mi˜
nones-Moyano,S.Porta,G.Escaram
´
ıs et al., “MicroRNA
proling of Parkinson’s disease brains identies early downreg-
ulation of miR-b/c which modulate mitochondrial function,”
Human Molecular Genetics,vol.,no.,pp.,.
[] A. H. Williams, G. Valdez, V. Moresi et al., “MicroRNA-
 delays ALS progression and promotes regeneration of
neuromuscular synapses in mice,Science,vol.,no.,pp.
–, .
[] H.Wang,J.Liu,Y.Zongetal.,“MiR-baberrantlyexpressed
in a double transgenic mouse model for Alzheimer’s disease
targets TGF-𝛽type II receptor,Brain Research,vol.,pp.
–, .
[] N. Schonrock, Y. D. Ke, D. Humphreys et al., “Neuronal
microRNA deregulation in response to Alzheimer’s disease
amyloid-beta,PloS ONE,vol.,no.,ArticleIDe,.
[] P. M. Gaughwin, M. Ciesla, N. Lahiri, S. J. Tabrizi, P. Brundin,
and M. Bj¨
orkqvist, “Hsa-miR-b is a plasma-stable microRNA
that is elevated in pre-manifest Huntington’s disease,” Human
Molecular Genetics, vol. , no. , Article ID ddr, pp. –
, .
[] M. Martins, A. Rosa, L. C. Guedes et al., “Convergence of
miRNA expression proling, alpha-synuclein interacton and
GWAS in Parkinson’s disease,PLoS ONE,vol.,no.,Article
ID e, .
[] H. Geekiyanage, G. A. Jicha, P. T. Nelson, and C. Chan, “Blood
serum miRNA: non-invasive biomarkers for Alzheimer’s dis-
ease,Experimental Neurology,vol.,no.,pp.,.
[] P. N. Alexandrov, P. Dua, J. M. Hill et al., “microRNA (miRNA)
speciation in Alzheimer’s disease (AD) cerebrospinal uid
(CSF) and extracellular uid (ECF),International Journal of
Biochemistry and Molecular Biology,vol.,no.,pp.,
.
[] R. Mishra, C. Chhatbar, and S. K. Singh, “HIV- Tat C-mediated
regulation of tumor necrosis factor receptor-associated factor-
by microRNA  in human microglia,” Journal of Neuroinam-
mation,vol.,p.,.
[] S. Rom, I. Rom, G. Passiatore et al., “CCL/MCP- is a target
for mir-a in HIV--infected human microglial cells,FASEB
Journal,vol.,no.,pp.,.
[] R. S. Dave and K. Khalili, “Morphine treatment of human
monocyte-derived macrophages induces dierential miRNA
and protein expression: impact on inammation and oxidative
stress in the central nervous system,Journal of Cellular Bio-
chemistry,vol.,no.,pp.,.
[] L. Zhang, L. Y. Dong, Y. J. Li, Z. Hong, and W. S. Wei, “miR-
 represses FasL in microglia and protects against microglia-
mediated neuronal cell death following hypoxia/ischemia,Glia,
vol. , no. , pp. –, .
[] L. Zhang, L. Y. Dong, Y. J. Li, Z. Hong, and W. S. Wei, “e
microRNA miR-c controls microglia-mediated neuronal
apoptosis by suppressing tumor necrosis factor,Journal of
Neuroinammation,vol.,p.,.
[] R. M. O’Connell, D. Kahn, W. S. J. Gibson et al., “MicroRNA-
promotes autoimmune inammation by enhancing inamma-
tory T cell development,Immunity,vol.,no.,pp.,
.
[] O. Butovsky, S. Siddiqui, G. Gabriely et al., “Modulating inam-
matory monocytes with a unique microRNA gene signature
ameliorates murine ALS,e Journal of Clinical Investigation,
vol. , no. , pp. –, .
[] S. M. Lehmann, C. Kruger, B. Park et al., “An unconventional
role for miRNA: let- activates Toll-like receptor  and causes
neurodegeneration,Nature Neuroscience,vol.,no.,pp.
, .
[] R. Saba, S. Gushue, R. L. Huzarewich et al., “MicroRNA
a (miR-a) is over-expressed during prion disease and
modulates the innate immune response and the microglial
activation state,PLoS ONE,vol.,no.,ArticleIDe,.
[] J.Gao,W.Y.Wang,Y.W.Maoetal.,“Anovelpathwayregulates
memory and plasticity via SIRT and miR-,Nature,vol.,
no.,pp.,.
[] Y.He,C.Yang,C.M.Kirkmire,andZ.J.Wang,“Regulation
of opioid tolerance by let- family microRNA targeting the 𝜇
opioid receptor,JournalofNeuroscience,vol.,no.,pp.
–, .
[] J.A.Hollander,H.I.Im,A.L.Amelioetal.,“StriatalmicroRNA
controls cocaine intake through CREB signalling,Nature,vol.
, no. , pp. –, .
[] P. M. Costa, A. L. Cardoso, C. Nobrega et al., “MicroRNA-
silencing enhances the cytotoxic eect of the antiangiogenic
drug sunitinib in glioblastoma,Human Molecular Genetics,vol.
,no.,pp.,.
[] H.L.Willemen,X.J.Huo,Q.L.Mao-Yingetal.,“MicroRNA-
 as a novel treatment for persistent hyperalgesia,Journal of
Neuroinammation,vol.,p.,.
[] A. Selvamani, P. Sathyan, R. C. Miranda, and F. Sohrabji, “An
antagomir to microRNA Letf promotes neuroprotection in an
ischemic stroke model,PLoS ONE,vol.,no.,ArticleID
e, .
[] M.G.Hemida,X.Ye,S.air,andD.Yang,“Exploitingthether-
apeutic potential of microRNAs in viral diseases: expectations
and limitations,Molecular Diagnosis and erapy,vol.,no.
,pp.,.
... MiR-155 is considered a pro-inflammatory miRNA and has been shown to play a major role in the regulation of the innate immune response by regulating the production of cytokines and chemokines (Thai et al., 2007;Guedes et al., 2013). Previous studies have suggested that miR-155 is one of the key molecules in the inflammatory response of macrophages after TLR activation, and its upregulation depends on the JNK pathway (O'connell et al., 2007). ...
... In the brains of 3xTg AD mice, miR-155 levels were strongly upregulated and coincided with an increase in microglia and astrocyte activation. Guedes et al. (2013Guedes et al. ( , 2014 suggested that miR-155 and c-Jun were upregulated early in 3xTg AD mice and Aβ-activated microglia and astrocytes, thereby promoting the production of inflammatory mediators such as IL-6 and IFN-β (Teter et al., 2019). This effect is related to the miR-155-dependent decrease of SOCS-1. ...
Article
Full-text available
Alzheimer’s disease (AD) is the most common cause of senile dementia. Although AD research has made important breakthroughs, the pathogenesis of this disease remains unclear, and specific AD diagnostic biomarkers and therapeutic strategies are still lacking. Recent studies have demonstrated that neuroinflammation is involved in AD pathogenesis and is closely related to other health effects. MicroRNAs (miRNAs) are a class of endogenous short sequence non-coding RNAs that indirectly inhibit translation or directly degrade messenger RNA (mRNA) by specifically binding to its 3′ untranslated region (UTR). Several broadly expressed miRNAs including miR-21, miR-146a, and miR-155, have now been shown to regulate microglia/astrocytes activation. Other miRNAs, including miR-126 and miR-132, show a progressive link to the neuroinflammatory signaling. Therefore, further studies on these inflamma-miRNAs may shed light on the pathological mechanisms of AD. The differential expression of inflamma-miRNAs (such as miR-29a, miR-125b, and miR-126-5p) in the peripheral circulation may respond to AD progression, similar to inflammation, and therefore may become potential diagnostic biomarkers for AD. Moreover, inflamma-miRNAs could also be promising therapeutic targets for AD treatment. This review provides insights into the role of inflamma-miRNAs in AD, as well as an overview of general inflamma-miRNA biology, their implications in pathophysiology, and their potential roles as biomarkers and therapeutic targets.
... Efek antiinflamasi ditunjukkan oleh AKBA melalui modulasi penurunan ekspresi mi-RNA155 dan meningkatkan ekspresi gen targetnya, yaitu suppressor of cytokine signaling 1 (SOCS-1) (Sayed et al., 2017). mi-RNA155 diketahui menarget SOCS-1 yang berperan sebagai protein antiinflamasi pada mikroglia, yang menyebabkan peningkatan regulasi beberapa jalur inflamasi (Guedes et al., 2013). Selain itu, efek neuroprotektif dari AKBA ditunjukkan dengan penurunan apoptosis melalui penurunan caspcase-3 yang didukung dengan penurunan kondisi stres oksidatif melalui peningkatan SOD, glutathione peroksidase (GPx), dan katalase serta penurunan MDA pada hippocampus (Marefati et al., 2020). ...
Article
Full-text available
Getah kemenyan dengan genus Boswellia telah lama digunakan secara tradisional pada beberapa penyakit. Kandungan Boswellia salah satunya adalah asam boswellic. Asam boswellic memiliki aktivitas neuroprotektif dan antiinflamasi. Beberapa jenis asam boswellic yang paling banyak diteliti mengenai efek farmakologi adalah asam 11-keto-β-boswelik (KBA), asam asetil-11-keto-β-boswellik (AKBA), dan β-boswelik (βBA). βBA diteliti memiliki efektivitas permeabel terhadap Blood Brain Barrier (BBB) dibandingkan jenis asam boswellic lainnya. Oleh karena itu, βBA memiliki potensi yang lebih besar dalam pengaruhnya pada otak terutama hippocampus. Review ini bertujuan untuk memberikan informasi lengkap dan membahas mengenai pengaruh boswellic acid pada performa hippocampus di otak terutama efek pada fungsi kognitif, efek farmakologis pada studi in vivo dan invitro, serta mekanisme molekuler sehingga diharapkan dapat menjadi pengetahuan sebagai acuan dalam melakukan penelitian lanjutan. Pemberian asam boswellic memberikan efek neuroprotektif dan antiinflamasi pada hippocampus sehingga dapat bekerja menghambat apoptosis sel di hippocampus, meningkatkan viabilitas sel saraf, dan meningkatkan kemampuan learning dan memori serta kognitif melalui anti-infalamasi. Beberapa mekanisme molekuler asam boswellic yang dapat mempengaruhi performa hippocampus adalah 5-Lipoxygenase (5-LOX), peningkatan Nuclear factor erythroid 2–related factor 2 (Nrf2), penekanan aktifitas acetylcholinesterase (AChE), dan pengaturan Ca (2+/-) dan protein kinase teraktivasi mitogen (MAPK). Akan tetapi, penelitian mengenai pengaruh boswellic acid pada performa hippocampus masih sangat terbatas dan perlu dilakukan penelitian lebih lanjut terutama pada manusia.
... MCM from M12.CB3 and M16.CB3 melanomas upregulated miR-124-5p. Upregulated expression of this microRNA is associated with the M2 anti-inflammatory, cancer-promoting phenotype of microglia [37] and with a reduction in human M1 pro-inflammatory macrophage functions [38]. In cancer cells, this miRNA serves as a tumor suppressor [39]. ...
Article
Full-text available
Previous studies from our lab demonstrated that the crosstalk between brain-metastasizing melanoma cells and microglia, the macrophage-like cells of the central nervous system, fuels progression to metastasis. In the present study, an in-depth investigation of melanoma-microglia interactions elucidated a pro-metastatic molecular mechanism that drives a vicious melanoma-brain-metastasis cycle. We employed RNA-Sequencing, HTG miRNA whole transcriptome assay, and reverse phase protein arrays (RPPA) to analyze the impact of melanoma-microglia interactions on sustainability and progression of four different human brain-metastasizing melanoma cell lines. Microglia cells exposed to melanoma-derived IL-6 exhibited upregulated levels of STAT3 phosphorylation and SOCS3 expression, which, in turn, promoted melanoma cell viability and metastatic potential. IL-6/STAT3 pathway inhibitors diminished the pro-metastatic functions of microglia and reduced melanoma progression. SOCS3 overexpression in microglia cells evoked microglial support in melanoma brain metastasis by increasing melanoma cell migration and proliferation. Different melanomas exhibited heterogeneity in their microglia-activating capacity as well as in their response to microglia-derived signals. In spite of this reality and based on the results of the present study, we concluded that the activation of the IL-6/STAT3/SOCS3 pathway in microglia is a major mechanism by which reciprocal melanoma-microglia signaling engineers the interacting microglia to reinforce the progression of melanoma brain metastasis. This mechanism may operate differently in different melanomas.
... One of the characteristics of M1 microglia is high expression of miR-155 [156]. Several studies have shown that miR-155 mediates the inflammatory response in ischemic brain tissue by regulating the expression of the Toll-like receptor 4 (TLR4)/ myeloid differentiation factor 88 (MyD88) pathway and suppressor of cytokine signaling protein 1 (SOCS1) [157][158][159]. ...
Article
Full-text available
The most frequent type of stroke, known as ischemic stroke (IS), is a significant global public health issue. The pathological process of IS and post-IS episodes has not yet been fully explored, but neuroinflammation has been identified as one of the key processes. Biomarkers are objective indicators used to assess normal or pathological processes, evaluate responses to treatment, and predict outcomes, and some biomarkers can also be used as therapeutic targets. After IS, various molecules are produced by different cell types, such as microglia, astrocytes, infiltrating leukocytes, endothelial cells, and damaged neurons, that participate in the neuroinflammatory response within the ischemic brain region. These molecules may either promote or inhibit neuroinflammation and may be released into extracellular spaces, including cerebrospinal fluid (CSF) and blood, due to reasons such as BBB damage. These neuroinflammatory molecules should be valued as biomarkers to monitor whether their expression levels in the blood, CSF, and brain correlate with the diagnosis and prognosis of IS patients or whether they have potential as therapeutic targets. In addition, although some molecules do not directly participate in the process of neuroinflammation, they have been reported to have potential diagnostic or therapeutic value against post-IS neuroinflammation, and these molecules will also be listed. In this review, we summarize the neuroinflammatory biomarkers in the brain, CSF, and blood after an IS episode and the potential value of these biomarkers for the diagnosis, treatment, and prognosis of IS patients.
... On the other hand, in the presence of strong inflammatory stimuli, microglia assume an M1 phenotype, characterized by high levels of miR-155 expression. Upregulation of miR-155 is believed to be crucial for the establishment of this phenotype, since this miRNA acts directly on anti-inflammatory molecules, such as SOCS-1, leading to over-regulation of the various inflammatory mediators characteristic of the M1 phenotype [50]. ...
Article
Full-text available
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression at the post-transcriptional level and that play an important role in many cellular processes, including modulation of inflammation. MiRNAs are present in high concentrations in the central nervous system (CNS) and are spatially and temporally expressed in a specific way. Therefore, an imbalance in the expression pattern of these small molecules can be involved in the development of neurological diseases. Generally, CNS responds to damage or disease through the activation of an inflammatory response, but many neurological disorders are characterized by uncontrolled neuroinflammation. Many studies support the involvement of miRNAs in the activation or inhibition of inflammatory signaling and in the promotion of uncontrolled neuroinflammation with pathological consequences. MiR-155 is a pro-inflammatory mediator of the CNS and plays an important regulatory role. The purpose of this review is to summarize how miR-155 is regulated and the pathological consequences of its deregulation during neuroinflammatory disorders, including multiple sclerosis, Alzheimer’s disease and other neuroinflammatory disorders. Modulation of miRNAs’ expression could be used as a therapeutic strategy in the treatment of pathological neuroinflammation.
... Microglia can differentiate into either M1 (pro-inflammatory) or M2 (anti-inflammatory) phenotypes depending on the provoking signals. M2 microglia release anti-inflammatory and protective cytokines such as IL-10, TGF-β, IL-4, and IL-13, which promote repair (Guedes et al., 2013). Whereas, M1 microglia release inflammatory mediators such as ROS, MMP-9 and pro-inflammatory cytokines such as TNFα, IL-6 and IL-1β.The balance between these different microglial phenotypic states promotes inflammation or tissue repair and influences the progression of neuroinflammatory disorders (Guedes et al., 2014). ...
Article
Full-text available
Alzheimer's disease (AD) is a progressive and deleterious neurodegenerative disease, strongly affecting the cognitive functions and memory of seniors worldwide. Around 58% of the affected patients live in low and middle-income countries, with estimates of increasing deaths caused by AD in the coming decade. AD is a multifactor pathology. Mitochondrial function declines in AD brain and is currently emerging as a hallmark of this disease. It has been considered as one of the intracellular processes severely compromised in AD. Many mitochondrial parameters decline already during aging; mitochondrial efficiency for energy production, reactive oxygen species (ROS) metabolism and the de novo synthesis of pyrimidines, to reach an extensive functional failure, concomitant with the onset of neurodegenerative conditions. Besides its impact on cognitive functions, AD is characterized by loss of synapses, extracellular amyloid plaques composed of the amyloid-β peptide (Aβ), and intracellular aggregates of hyperphosphorylated Tau protein, accompanied by drastic sleep disorders, sensory function alterations and pain sensitization. Unfortunately, till date, effective management of AD-related disorders and early, non-invasive AD diagnostic markers are yet to be found. MicroRNAs (miRNAs) are small non-coding nucleic acids that regulate key signaling pathway(s) in various disease conditions. About 70% of experimentally detectable miRNAs are expressed in the brain where they regulate neurite outgrowth, dendritic spine morphology, and synaptic plasticity. Increasing studies suggest that miRNAs are intimately involved in synaptic function and specific signals during memory formation. This has been the pivotal key for considering miRNAs crucial molecules to be studied in AD. MicroRNAs dysfunctions are increasingly acknowledged as a pivotal contributor in AD via deregulating genes involved in AD pathogenesis. Moreover, miRNAs have been proved to control pain sensitization processes and regulate circadian clock system that affects the sleep process. Interestingly, the differential expression of miRNA panels implies their emerging potential as diagnostic AD biomarkers. In this review, we will present an updated analysis of miRNAs role in regulating signaling processes that are involved in AD-related pathologies. We will discuss the current challenges against wider use of miRNAs and the future promising capabilities of miRNAs as diagnostic and therapeutic means for better management of AD.
... It is well known that chronic neuroinflammation is one of the prominent hypotheses put forward to describe the pathogenesis of AD [17]. Several miRNA networks, including miRNAs related with innate immunity and neuroinflammation, have been found to be dysregulated in AD [18,19]. ...
Article
Full-text available
Age-related disorders, such as Alzheimer's disease (AD) and age-related macular degeneration (AMD) share common features such as amyloid-β (Aβ) protein accumulation. Retinal deposition of Aβ aggregates in AMD patients has suggested a potential link between AMD and AD. In the present study, we analyzed the expression pattern of a focused set of miRNAs, previously found to be involved in both AD and AMD, in the retina of a triple transgenic mouse model of AD (3xTg-AD) at different time-points. Several miRNAs were differentially expressed in the retina of 3xTg-AD mice, compared to the retina of age-matched wild-type (WT) mice. In particular, bioinformatic analysis revealed that miR-155 had a central role in miRNA-gene network stability, regulating several pathways, including apoptotic and inflammatory signaling pathways modulated by TNF-related apoptosis-inducing ligand (TNFSF10). We showed that chronic treatment of 3xTg-AD mice with an anti-TNFSF10 monoclonal antibody was able to inhibit the retinal expression of miR-155, which inversely correlated with the expression of its molecular target SOCS-1. Moreover, the fine-tuned mechanism related to TNFSF10 immunoneutralization was tightly linked to modulation of TNFSF10 itself and its death receptor TNFRSF10B, along with cytokine production by microglia, reactive gliosis, and specific AD-related neuropathological hallmarks (i.e., Aβ deposition and Tau phosphorylation) in the retina of 3xTg-AD mice. In conclusion, immunoneutralization of TNFSF10 significantly preserved the retinal tissue in 3xTg-AD mice, suggesting its potential therapeutic application in retinal degenerative disorders. Cell Death and Disease (2021) 12:905 ; https://doi.
Article
Full-text available
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by complex pathophysiological features. Amyloid plaques resulting from extracellular amyloid deposition and neurofibrillary tangles formed by intracellular hyperphosphorylated tau accumulation serve as primary neuropathological criteria for AD diagnosis. The activation of microglia has been closely associated with these pathological manifestations. Non-coding RNA (ncRNA), a versatile molecule involved in various cellular functions such as genetic information storage and transport, as well as catalysis of biochemical reactions, plays a crucial role in microglial activation. This review aims to investigate the regulatory role of ncRNAs in protein expression by directly targeting genes, proteins, and interactions. Furthermore, it explores the ability of ncRNAs to modulate inflammatory pathways, influence the expression of inflammatory factors, and regulate microglia activation, all of which contribute to neuroinflammation and AD. However, there are still significant controversies surrounding microglial activation and polarization. The categorization into M1 and M2 phenotypes may oversimplify the intricate and multifaceted regulatory processes in microglial response to neuroinflammation. Limited research has been conducted on the role of ncRNAs in regulating microglial activation and inducing distinct polarization states in the context of neuroinflammation. Moreover, the regulatory mechanisms through which ncRNAs govern microglial function continue to be refined. The current understanding of ncRNA regulatory pathways involved in microglial activation remains incomplete and may be influenced by spatial, temporal, and tissue-specific factors. Therefore, further in-depth investigations are warranted. In conclusion, there are ongoing debates and uncertainties regarding the activation and polarization of microglial cells, particularly concerning the categorization into M1 and M2 phenotypes. The study of ncRNA regulation in microglial activation and polarization, as well as its mechanisms, is still in its early stages and requires further investigation. However, this review offers new insights and opportunities for therapeutic approaches in AD. The development of ncRNA-based drugs may hold promise as a new direction in AD treatment.
Article
Full-text available
Alzheimer's disease (AD) is the most common form of senile dementia and is characterized by progressive cognitive impairment and neuronal degeneration. Microglial activation is an important pathologic hallmark of AD. During disease progression, microglial cells switch from an alternative or anti‐inflammatory and neuroprotective profile (M2) to a classic or proinflammatory and neurotoxic profile (M1). Phenotypically, M1 microglia is characterized by the activation of inflammatory signaling pathways that cause increased expression of proinflammatory genes, including those coding for cytokines and chemokines. This microglia‐mediated neuroinflammation contributes to neuronal cell death. Recent studies in microglial cells have shown that a group of plant‐derived compounds, known as flavonoids, possess anti‐inflammatory properties and therefore exert a neuroprotective effect through regulating microglia activation. Here, we discuss how flavonoids can promote the switch from an inflammatory M1 phenotype to an anti‐inflammatory M2 phenotype in microglia and how this represents a valuable opportunity for the development of novel therapeutic strategies to blunt neuroinflammation and boost neuronal recovery in AD. We also review how certain flavonoids can inhibit neuroinflammation through their action on the expression of microglia‐specific microRNAs (miRNAs), which also constitute a key therapeutic approach in different neuropathologies involving an inflammatory component, including AD. Finally, we propose novel targets of microglia‐specific miRNAs that may be considered for AD treatment. Review of how flavonoids control microglial activation states through regulation of microRNAs and the implications in Alzheimer's disease appearance and progression.
Article
Microglial activation followed by recruitment of blood-borne macrophages into the central nervous system (CNS) aggravates neuroinflammation. Specifically, in multiple sclerosis (MS) as well as in experimental autoimmune encephalomyelitis (EAE), a rodent model of MS, activated microglia and macrophages (Mg/Mφ) promote proinflammatory responses and expand demyelination in the CNS. However, a potent therapeutic approach through the systemic route for regulating their functions has not yet been developed. Here, we demonstrated that a systemically injected DNA/RNA heteroduplex oligonucleotide (HDO), composed of an antisense oligonucleotide (ASO) and its complementary RNA, conjugated to cholesterol (Chol-HDO) distributed more efficiently to demyelinating lesions of the spinal cord in EAE mice with significant gene silencing than the parent ASO. Importantly, systemic administration of Cd40-targeting Chol-HDO improved clinical signs of EAE with significant downregulation of Cd40 in Mg/Mφ. Furthermore, we successfully identified that macrophage scavenger receptor 1 (MSR1) is responsible for the uptake of Chol-HDO by Mg/Mφ of EAE mice. Overall, our findings demonstrate the therapeutic potency of systemically administered Chol-HDO to regulate activated Mg/Mφ in neuroinflammation.
Article
Full-text available
Toll-like receptors (TLRs) play a critical role in the initiation of immune responses against invading pathogens. MicroRNAs have been shown to be important regulators of TLR signaling. In this study, we have found that the stimulation of multiple TLRs rapidly reduced the levels of microRNA-92a (miRNA-92a) and some other members of the miRNA-92a family in macrophages. miR-92a mimics significantly decreased, whereas miR-92a knockdown increased, the activation of the JNK/c-Jun pathway and the production of inflammatory cytokines in macrophages when stimulated with ligands for TLR4. Furthermore, mitogen-activated protein kinase kinase 4 (MKK4), a kinase that activates JNK/stress-activated protein kinase, was found to be directly targeted by miR-92a. Similar to the effects of the miR-92a mimics, knockdown of MKK4 inhibited the activation of JNK/c-Jun signaling and the production of TNF-α and IL-6. In conclusion, we have demonstrated that TLR-mediated miR-92a reduction feedback enhances TLR-triggered production of inflammatory cytokines in macrophages, thus outlining new mechanisms for fine-tuning the TLR-triggered inflammatory response.
Article
Full-text available
Developmental processes require a precise spatio-temporal regulation of gene expression wherein a diverse set of transcription factors control the signalling pathways. MicroRNAs (miRNAs), a class of small non-coding RNA molecules have recently drawn attention for their prominent role in development and disease. These tiny sequences are essential for regulation of processes, including cell signalling, cell development, cell death, cell proliferation, patterning and differentiation. The consequence of gene regulation by miRNAs is similar to that by transcription factors (TFs). A regulatory cascade essential for appropriate execution of several biological events is triggered through a combinatorial action of miRNAs and TFs. These two important regulators share similar regulatory logics and bring about a cooperative action in the gene regulatory network, dependent on the binding sites present on the target gene. The review addresses the biogenesis and nomenclature of miRNAs, outlines the mechanism of action and regulation of their expression, and focuses on the combinatorial action of miRNAs and TFs for the expression of genes in various regulatory cascades.
Article
Full-text available
Highly malignant glioblastoma (GBM) is characterized by high genetic heterogeneity and infiltrative brain invasion patterns, and aberrant miRNA expression has been associated with hallmark malignant properties of glioblastoma. The lack of effective GBM treatment options prompted us to investigate whether miRNAs would constitute promising therapeutic targets towards the generation of a gene therapy approach with clinical significance for this disease. Here, we show that microRNA-21 (miR-21) is upregulated and microRNA-128 (miR-128) is downregulated in mouse and human glioblastoma samples, a finding that is corroborated by analysis of a large set of human GBM data from The Cancer Genome Atlas. Moreover, we demonstrate that oligonucleotide-mediated miR-21 silencing in U87 human GBM cells resulted in increased levels of the tumor suppressors PTEN and PDCD4, caspase 3/7 activation and decreased tumor cell proliferation. Cell exposure to pifithrin, an inhibitor of p53 transcriptional activity, reduced the caspase activity associated with decreased miR-21 expression. Finally, we demonstrate for the first time that miR-21 silencing enhances the anti-tumoral effect of the tyrosine kinase inhibitor sunitinib, whereas no therapeutic benefit is observed when coupling miR-21 silencing with the first line drug temozolomide. Overall, our results provide evidence that miR-21 is uniformly overexpressed in glioblastoma and constitutes a highly promising target for multimodal therapeutic approaches towards GBM.
Article
Full-text available
Background Post-ischemic microglial activation may contribute to neuronal damage through the release of large amounts of pro-inflammatory cytokines and neurotoxic factors. The involvement of microRNAs (miRNAs) in the pathogenesis of disorders related to the brain and central nervous system has been previously studied, but it remains unknown whether the production of pro-inflammatory cytokines is regulated by miRNAs. Methods BV-2 and primary rat microglial cells were activated by exposure to oxygen-glucose deprivation (OGD). Global cerebral ischemia was induced using the four-vessel occlusion (4-VO) model in rats. Induction of pro-inflammatory and neurotoxic factors, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide (NO), were assessed by ELISA, immunofluorescence, and the Griess assay, respectively. The miRNA expression profiles of OGD-activated BV-2 cells were subsequently compared with the profiles of resting cells in a miRNA microarray. BV-2 and primary rat microglial cells were transfected with miR-181c to evaluate its effects on TNF-α production after OGD. In addition, a luciferase reporter assay was conducted to confirm whether TNF-α is a direct target of miR-181c. Results OGD induced BV-2 microglial activation in vitro, as indicated by the overproduction of TNF-α, IL-1β, and NO. Global cerebral ischemia/reperfusion injury induced microglial activation and the release of pro-inflammatory cytokines in the hippocampus. OGD also downregulated miR-181c expression and upregulated TNF-α expression. Overproduction of TNF-α after OGD-induced microglial activation provoked neuronal apoptosis, whereas the ectopic expression of miR-181c partially protected neurons from cell death caused by OGD-activated microglia. RNAinterference-mediated knockdown of TNF-α phenocopied the effect of miR-181c-mediated neuronal protection, whereas overexpression of TNF-α blocked the miR-181c-dependent suppression of apoptosis. Further studies showed that miR-181c could directly target the 3′-untranslated region of TNF-α mRNA, suppressing its mRNA and protein expression. Conclusions Our data suggest a potential role for miR-181c in the regulation of TNF-α expression after ischemia/hypoxia and microglia-mediated neuronal injury.
Article
Full-text available
Background Microglia are the resident immune cells of the central nervous system and are accepted to be involved in a variety of neurodegenerative diseases. Several studies have demonstrated that microglia, like peripheral macrophages, exhibit two entirely different functional activation states, referred to as classical (M1) and alternative (M2) activation. TGFβ is one of the most important anti-inflammatory cytokines and its effect on inhibiting microglia or macrophage classical activation has been extensively studied. However, the role of TGFβ during alternative activation of microglia has not been described yet. Methods To investigate the role of TGFβ in IL4-induced microglia alternative activation, both, BV2 as well as primary microglia from new born C57BL/6 mice were used. Quantitative RT-PCR and western blots were performed to detect mRNA and protein levels of the alternative activation markers Arginase1 (Arg1) and Chitinase 3-like 3 (Ym1) after treatment with IL4, TGFβ or both. Endogenous TGFβ release after IL4 treatment was evaluated using the mink lung epithelial cell (MLEC) assay and a direct TGFβ2 ELISA. TGFβ receptor type I inhibitor and MAPK inhibitor were applied to address the involvement of TGFβ signalling and MAPK signalling in IL4-induced alternative activation of microglia. Results TGFβ enhances IL4-induced microglia alternative activation by strongly increasing the expression of Arg1 and Ym1. This synergistic effect on Arg1 induction is almost completely blocked by the application of the MAPK inhibitor, PD98059. Further, treatment of primary microglia with IL4 increased the expression and secretion of TGFβ2, suggesting an involvement of endogenous TGFβ in IL4-mediated microglia activation process. Moreover, IL4-mediated induction of Arg1 and Ym1 is impaired after blocking the TGFβ receptor I indicating that IL4-induced microglia alternative activation is dependent on active TGFβ signalling. Interestingly, treatment of primary microglia with TGFβ alone results in up regulation of the IL4 receptor alpha, indicating that TGFβ increases the sensitivity of microglia for IL4 signals. Conclusions Taken together, our data reveal a new role for TGFβ during IL4-induced alternative activation of microglia and consolidate the essential functions of TGFβ as an anti-inflammatory molecule and immunoregulatory factor for microglia.
Article
Full-text available
A growing body of evidence suggests that microRNA (miRNA) dysregulation contributes to many types of human disease, including central nervous system disorders. In this study, we identified an inverse correlation between the expression of miR-21 and Fas ligand (FasL) during hypoxia-induced microglial activation. Specifically, hypoxia caused the upregulation of FasL expression but the downregulation of miR-21 expression in microglia. Furthermore, we demonstrated that miR-21 suppresses FasL production by directly binding to its 3'-untranslated region. The overproduction of FasL following hypoxic microglial activation induced neuronal apoptosis, whereas the ectopic expression of miR-21 partially protected neurons from cell death caused by hypoxia-activated microglia. Finally, we confirmed that the function of miR-21 in microglia-mediated neuronal injury is dependent on FasL. Our study demonstrates an important role for miRNAs in microglia-mediated neuronal apoptosis, and suggests potential novel therapeutic interventions for cerebral hypoxic diseases associated with microglial activation. © 2012 Wiley Periodicals, Inc.
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) is a progressive disease associated with neuronal cell death that is thought to involve aberrant immune responses. Here we investigated the role of innate immunity in a mouse model of ALS. We found that inflammatory monocytes were activated and that their progressive recruitment to the spinal cord, but not brain, correlated with neuronal loss. We also found a decrease in resident microglia in the spinal cord with disease progression. Prior to disease onset, splenic Ly6Chi monocytes expressed a polarized macrophage phenotype (M1 signature), which included increased levels of chemokine receptor CCR2. As disease onset neared, microglia expressed increased CCL2 and other chemotaxis-associated molecules, which led to the recruitment of monocytes to the CNS by spinal cord-derived microglia. Treatment with anti-Ly6C mAb modulated the Ly6Chi monocyte cytokine profile, reduced monocyte recruitment to the spinal cord, diminished neuronal loss, and extended survival. In humans with ALS, the analogous monocytes (CD14+CD16-) exhibited an ALS-specific microRNA inflammatory signature similar to that observed in the ALS mouse model, linking the animal model and the human disease. Thus, the profile of monocytes in ALS patients may serve as a biomarker for disease stage or progression. Our results suggest that recruitment of inflammatory monocytes plays an important role in disease progression and that modulation of these cells is a potential therapeutic approach.
Article
The histologically apparent polymorphism of plaques containing [beta]-amyloid in Alzheimer's disease is thought to represent different stages in plaque evlution. [beta]-amyloid-immunopostitive plaques were classified according to the pattern of [beta]-amyloid distribution (diffuse vs dense-core) and the presence or absence of dystrophic [beta]-amyloid precursor protein-immunopositive ([beta]-APP+) neurites (neuritic vs non-neuritic). The potential contribution of microgilia-derived interleukin-1 (IL-1), an immune response cytokine that induces synthesis and processing of [beta]-APP, to the possible sequential development of these plaque types was examined through determination of the number of IL-1[alpha]+ microglia associated with each of four identified plaque types. Diffuse non-neuritic plaques had the least dense and most widely dispersed [beta]-amyloid, did not exhibit [beta]-APP+ dystrophic neurites, but most (78%) contained activated IL-1[alpha]+ microglia (2 [gamma] 0.2/Plaque; mean [gamma] SEM). Diffuse neuritic plaques had more dense, but still widely dispersed [beta]-amyloid, displayed a profusion of [beta]-APP+ dystrophic neurites, and had the greatest numbers of associated activated IL-1[alpha]+ microglia (4 [gamma] 0.4/plaque). Dense-core, non-neuritic plaques had compact [beta]-amyloid, lacked associated diffuse [beta]-amyloid, and were devoid of both IL-1[alpha]+ microglia and [beta]-APP+ dystrophic neurites. These results suggest an important immunological component in the evolution of amyloid-contining plaques in Alzheimer's disease and further suggest that IL-1-expressing cells are necessary to intitiate dystrophic neurite formation in diffuse [beta]-amyloid deposits. Our data indicate that activation of microglia with expression of IL-1 in Alzheimer's disease is required to drive the metamorphosis of diffuse non-neuritic [beta]-amyloid deposits to the characteristic and diagnostic neuritic plaques of Alzheimer's disease.
Article
Human cerebrospinal fluid (CSF), produced by the choroid plexus and secreted into the brain ventricles and subarachnoid space, plays critical roles in intra-cerebral transport and the biophysical and immune protection of the brain. CSF composition provides valuable insight into soluble pathogenic bio-markers that may be diagnostic for brain disease. In these experiments we analyzed amyloid beta (Aβ) peptide and micro RNA (miRNA) abundance in CSF and in short post-mortem interval (PMI <2.1 hr) brain tissue-derived extracellular fluid (ECF) from Alzheimer's disease (AD) and age-matched control neocortex. There was a trend for decreased abundance of Aβ42 in the CSF and ECF in AD but it did not reach statistical significance (mean age ~72 yr; N=12; p~0.06, ANOVA). The most abundant nucleic acids in AD CSF and ECF were miRNAs, and their speciation and inducibility were studied further. Fluorescent miRNA-array-based analysis indicated significant increases in miRNA-9, miRNA-125b, miRNA-146a, miRNA-155 in AD CSF and ECF (N=12; p<0.01, ANOVA). Primary human neuronal-glial (HNG) cell co-cultures stressed with AD-derived ECF also displayed an up-regulation of these miRNAs, an effect that was quenched using the anti-NF-кB agents caffeic acid phenethyl ester (CAPE) or 1-fluoro-2-[2-(4-methoxy-phenyl)-ethenyl]-benzene (CAY10512). Increases in miRNAs were confirmed independently using a highly sensitive LED-Northern dot-blot assay. Several of these NF-кB-sensitive miRNAs are known to be up-regulated in AD brain, and associate with the progressive spreading of inflammatory neurodegeneration. The results indicate that miRNA-9, miRNA-125b, miRNA-146a and miRNA-155 are CSF- and ECF-abundant, NF-кB-sensitive pro-inflammatory miRNAs, and their enrichment in circulating CSF and ECF suggest that they may be involved in the modulation or proliferation of miRNA-triggered pathogenic signaling throughout the brain and central nervous system (CNS).
Article
Microglia in the central nervous system are usually maintained in a quiescent state. When activated, they can perform many diverse functions which may be either beneficial or harmful depending on the situation. Although microglial activation may be accompanied by changes in morphology, morphological changes cannot accurately predict the function being undertaken by a microglial cell. Studies of peripheral macrophages and in vitro and animal studies of microglia have resulted in the definition of specific activation states: M1 (classical activation) and M2 (sometimes sub-divided into alternative activation and acquired deactivation). Some authors have suggested that these might be an overlapping continuum of functions rather than discrete categories. In this review, we consider translational aspects of our knowledge of microglia: specifically, we discuss the question as to what extent different activation states of microglia exist in the human central nervous system, which tools can be used to identify them and emerging evidence for such changes in ageing and in Alzheimer's disease.