ArticlePDF Available

Neuroprotective Effects of Dantrolene in Neurodegenerative Disease: Role of Inhibition of Pathological Inflammation

Authors:

Abstract and Figures

Neurodegenerative diseases (NDs) refer to a group of diseases in which slow, continuous cell death is the main pathogenic event in the nervous system. Most NDs are characterized by cognitive dysfunction or progressive motor dysfunction. Treatments of NDs mainly target alleviating symptoms, and most NDs do not have disease-modifying drugs. The pathogenesis of NDs involves inflammation and apoptosis mediated by mitochondrial dysfunction. Dantrolene, approved by the US Food and Drug Administration, acts as a RyRs antagonist for the treatment of malignant hyperthermia, spasticity, neuroleptic syndrome, ecstasy intoxication and exertional heat stroke with tolerable side effects. Recently, dantrolene has also shown therapeutic effects in some NDs. Its neuroprotective mechanisms include the reduction of excitotoxicity, apoptosis and neuroinflammation. In summary, dantrolene can be considered as a potential therapeutic candidate for NDs.
Content may be subject to copyright.
Journal Pre-proof
Neuroprotective Effects of Dantrolene in
Neurodegenerative Disease: Role of Inhibition of
Pathological Inflammation
Wenjia Zhang, Xu Zhao, Piplu Bhuiyan, Henry
Liu, Huafeng Wei
PII: S2957-3912(24)00057-3
DOI: https://doi.org/10.1016/j.jatmed.2024.04.002
Reference: JATMED7
To appear in: Journal of Anesthesia and Translational Medicine
Please cite this article as: Wenjia Zhang, Xu Zhao, Piplu Bhuiyan, Henry Liu and
Huafeng Wei, Neuroprotective Effects of Dantrolene in Neurodegenerative
Disease: Role of Inhibition of Pathological Inflammation, Journal of Anesthesia
and Translational Medicine, (2024)
doi:https://doi.org/10.1016/j.jatmed.2024.04.002
This is a PDF file of an article that has undergone enhancements after acceptance,
such as the addition of a cover page and metadata, and formatting for readability,
but it is not yet the definitive version of record. This version will undergo
additional copyediting, typesetting and review before it is published in its final
form, but we are providing this version to give early visibility of the article.
Please note that, during the production process, errors may be discovered which
could affect the content, and all legal disclaimers that apply to the journal pertain.
© 2024 Published by Elsevier.
Neuroprotective Effects of Dantrolene in Neurodegenerative Disease:
Role of Inhibition of Pathological Inflammation
Wenjia Zhang1,2, Xu Zhao 2, Piplu Bhuiyan 1,
Henry Liu 1, Huafeng Wei 1
1Department of Anesthesiology and Critical Care
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA 19104, USA
2Department of Anesthesiology
Shandong Provincial Hospital
Shandong First Medical University
Jinan, Shandong, 250021, China
Corresponding Author:
Huafeng Wei, MD, PhD
Department of Anesthesiology and Critical Care
University of Pennsylvania Perelman School of Medicine
305 John Morgan Building
3620 Hamilton Walk
Philadelphia, PA 19104
Phone: 215-2609840, FAX: 215-349-507
Email: huafeng.wei@pennmedicine.upenn.edu
Abstract
Neurodegenerative diseases (NDs) refer to a group of diseases in which slow,
continuous cell death is the main pathogenic event in the nervous system. Most
Journal Pre-proof
NDs are characterized by cognitive dysfunction or progressive motor
dysfunction. Treatments of NDs mainly target alleviating symptoms, and most NDs do
not have disease-modifying drugs. The pathogenesis of NDs involves inflammation
and apoptosis mediated by mitochondrial dysfunction. Dantrolene, approved by
the US Food and Drug Administration, acts as a RyRs antagonist for the
treatment of malignant hyperthermia, spasticity, neuroleptic syndrome, ecstasy
intoxication and exertional heat stroke with tolerable side effects. Recently,
dantrolene has also shown therapeutic effects in some NDs. Its neuroprotective
mechanisms include the reduction of excitotoxicity, apoptosis and
neuroinflammation. In summary, dantrolene can be considered as a potential
therapeutic candidate for NDs.
Keywords:
Alzheimer's disease, cognitive dysfunction, progressive motor dysfunction,
apoptosis, calcium, pyroptosis
Introduction
Neurodegenerative diseases (NDs) refer to a group of diseases
characterized by progressive and chronic cell death within the nervous system,
manifesting clinically as symptoms reflective of the pathophysiological changes
caused by neuronal loss [1]. Many diseases can be classified into NDs. The
prototypical examples include Parkinson's disease (PD), Alzheimer's disease
(AD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and
multiple sclerosis (MS). Aging is the biggest risk factor for most NDs [2].
Common NDs and their estimated prevalence, as reported in primary literature,
are shown in Table 1.
Journal Pre-proof
Table 1 Prevalence and major symptoms of common NDs
Disease
Prevalence
Major Symptoms
Alzheimer's
disease
(AD)
6.7 million age 65 and
older in the US in 2023[2]
Impairment of learning and
memory, speech difficulties
Parkinson's
disease
(PD)
23% of the global
population aged
>65 years in 2017 [3]
Muscle tremor, rigidity,
bradykinesia/akinesia, and
postural instability
Huntington's
disease
(HD)
10.613.7 per 100 000 in
Western countries. 17 per
million in Japan, Taiwan,
and Hong Kong in 2017 [4]
Chorea, dystonia, loss of
coordination, cognitive
decline,
behavioral difficulties
Amyotrophic
lateral
sclerosis
(ALS)
Prevalence rates (per
100,000 persons) and
incidence rates (per
100,000 person-years) are
6.22 and 2.31 for Europe,
5.20 and 2.35 for North
America, 3.41 and 1.25 for
Latin America, 3.01 and
0.93 for Asian countries
excluding Japan, and 7.96
and 1.76 for Japan,
respectively.[5]
Progressive motor defects,
with
muscle weakness, atrophy
and
spasms
Multiple
sclerosis
(MS)
2.8 million people
worldwide in 2021[6]
Blurred vision, weak limbs,
tingling sensations,
unsteadiness, and fatigue
Most NDs are characterized by cognitive dysfunction or progressive motor
dysfunction, thus placing an increasing burden on caregivers and economy.
More importantly, PD stands as the the only treatable ND currently with the
individualized treatment objectives, emphasizing the need of personalized
management. Levodopa serves as the primary first-line treatment. Achieving
optimal treatment begins with diagnosis and requires a multidisciplinary
approach that includes a number of nonpharmacological interventions [7]. All
other NDs are incurable. To date, two types of drugs have received approval
Journal Pre-proof
for AD, including cholinesterase inhibitors and the N - methyl - D - aspartate
(NMDA) receptor antagonist. However, these medications only alleviate the
symptoms of AD and do not provide a cure or preventive measures against the
disease itself. [8]. Potential interventions for HD include therapies targeting
huntingtin DNA and RNA, clearance of huntingtin protein, DNA repair pathways,
and other treatment strategies targeting inflammation and cell death [9]. For
ALS, survival can be improved through supportive and symptomatic care
delivered by a multidisciplinary team of experts. This approach may involve
expert consensus guidelines and an evidence-based approach to treatment.
[10]. The greatest challenge in MS lies in developing therapies that combine
neuroprotection and remyelination to treat, and ultimately disable, the
progression of the disease [11]. Currently, many disease-modifying therapies
for patients in early aggressive courses have shown variable efficacy in
preventing recurrence, lesion accumulation on magnetic resonance imaging
(MRI), and disability progression [12].
Dantrolene, a Food and Drug Administration (FDA)-approved drug for the
treatment of malignant hyperthermia, has recently been evaluated for
prospective use as a neuroprotective agent for the treatment of
neurodegenerative syndromes including AD [13]. Researchers have
demonstrated that dantrolene improves memory loss and neuropathology in
different animal models of AD [14, 15]. Through in-vitro experiments,
researchers have also demonstrated that dantrolene ameliorated the
impairments in neurogenesis and synaptogenesis [16]. Recent studies also
show dantrolene’s therapeutic effects in different NDs including AD [14, 15] and
HD [80]. The potential mechanisms by which dantrolene protects neurons are
decreasing excitotoxicity, apoptosis, and neuroinflammation.
Pathogenesis of NDs and potential therapeutic pathways
Although NDs are heterogeneous and contain a series of different diseases,
the study of genetic defects and disease-related proteins indicates that different
NDs share similar features, mechanisms, and even genetic or protein
abnormalities [17].
Journal Pre-proof
There are two main mechanisms that may explain this challenging process.
One is an inflammation-mediated immune response, and the other is apoptosis,
a form of programmed cell death. The two mechanisms are implicated in the
pathogenesis of NDs, including AD, PD, HD, MS, and ALS [18, 19].
To date, mitochondrial dysfunction has been thought to contribute to
inflammation and apoptosis in NDs through the accumulation of mitochondrial
DNA (mtDNA) mutations and the generation of reactive oxygen species (ROS)
[17, 20]. Specific interactions of disease-related proteins with mitochondria
have recently been uncovered: APP, β-amyloid (Aβ), presenilin, α-synuclein,
parkin, DJ-1, PINK1 (phosphatase and tension homologue-induced kinase 1),
LRRK2 (leucine-rich-repeat kinase 2), HTRA2, SOD1 and huntingtin have all
been demonstrated to localize within the mitochondria [20] [21].
The pathology of inflammation and mitochondrial dysfunction-mediated
apoptosis in AD is characterized by the accumulation of amyloid β-containing
neurotic plaques, neurofibrillary tangles, and dystrophic neurites containing
hyperphosphorylated tau protein. Microglia, the resident macrophages in the
brain, become activated by pathological events such as neuronal death or
protein aggregates. Once activated, they can bind to oligomers and fibrils
via cell surface receptors. SCARA1, CD36, CD14, α6β1 integrin, CD47, and
Toll-like receptors (TLR2, TLR4, TLR6, and TLR9) are receptors. The process
of microglia activation is thought to be part of the inflammatory reaction in AD
[22]. Inefficient clearance of has been identified as a major pathogenic
pathway in sporadic cases of AD [22]. By decreasing the expression of
phagocytosis receptors, microglia show insufficient phagocytic capacity which
is thought to be responsible for increased cytokine concentration. Reduced
mitophagy process enhances neuroinflammation due to reduced phagocytosis
and over stimulation of microglia, leading to increased accumulation of
plaques and hyperphosphorylated tau protein [22, 23]. In addition, the
researchers showed that in TgAPPsw and PSAPP transgenic mice, the
increases of concentration were highly correlated with increased
concentration of proinflammatory cytokines, including TNFα, interleukin 6 (IL-
6), interleukin (IL-1α), and granulocyte-macrophage colony stimulating
factor (GM-CSF) [22].
Journal Pre-proof
Oxidative damage in the brain of AD transgenic APP mice proceeds the
occurrence of Aβ deposition and significant plaque pathological changes. The
signaling pathways of APP or tau processing may be activated by oxidative
stress. In vitro, studies have shown that exposure to Aβ increases oxidative
stress and decreases energy availability, thereby increasing the susceptibility
of cell death by inducing apoptosis. However, evidence regarding the role of
apoptosis in neuronal cell death in AD remains limited [24, 25].
Inflammation and mitochondrial dysfunction-mediated apoptosis are
important pathologies of PD. The pathological feature of PD is the presence of
Lewy bodies, resulting from α-synuclein aggregation and dopaminergic
neurodegeneration in the substantia nigra pars compacta. Dopaminergic
neurons are selectively vulnerable to mitochondrial dysfunction. Subsequently,
the mitochondrial PT pore opens, resulting in the release of cytochrome c into
the cytoplasm, triggering the mitochondria-mediated apoptotic pathway, which
is considered to be the primary mechanism of dopaminergic neuronal death in
PD. MPTP (1-methyl 4-phenyl-1,2,3,6-tetrahydropyridine), whose metabolite
MPP+ inhibits complex I of the mitochondrial electron-transport chain, causing
parkinsonism in designer-drug abusers [26, 27].
Another important pathogenesis is pro-inflammatory immune-mediated
mechanisms. Results from animal models confirmed the activation of microglia
and the upregulation of pro-inflammatory mediators such as cytokines found in
postmortem PD brains [18]. Antigens such as infectious pathogens, prions,
pathologically modified proteins, aggregates, and apoptotic cells can activate
microglia. Activated microglia subsequently secrete a wide range of
inflammatory mediators, such as TNFα, IL-6, nitric oxide synthase 2 (NOS2),
COX2, and ROS, leading to cell proliferation, followed by slow degeneration,
and finally the death of dopaminergic neurons [28]. Furthermore, clinical studies
in PD patients examined the CSF and peripheral blood, showing elevated
serum IL-1, IL-6, and TGF-β in CSF [29]. Recently, mitochondrial dysfunction
has been proven to induce neuroinflammation [18].
Journal Pre-proof
Inflammation and mitochondrial dysfunction also mediate apoptosis in HD.
HD is caused by mutations in an abnormally expanded CAG trinucleotide
repeat in a gene encoding the polyglutamine repeats in the huntingtin protein.
Inflammation in HD can be triggered by glial cells responses to autonomous
neuronal degeneration or immune cells activation due to mHtt (mutant HTT
protein) expression, or both at the same time [30]. The main inflammatory
processes are carried out by activated microglia and astrocytes. Reactive
astrocytes are observed in presymptomatic HD and correlate with disease
severity [31].
Studies have shown that the activity of complexes II and III of the electron-
transport chain is reduced in the brains of HD patients. Additionally,
mitochondrial respiration and ATP production were also significantly impaired
in striatal cells in mutant HTT-knock-in mouse embryos. Moreover, cytochrome
c discharge, BAX overexpression, and active caspase-3, -8, and -9 have been
illustrated in the brains of HD patients and in experimental models of HD [32].
These evidence suggest that mitochondrial dysfunction-mediated apoptosis
plays an important role in the pathogenesis of HD.
Inflammation and mitochondrial dysfunction mediate apoptosis in ALS.
ALS is recognized by the progressive and specific loss of motor neurons in the
cortex and the ventral horn of the spinal cord. Studies in ALS patients and
animal models have shown that activated microglia and astroglia,
proinflammatory peripheral lymphocytes, and macrophages are associated
with ALS [33]. The anti-inflammatory response is initially protective, but
subsequently, inflammatory cytotoxic cytokines (IL-1β, IL-6, IL-18, TNF-α etc.)
and reactive oxygen species (ROS) can lead to cell death and further tissue
damage [34]. In recent years, an increasing number of studies have revealed
that both CNS and peripheral immune cells release extracellular vesicles, which
can regulate the behavior of neighboring receptor cells and play an essential
role in the pathogenesis of ALS [33].
The spinal cord, nerve, and muscle biopsy samples showed abnormal
mitochondria structures and numbers. The study focused on the expression of
mutant SOD1 in animal and cellular models of ALS. In fact, SOD1 plays a vital
Journal Pre-proof
role in the clearance of ROS, and the abnormal activity of mutant human SOD1
(mSOD1) leads to oxidative damage [35]. Transgenic mice overexpressing the
G93A Sod1 mutation exhibit impaired mitochondrial energy metabolism in the
brain and spinal cord during disease onset. The illustration of DNA
fragmentation has suggested the role of apoptosis in ALS pathogenesis,
Caspase 9 activation, cytochrome c discharge, BAX overexpression, and
reduced Bcl-2 expression in postmortem tissue and transgenic mouse models
of ALS[36-38].
Inflammation and mitochondrial dysfunction mediated apoptosis in MS.
The pathological hallmark of all MS phenotypes is focal plaques (also called
lesions), which are areas of demyelination typically located around post-
capillary venules and characterized by breakdown of the blood-brain barrier
(BBB). The mechanisms underlying BBB breakdown are not fully understood
but seem to involve direct action of pro-inflammatory cytokines and chemokines
(such as TNF-α, IL-1β ,and IL-6) produced by resident and endothelial cells, as
well as indirect cytokine-dependent and chemokine-dependent leukocyte-
mediated damage [39]. BBB dysregulation increases the trans-endothelial
migration of activated leukocytes, including macrophages, T cells, and B cells,
into the CNS. This process leads to further inflammation and demyelination,
resulting in oligodendrocyte loss, reactive gliosis, and neuro-axonal
degeneration[39].
In addition to inflammatory mechanism, researchers are currently focusing
on the role of mitochondrial damage in demyelination and neurodegeneration.
Biochemical studies identified impaired NADH dehydrogenase activity and
increased complex IV activity in mitochondria in MS lesions[40]. Furthermore,
in oligodendrocytes, mitochondrial damage results in the release of apoptosis-
inducing factor, its translocation into the nuclei, and the activation of poly-ADP-
ribose polymerase (PARP), a mechanism demonstrated in oligodendrocyte
destruction and demyelination induced by cuprizone intoxication in an
experiment in vivo [40].
Journal Pre-proof
Therapeutic pathways
Given the growing evidence supporting the role of inflammation and
mitochondrial dysfunction-mediated apoptosis in the pathogenesis of various
NDs, treatments involving anti-inflammation and improving mitochondrial
function may serve as potential therapeutic approaches.
1. Immunomodulatory therapies
Experimental evidence and animal studies showed that non-steroidal anti-
inflammatory drugs (NSAIDs), particularly ibuprofen and piroxicam have
promising results and appeared to reduce PD risk [18]. Other potential
immunomodulatory therapies, such as anti-TNF therapies, are based on in vitro
studies and need more in vivo evidence[18]. Drugs such as PPARγ agonists,
COX inhibitors, vitamin E, vitamin C, curcumin and catechin, which reduce the
neuroinflammatory pathways, are thought to prevent the progression of AD[23].
Preclinical studies in HD mouse models have shown that active immunity is
achieved by the introduction of an immunogenic short peptide. This peptide
generates host B cell-mediated antigen-specific antibodies against exon1 of
mutant HTT protein. Consequently, this approach downregulates aberrant
neuroinflammatory and cell death pathways[31]. Minocycline, which can
suppress microglial activation and modulate apoptosis, was shown to reduce
motor neurons loss, delay disease onset, and extend the survival of SOD1G93A
mice. NP001, which has been shown to be safe and well tolerated, can slow
disease progression by modulating monocyte activation and downregulating
NF-kB in macrophages[41].
2. Inhibition of mitochondrial cytochrome c release
Minocycline can also inhibit mitochondrial cytochrome c release, which is
an essential step in the progression of the mitochondria-mediated apoptotic
pathway. Moreover, it displays broad neuroprotection in experimental models
of NDs[42, 43]
3. The use of antioxidants
One of the most studied antioxidant compounds is mitoquinone (MitoQ),
which exerts direct antioxidant action by scavenging superoxide, peroxyl, and
Journal Pre-proof
peroxynitrite ROS[44]. In an AD transgenic mouse model expressing three
human mutant genes, APP, PSEN1 and tau, MitoQ treatment showed improved
behavioral phenotype. Other antioxidants, such as Skulachev (SkQ1), MitoApo,
melatonin, have also been reported to show neuroprotective effects in different
research models [45-48].
4. Overexpression of the anti-apoptotic protein Bcl-2
CGP 3466 is a drug that ultimately results in the upregulation of anti-
apoptotic Bcl-2. CGP 3466 has been reported to rescue dopaminergic neurons
from death and to subsequently inhibit the development of motor symptoms in
rodent models of PD by preventing mitochondria-mediated apoptosis[49].
Additionally, Bcl-2 overexpression and the deletion of pro-apoptotic BAX and
BAK have been shown to exert neuroprotection in ALS mice by blocking the
apoptosis of lumbar spinal motor neurons and delaying the onset of
symptoms[50].
Dantrolene Pharmacology
Dantrolene is the only effective treatment for malignant hyperthermia (MH),
a fatal condition during general anesthesia. Before the advent of dantrolene,
the mortality rate of MH was reported as high as 80%[51]. Dantrolene was
approved by the Food and Drug Administration (FDA) in 1979, and can
effectively reverse the symptoms and reduce the mortality to less than 5%[52].
Mechanism of action
The ryanodine receptors (RyRs) are major intracellular Ca 2+ release
channels located in the plasma membrane of the endoplasmic/sarcoplasmic
reticulum. Three mammalian isoforms of RyRs have been isolated. Type 1
RyRs (RyR-1) is preferentially expressed in skeletal muscle and in cerebellar
Purkinje neurons. Type 2 RyRs (RyR-2) is predominantly expressed in cardiac
muscle and is the most abundant isoform in the brain. Type 3 RyRs (RyR-3)
was first identified in the brain and is mainly found in cortical and hippocampal
regions involved in learning and memory[53]. Both RyR-1 and RyR-3 are the
targets of dantrolene in vivo, while previously RYR2 in cardiac SR vesicles was
thought insensitive to clinical concentrations of dantrolene[54]. A recent in vitro
Journal Pre-proof
study shows that the inhibition of dantrolene to RyR2 complex is very sensitive
but depends on the presence of both FKBP12.6(a regulatory protein) and
calmodulin (CaM)[55].
The hypermetabolic symptoms of MH are due to the abnormal release of
the Ca2+ from the sarcoplasmic reticulum (SR) via RyR-1 in skeletal
muscles[56]. Symptoms of chronic ischemia heart disease including ventricular
tachycardia, left-ventricular remodeling, and contractile dysfunction are highly
associated with the RyR-2 hyperactivity [57]. Dantrolene is a ryanodine receptor
antagonist that acts directly on the RyR-1 and RyR-3 to reduce channel
activation by CaM. Thereby it decreases the high Ca2+ concentration that leads
to sustained pathological muscle contraction[51, 53, 58]. Dantrolene also
stabilizes RyR-2 tetrameric structure and improves survival after myocardial
infarction[59].
Chemistry and Pharmacokinetics
Dantrolene is highly lipophilic and therefore poorly soluble in water.
Dantrolene can now be administered intravenously. One bottle of 20 mg
dantrolene sodium contains 3g mannitol to improve water solubility. The powder
should be dissolved in 60 ml water to achieve a concentration of 0.33mg/ml,
and adjusted the final pH to 9.5.
In 2014, , FDA approved Ryanodex, a new IV formulation of dantrolene
nanoparticles. Ryanodex (Eagle)[60] contains 250 mg of dantrolene as a
lyophilized powder. Each vial of powder requires only 5ml of sterile water (SWI)
to reconstitute for injection, while other previously approved formulations
required 60ml of SWI to reconstitute a 20 mg dose. Ryanodex contains less
mannitol, so patients may need to supplement with mannitol to produce more
urine[60].
Approximately 70% of dantrolene is absorbed after oral administration, with
a peak plasma time of six hours. However, plasma concentration varies
significantly from patient to patient, especially in children[61].The plasma
elimination half-life is estimated to be 12 hours. In children, the pharmacokinetic
profile is similar, with a half-life of approximately 10 hours [62]. Hepatic
Journal Pre-proof
microsomes metabolize Dantrolene to 5-hydroxydantrolene. Dantrolene and its
metabolites are mainly excreted through urine and bile[63, 64].
Therapeutic Uses
In addition to malignant hyperthermia, dantrolene is also used to treat
other diseases in clinical work.
Muscle Spasm
The first approved use of oral dantrolene was for the treatment of
spasticity[65]. Spasticity is a common symptom of many neurological disorders,
such as cerebral infarction and MS[66]. Non-drug treatments for spasticity
include passive movements, exercises, posture and standing. Commonly used
anti-spasmodic drugs are those that act on the gamma aminobutyric acid
(GABA) energic system (baclofen, gabapentin, and benzodiazepines), the α-2
adrenergic system (tizanidine) Unlike other drugs that treat spasm, dantrolene
works directly on the muscle and so is less sedative.
Neuroleptic malignant syndrome
The incidence of neuroleptic malignant syndrome (NMS) has been reported
to be rare, and the most obvious clinical features of NMS are muscle rigidity
and hyperthermia. As the cause of altered thermoregulation remains unclear,
skeletal muscle contraction may generate heat. Therefore, paralytics will be a
key cooling measures[67]. Dantrolene, along with dopamine agonists such as
bromocriptine and amantadine, may provide benefits. However, these findings
are solely based on case reports due to the absence of controlled clinical
trials[68].
Ecstasy intoxication
One of the crucial signs of ecstasy intoxication is hyperthermia, which may
be caused by overstimulation of central serotonin. Dantrolene has been
proposed as a potentially effective treatment for ecstasy poisoning in
emergency rooms. However, recent animal studies suggest that dantrolene
sodium formulations are mechanistically unsuitable for the treatment of MDMA-
and METH-induced hyperthermia[69]
Journal Pre-proof
Exertional heat stroke
Exertional heat stroke (EHS) is an acute injury with high morbidity and
mortality that is common in military and special operations environments. The
most effective way to treat EHS is immediately cooling. Dantrolene is
administered to the victim to prevent further muscle contractions and
hyperthermia. However, these data come from case reports. Whether
dantrolene treatment is beneficial for EHS still requires randomized, controlled
clinical studies[70].
Adverse Effects
Adverse effects may occur following acute or chronic parenteral
administration of dantrolene. The most common side effect was muscle
weakness (22%), followed by phlebitis (10%), respiratory failure (3%), and
gastrointestinal discomfort (3%). Other adverse symptoms of dantrolene
treatment include drowsiness, dizziness, and confusion[61]. Long-term oral
therapy at high dose has been associated with liver dysfunction, but dantrolene
is not the only potentially hepatotoxic substance administered to patients
reported to have this complication [65].
Dantrolene in Neurodegenerative Diseases
Dantrolene’s anti-inflammation effect
In recent years, research on application of dantrolene in different diseases
has been studied, including in vitro cell models and in vivo animal models.
Recent evidence suggests that disruption of intracellular Ca2+ homeostasis
resulting from overreaction of RyRs on the ER and/or SR, plays a crucial role
in sepsis and COVID-19-induced inflammation[71, 72]. Dantrolene, as a RyRs
antagonist, has been expected to improve SARS-CoV-2-mediated
inflammation and inhibit SIRS during sepsis by suppressing the concentration
of IL-6, IL-8, IL-1β, TNF-α, and IFN-γ3 in plasma and tissues[71, 72].
Wenk and his colleagues infused LPS, a commonly used inflammation
inducer, into the brain of 3-months-old F-344 AD rat for 28 days. At the
beginning of the inflammation induction, rats were treated subcutaneously with
Journal Pre-proof
dantrolene (5 mg/kg/day) or nimodipine (5 mg/kg/day). They found that
treatment with nimodipine or dantrolene restored the number of TH-
immunoreactive cells in the locus coeruleus (LC) and significantly reduced
microglia activation in the hippocampus[73] and substantia nigra pars
compacta[74]. Furthermore, dantrolene treatment decreased inflammatory
gene expression of TLR4, iNOS, and TGFβ in the hippocampus, resulting in
neurotoxicity[73].
In another study of autoimmune disease, Fomina’s team used a mouse
model of experimental autoimmune encephalomyelitis (EAE), a T cell-mediated
autoimmune neuroinflammatory disease. They found that daily intraperitoneal
injection of dantrolene at 5 or 10 mg/kg beginning at the time of EAE induction
significantly reduced dampened inflammation in the spinal cord[75]. These
findings indicate that dantrolene has anti-inflammation effects in CNS.
Dantrolene’s therapeutic effects in AD and HD
In 2012, Professor Wei’s lab first reported the use of dantrolene for early
and long-term treatment in a murine AD model. Their team published a series
of articles on long-term treatment with dantrolene in both 3xTg and 5XFAD
mouse models[14, 15]. Their research also revealed that dantrolene reduced
amyloid accumulation in neurons by up to 76% compared to the vehicle control
and tended to reduce hippocampal phosphorylated tau protein[76]. Their
studies showed that dantrolene can ameliorate cognitive dysfunction in vivo and
has tolerable side effects with long-term use (treatment up to 10 months)[14].
In in vitro experiments on pluripotent stem cells, they found that dantrolene
could ameliorates the impairment of neurogenesis and synaptogenesis[16].
In recent years, impaired neural-lysosomal and autophagy-mediated
degradation of cellular debris has been suggested to contribute to neuritic
dystrophy and synaptic loss. Mustaly-Kalimi’s group demonstrated that
dantrolene can restore autophagic clearance of intracellular protein aggregates
by increasing vATPase levels, lysosomal acidification, and proteolytic activity
Journal Pre-proof
in human iPSC-derived neurons from AD patients. Their work showed that
abnormal upstream Ca2+ dysregulation contributes to the pathological
accumulation of intracellular protein aggregates before the development of
overt histopathological or cognitive deficits in AD [77]. Studies also found that
dantrolene preserved synaptic plasticity. Zhang et al. used PS1-M146V knock-
in (KI) FAD mice with impaired long-term LTP maintenance (L-LTP). They found
that dantrolene could reverse insufficient mushroom spines maintenance in KI
neurons[78].
Besides these exciting findings, there are also controversial results. A study
genetically inhibiting RyRs expression in APPPS1 mice showed that blocking
RyR-3 was beneficial in older AD neurons while causing more synaptic
dysfunction in young AD neurons[56].
Altogether, these studies of dantrolene in AD are inspiring, but the
mechanisms by which dantrolene as a neuroprotective agent still require further
investigation.
HD is a progressive neurodegenerative disease caused by polyglutamine
amplification in the Huntington protein, resulting in selective degeneration of
spine neurons in the striatum. Bezprozvanny’s team fed yeast artificial
chromosome transgenic mice (YAC128), a model of HD, dantrolene (5 mg/kg)
twice a week from 2 months to 11.5 months of age. Their results showed that
the treatment group performed significantly better on beam-walking and gait-
walking tests. Furthermore, they performed neuropathological analysis on
YAC128 mice and found that the loss of NeuN-positive striatal neurons was
significantly reduced after long-term feeding of dantrolene[79].
Michael S and his colleagues combined whole-cell patch clamp and Two-
Photon Ca2+ Imaging to record action potential-evoked Ca2+ transients in
cortical pyramidal neurons (CPNs) in the R6/2 mouse model of juvenile HD at
different stages of disease progression. They demonstrated that in the
presence of dantrolene, the amplitude and area of Ca2+ transients were
significantly reduced compared with WT CPNs. The results suggest that
Journal Pre-proof
dantrolene is a potential therapeutic agent for HD.
Potential mechanism of dantrolene ’s neuroprotective effects
Figure 1. Pyroptosis. Glutamate excitotoxicity result in pathological and
excessive activation of NMDAR and/or AMPAR, leading to pathological Ca2+
influx into cytosol space, increasing cytosolic and mitochondia Ca2+
concentration, mitochondrial reactive oxygen species (ROS) and then cytosol
ROS. Over activation of RyR results in excessive Ca2+ release from ER and
subsequent ER stress. The upstream Ca2+ dysregulation eventually results in
activation of NLRP3 inflammasome, activated caspase-1 and cleaved GSDMD
to generate N terminal GSDMD and releasing IL-1beta and IL-18 to generate
pathological inflammation.
RyRs are located on the sarco/endoplasmic reticulum (SR/ER) of nearly all
cell types. As we summarized previously, the mechanism of NDs is cell death
caused by mitochondrial dysfunction. Increasing studies show that aberrant
Ca2+ release from intracellular stores (SR/ER) is thought to be an upstream
cause of mitochondrial dysfunction. Therefore, aberrant Ca2+ release from
intracellular stores appears to be a key point.
Calcium is an extremely important signaling ion, transmitting cell signals for
various external stimuli. Under physiological conditions, more than 99% of
Journal Pre-proof
intracellular Ca2+ is bound to cytosolic proteins or stored in the ER. Free
cytosolic Ca2+ is normally maintained at approximately 100 nm, but stimulation
can lead to an overall increase to approximately 1 μM[81]. The mechanisms
by which free cytosolic Ca2+ rises are Ca2+ entry through plasma membrane
channels and intracellular pools, in which ER plays an important role. As early
as 1961, Engstrom’s group first discovered that mitochondria could rapidly and
efficiently take up large amounts of Ca2+ when exposed to Ca2+ pulses, [82] . In
resting neurons, mitochondrial total Ca2+ and free Ca2+ levels are low, estimated
to be approximately 0.1 mM and 100 nM, respectively. However, upon
stimulation, mitochondria are able to accumulate enormous amounts of Ca2+[83,
84]. Elevated Ca2+ in mitochondria has many physiological effects, which are
especially important for cellular function. When Ca2+ concentration increases,
aerobic ATP production will be adjusted, and ROS production will be regulated
accordingly. Additionally, elevated Ca2+ contributes to synaptic transmission
and excitability, organelle dynamics, and activation of the release of death
signals[85, 86]. Accordingly, dantrolene (a RyRs inhibitor), has been shown to
block the release of ER Ca2+ stores and partially protect neurons from oxygen-
glucose deprivation toxicity [87]. Inhibiting the release of ER Ca2+ stores may
be one of the important mechanisms of dantrolene’s neuroprotective effects.
Chronic inflammation leads to excess Ca2+ to be transferred from the ERs
to mitochondria, causing mitochondrial calcium overload and further damaging
mitochondria[88]. Moreover, calpains are intracellular Ca2+- dependent
cysteine proteases that play a physiologic role by cleaving several substrates.
Overactivation of calpains can lead to changes in hippocampal structure and
function, and is linked to neuronal death. Furthermore, calpain is overactivated
by increased Ca2+ concentrations[89]. Hence, inhibiting the release of Ca2+
from the SRs may be one of the mechanisms by which dantrolene suppresses
the neuroinflammation.
Glutamate is the primary excitatory neurotransmitter in CNS, and one of the
glutamate receptors is NMDAR, which plays an important role in excitotoxic
Journal Pre-proof
injury. Under physiological conditions, once NMDAR are activated, Na+ and
Ca2+ flow will be permitted, which is essential for normal synaptic transmission
as well as for a wide range of Ca2+-dependent signaling pathways. However,
significantly increased glutamate concentration, such as in stroke, leads to loss
of ion homeostasis and necrotic cell death by triggering massive NMDAR
stimulation [90]. The elevation of intracellular Ca2+ concentration triggers the
release of Ca2+ from ER through RyRs on ER[91]. Elevated intracellular Ca2+
levels lead to mitochondrial Ca2+ overload, which initiates a neurotoxic cascade
through multiple mechanisms, including mitochondrial membranes rupture,
ROS generation and release of cytochrome c and other proapoptotic factors[92].
Ultimately these mitochondrial-related events result in necrotic or apoptotic-like
cell death[93]. Furthermore, emerging studies show that abnormal Ca2+ release
from the ER/SR through RyRs Ca2+ channels play an important role in
neuroinflammation[94].
Taken together, dantrolene, a RyRs antagonist acts as a neuroprotective
agent through multiple mechanisms proposed below.
. Dantrolene protects neurons by decreasing excitotoxicity
In in vitro experiment on mouse cerebral cortex neurons, dantrolene reduced
glutamate-induced increases in intracellular Ca2+ by 70% under physiological
conditions, protecting neurons from glutamate-induced neurotoxicity[95]. In
another animal experiment, dantrolene significantly reduced the ischemia-
induced increase in glutamate concentration and prevented neuronal loss in the
CA1 region of the rat hippocampus [96]. In several other in vitro experiments,
dantrolene has been shown to inhibit activation of the NMDA receptors in
multiple different cell lines [97-99].
. Dantrolene protects neurons by decreasing apoptosis
Several animal models have showed neuroprotective effect through apoptosis
mechanism. In an in vivo experiment using repeated electroconvulsions as a
rat status epilepticus model, the TUNEL method was used to detect cell
Journal Pre-proof
apoptosis. More TUNEL (+) neurons were observed in the hippocampus of the
dantrolene-treated group compared with the control group[100]. In another rat
spinal cord injury model, the author combined riluzole and dantrolene for
treatment; they found that rats treated with a combination of riluzole and
dantrolene showed a greater number of NeuN-positive neurons, suggesting
that dantrolene prevents neuronal apoptosis [101].
An in vitro experiment using neuron-like PC12 cells demonstrated that
dantrolene, as well as 2-aminoethoxydiphenyl borate (2-APB), another RyRs
and InsP3 receptor (InsP3R) inhibitor, protect PC12 cells from H2O2-induced
apoptosis and activate autophagic pathways[102].
. Dantrolene protects neurons by decreasing neuroinflammation
An increasing number of studies, including different animal models of NDs
show that Pro-inflammatory markers are significantly reduced after treatment
with dantrolene as we mentioned before. The proposed mechanisms by which
dantrolene decreases neuroinflammation include the following approaches.
1. Dantrolene reduces neuroinflammation by inhibiting microglia activation
Neuroinflammation is characterized by the production of pro-inflammatory
cytokines, including IL-1β, IL-6, IL-18 and tumor necrosis factor (TNF), in which
microglia play an important role. Once the microglia are activated by
pathological factors such as apoptotic neurons or infectious pathogens, they
produce increased amounts of pro-inflammatory cytokines[103]. Furthermore,
the insufficient phagocytic capacity of activated microglia leads to the inefficient
clearance of protein aggregates such as Aβ in AD[22]. Activated microglia can
also directly eliminate synaptic structures[103]. The emerging role of microglial
activation in the pathogenesis of NDs makes microglia a therapeutic target.
Ca2+ is required for LPS-mediated microglia activation in vitro, and the
application of Ca2+ chelators is sufficient to prevent activation and production of
proinflammatory cytokines[104]. A study has shown that nitrogen-doped
graphene quantum dots (N-GQDs) can induce intracellular calcium overload by
Journal Pre-proof
activating calcium channels of L VGCCs and RyRs, thereby activating mouse
hippocampal microglia. [105]. These evidences revealed that RyRs and L-
VDCCs participated in mediating Ca2+-related microglia activation. Microglia
express mRNA for RyR1 and RyR2 isoforms, and application of RyRs
antagonists prevents microglia-mediated LPS-induced neurotoxicity [106].
Another study demonstrated that dantrolene significantly reduced the number
of activated microglia in the hippocampus and reduced the expression of
various pro-inflammatory cytokines[73]. Taken together, dantrolene decrease
microglia activation by reducing intracellular calcium overload through RyRs
channels.
2. Dantrolene reduces neuroinflammation by inhibiting pro-inflammatory
cytokines
As we summarized previously, most NDs are characterized by a
pathological inflammatory response. The inflammatory response is initially
protective, but subsequently, inflammatory cytokines such as TNFα, IL-6,
interleukin 1α(IL-1α), and GM-CSF[22] lead to cell death and tissue damage
[34]. Intracellular Ca2+ signaling and the elevation of intracellular Ca2+ are
critical for the release of pro-inflammatory cytokines [71]. Furthermore, calcium
channels contribute to the release of pro-inflammatory cytokines in NDs. A
recent study showed that knockdown of transient receptor potential vanilloid
4(TRPV4), a nonselective Ca2+ channel, can reduce the concentration of pro-
inflammatory cytokines including IL-18, COX-2, and 5-LOX in PD mice[107].
The calcium channel blocker carvedilol has been demonstrated to decrease the
release of pro-inflammatory cytokine TNF-α IL-2 and IL-6[108]. Many research
groups also reported that VGCCs and RyRs play an important role in
neuroinflammation[68, 109].
Dantrolene, a calcium channel blocker with the ability to ameliorate Ca2+
dysregulation by inhibiting RyRs, has been discovered to suppress plasma and
tissue concentration of IL-6, IL-8, IL-1β, TNF-α, and IFN-γ36 in vivo and in
Journal Pre-proof
vitro[110-113]. Taken together, dantrolene inhibited ER-mediated Ca2+ release
and reduced pro-inflammatory cytokine release.
3. Dantrolene reduces neuroinflammation by ameliorating oxidative stress
Oxidative stress (OS) and neuroinflammation are two different but
fundamental pathological factors that play important roles in the onset and
progression of NDs [114]. Inflammatory cells such as microglia and astrocytes
secrete reactive substances that promote OS [114]. Some ROS and RNS
(reactive nitrogen species) can reversely stimulate intracellular signaling
cascades, resulting in an increased release of pro-inflammatory cytokines.
Therefore, neuroinflammation and OS can promote each other, especially in
the NDs state.
Given that oxidative stress plays a crucial role in NDs by causing cell death
and promoting neuroinflammation, antioxidant drugs have been a potential
treatment for NDs. Dantrolene has been reported to protect cells from oxidative
stress by increasing the concentration of GSH and GSH/GSSG [115, 116].
Furthermore, calcium influx through RyRs on ER is associated with ROS
generation [117].
4. Dantrolene reduces neuroinflammation by ameliorating pyroptosis
Pyroptosis is a type of programmed cell death during which the
inflammatory cytokines are released. Pyroptosis is mediated by GSDMD-N (N-
terminal fragment of gasdermin D) which binds to membranes to form
membrane pores and promotes the release of inflammatory cytokines,
especially IL-. NLRP3 inflammasome activation plays a leading role in the
initiation of this process. Pyroptosis is closely related to neuroinflammatory
diseases, including subarachnoid hemorrhage, spinal cord injury [118] and PD
[119]. Recent studies have shown that ER stress and excessive Ca2+ release
are associated with NLRP3 inflammasome activation. A recent study shows
that restriction of Ca2+ release in the ER decreases the NLRP3 inflammasome
Journal Pre-proof
activation[118]. Dantrolene, as a RyRs antagonist, can also inhibit Ca2+ release
in the ER and therefore, potentially ameliorate pyroptosis by inhibiting NLRP3
inflammasome activation.
Future trends in NDs and potential application of dantrolene
In summary, the pathogenesis of NDs primarily involves
neuroinflammation and apoptosis mediated by mitochondrial dysfunction. To
date there are no disease-modified drugs. On the one hand, dantrolene shows
anti-inflammatory effect in a variety of NDs; on the other hand, there are
disease-modified drugs for the treatment of AD. All these evidences suggest
that dantrolene, as a potential anti-inflammation drug, can be considered as a
potential therapeutic drug for the treatment of different NDs.
CRediT authorship statement
Conceptualization: Wenila Zhang, Huafeng Wei; Data curation: Xu Zhao ;
Formal analysis: Xu Zhao ; Investigation: Piplu Bhuiyan ; Methodology: Piplu
Bhuiyan; Project administration: Huafeng Wei; Supervision: Huafeng Wei;
Validation: Huafeng Wei; Writing original draft: Wenila Zhang; Writing
review & editing: Henry Liu. All authors have read and agreed to the published
version of the manuscript.
Disclosure statement
Not applicable.
Ethical statement
Not applicable.
Data availability statement
All study data are included in the article.
Funding
This work was supported by grants to HW from the National Institution of
Aging (R01AG061447, 3R01AG061447- 03S1).
Journal Pre-proof
Acknowledgments
The author would like to thank Luna Sato and Yutong Yi for English editing
the manuscript.
Declaration of competing interests
Henry Liu holds the position of Editor-in-Chief for JATM, and was blinded from
reviewing or making decisions for the manuscript. Dr. Huafeng Wei is one of
the inventors for patents application titled "Intranasal dantrolene for treatment
of Alzheimer's disease" owned by the University of Pennsylvania Trustee in
following countries: International (PCTUS2020/040198, 2020), USA
(62/868,820, 2019), Europe (20833145.4-1112, 2022), Japan (2021-577376,
2021), China (202080054348.2, 2022), Hong Kong (62022053033.60, 2020),
South Korea (10-2022-7003375, 2020), Singapore (11202114347V, 2020),
Brazil (BR1120210265970, 2020), Mexico (MX/a/2022/000231, 2022). The
authors declare that they have no conflicts of interest.
References:
[1]. Masi, A., R. Narducci and G. Mannaioni, Harnessing ionic mechanisms to achieve disease
modification in neurodegenerative disorders. Pharmacol Res, 2019. 147: p. 104343.
[2]. 2023 Alzheimer's disease facts and figures. Alzheimers Dement, 2023. 19(4): p. 1598-1695.
[3]. Hou, Y., et al., Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol, 2019. 15(10):
p. 565-581.
[4]. Bates, G.P., et al., Huntington disease. Nat Rev Dis Primers, 2015. 1: p. 15005.
[5]. Brown, C.A., et al., Estimated Prevalence and Incidence of Amyotrophic Lateral Sclerosis and
SOD1 and C9orf72 Genetic Variants. Neuroepidemiology, 2021. 55(5): p. 342-353.
[6]. Federation, M.I., What is MS?. 2021.
[7]. Bloem, B.R., M.S. Okun and C. Klein, Parkinson's disease. Lancet, 2021. 397(10291): p. 2284-
2303.
[8]. Breijyeh, Z. and R. Karaman, Comprehensive Review on Alzheimer's Disease: Causes and
Treatment. Molecules, 2020. 25(24).
[9]. Tabrizi, S.J., et al., Potential disease-modifying therapies for Huntington's disease: lessons learned
and future opportunities. Lancet Neurol, 2022. 21(7): p. 645-658.
[10]. Hobson, E.V. and C.J. McDermott, Supportive and symptomatic management of amyotrophic
lateral sclerosis. Nat Rev Neurol, 2016. 12(9): p. 526-38.
[11]. Thompson, A.J., et al., Multiple sclerosis. Lancet, 2018. 391(10130): p. 1622-1636.
Journal Pre-proof
[12]. Yamout, B.I. and R. Alroughani, Multiple Sclerosis. Semin Neurol, 2018. 38(2): p. 212-225.
[13]. Bolognino, I., et al., Synthesis and Biological Evaluation of Dantrolene-Like Hydrazide and
Hydrazone Analogues as Multitarget Agents for Neurodegenerative Diseases. ChemMedChem, 2021.
16(18): p. 2807-2816.
[14]. Shi, Y., et al., Intranasal Dantrolene as a Disease-Modifying Drug in Alzheimer 5XFAD Mice.
J Alzheimers Dis, 2020. 76(4): p. 1375-1389.
[15]. Peng, J., et al., Dantrolene ameliorates cognitive decline and neuropathology in Alzheimer
triple transgenic mice. Neurosci Lett, 2012. 516(2): p. 274-9.
[16]. Wang, Y., et al., Dantrolene Ameliorates Impaired Neurogenesis and Synaptogenesis in
Induced Pluripotent Stem Cell Lines Derived from Patients with Alzheimer's Disease. Anesthesiology,
2020. 132(5): p. 1062-1079.
[17]. Huang, M. and S. Chen, DJ-1 in neurodegenerative diseases: Pathogenesis and clinical
application. Prog Neurobiol, 2021. 204: p. 102114.
[18]. Marogianni, C., et al., Neurodegeneration and Inflammation-An Interesting Interplay in
Parkinson's Disease. Int J Mol Sci, 2020. 21(22).
[19]. Erekat, N.S., Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin
Anat, 2022. 35(1): p. 65-78.
[20]. Lin, M.T. and M.F. Beal, Mitochondrial dysfunction and oxidative stress in neurodegenerative
diseases. Nature, 2006. 443(7113): p. 787-95.
[21]. Wu, Y., M. Chen and J. Jiang, Mitochondrial dysfunction in neurodegenerative diseases and
drug targets via apoptotic signaling. Mitochondrion, 2019. 49: p. 35-45.
[22]. Heneka, M.T., et al., Neuroinflammation in Alzheimer's disease. Lancet Neurol, 2015. 14(4):
p. 388-405.
[23]. Dhapola, R., et al., Recent advances in molecular pathways and therapeutic implications
targeting neuroinflammation for Alzheimer's disease. Inflammopharmacology, 2021. 29(6): p. 1669-
1681.
[24]. Turunc, B.E., et al., Nicotinamide treatment reduces the levels of oxidative stress, apoptosis,
and PARP-1 activity in Abeta(1-42)-induced rat model of Alzheimer's disease. Free Radic Res, 2014.
48(2): p. 146-58.
[25]. Zhang, Q.S., W. Liu and G.X. Lu, miR-200a-3p promotes b-Amyloid-induced neuronal
apoptosis through down-regulation of SIRT1 in Alzheimer's disease. J Biosci, 2017. 42(3): p. 397-404.
[26]. Huang, Q., X. Zhu and M. Xu, Silencing of TRIM10 alleviates apoptosis in cellular model of
Parkinson's disease. Biochem Biophys Res Commun, 2019. 518(3): p. 451-458.
[27]. Liu, X.L., et al., Mitochondria-mediated damage to dopaminergic neurons in Parkinson's
disease (Review). Int J Mol Med, 2018. 41(2): p. 615-623.
[28]. De Lella, E.A., et al., Chronic expression of low levels of tumor necrosis factor-alpha in the
substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and
microglia/macrophage activation. Neurobiol Dis, 2010. 37(3): p. 630-40.
[29]. Imamura, K., et al., Cytokine production of activated microglia and decrease in neurotrophic
factors of neurons in the hippocampus of Lewy body disease brains. Acta Neuropathol, 2005. 109(2): p.
141-50.
[30]. Saba, J., et al., Neuroinflammation in Huntington's Disease: A Starring Role for Astrocyte and
Microglia. Curr Neuropharmacol, 2022. 20(6): p. 1116-1143.
[31]. Faideau, M., et al., In vivo expression of polyglutamine-expanded huntingtin by mouse striatal
Journal Pre-proof
astrocytes impairs glutamate transport: a correlation with Huntington's disease subjects. Hum Mol Genet,
2010. 19(15): p. 3053-67.
[32]. Ji, E.S., et al., Treadmill exercise enhances spatial learning ability through suppressing
hippocampal apoptosis in Huntington's disease rats. J Exerc Rehabil, 2015. 11(3): p. 133-9.
[33]. Carata, E., et al., Looking to the Future of the Role of Macrophages and Extracellular Vesicles
in Neuroinflammation in ALS. Int J Mol Sci, 2023. 24(14).
[34]. Thonhoff, J.R., et al., Expanded autologous regulatory T-lymphocyte infusions in ALS: A
phase I, first-in-human study. Neurol Neuroimmunol Neuroinflamm, 2018. 5(4): p. e465.
[35]. Wiedau-Pazos, M., et al., Altered reactivity of superoxide dismutase in familial amyotrophic
lateral sclerosis. Science, 1996. 271(5248): p. 515-8.
[36]. Patel, B.P., et al., Caloric restriction shortens lifespan through an increase in lipid peroxidation,
inflammation and apoptosis in the G93A mouse, an animal model of ALS. PLoS One, 2010. 5(2): p.
e9386.
[37]. Sathasivam, S. and P.J. Shaw, Apoptosis in amyotrophic lateral sclerosis--what is the evidence?
Lancet Neurol, 2005. 4(8): p. 500-9.
[38]. Vaz, A.R., et al., Glycoursodeoxycholic acid reduces matrix metalloproteinase-9 and caspase-
9 activation in a cellular model of superoxide dismutase-1 neurodegeneration. Mol Neurobiol, 2015.
51(3): p. 864-77.
[39]. Filippi, M., et al., Multiple sclerosis. Nat Rev Dis Primers, 2018. 4(1): p. 43.
[40]. Lassmann, H., J. van Horssen and D. Mahad, Progressive multiple sclerosis: pathology and
pathogenesis. Nat Rev Neurol, 2012. 8(11): p. 647-56.
[41]. Liu, J. and F. Wang, Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular
Mechanisms and Therapeutic Implications. Front Immunol, 2017. 8: p. 1005.
[42]. Cankaya, S., et al., The therapeutic role of minocycline in Parkinson's disease. Drugs Context,
2019. 8: p. 212553.
[43]. Garrido-Mesa, N., A. Zarzuelo and J. Galvez, Minocycline: far beyond an antibiotic. Br J
Pharmacol, 2013. 169(2): p. 337-52.
[44]. Murphy, M.P., Targeting lipophilic cations to mitochondria. Biochim Biophys Acta, 2008.
1777(7-8): p. 1028-31.
[45]. Stefanova, N.A., et al., An antioxidant specifically targeting mitochondria delays progression
of Alzheimer's disease-like pathology. Aging (Albany NY), 2016. 8(11): p. 2713-2733.
[46]. Silachev, D.N., et al., Neuroprotective Effects of Mitochondria-Targeted Plastoquinone and
Thymoquinone in a Rat Model of Brain Ischemia/Reperfusion Injury. Molecules, 2015. 20(8): p. 14487-
503.
[47]. Ghosh, A., et al., Mitoapocynin Treatment Protects Against Neuroinflammation and
Dopaminergic Neurodegeneration in a Preclinical Animal Model of Parkinson's Disease. J
Neuroimmune Pharmacol, 2016. 11(2): p. 259-78.
[48]. Antolin, I., et al., Protective effect of melatonin in a chronic experimental model of Parkinson's
disease. Brain Res, 2002. 943(2): p. 163-73.
[49]. Erekat, N.S., Apoptosis and its Role in Parkinson's Disease. 2018.
[50]. Moujalled, D., A. Strasser and J.R. Liddell, Molecular mechanisms of cell death in
neurological diseases. Cell Death Differ, 2021. 28(7): p. 2029-2044.
[51]. Gregory, H. and K.A. Weant, Pathophysiology and Treatment of Malignant Hyperthermia.
Adv Emerg Nurs J, 2021. 43(2): p. 102-110.
Journal Pre-proof
[52]. Ellinas, H. and M.A. Albrecht, Malignant Hyperthermia Update. Anesthesiol Clin, 2020. 38(1):
p. 165-181.
[53]. Lanner, J.T., Ryanodine receptor physiology and its role in disease. Adv Exp Med Biol, 2012.
740: p. 217-34.
[54]. Zhao, F., et al., Dantrolene inhibition of ryanodine receptor Ca2+ release channels. Molecular
mechanism and isoform selectivity. J Biol Chem, 2001. 276(17): p. 13810-6.
[55]. Walweel, K., et al., Dantrolene inhibition of ryanodine channels (RyR2) in artificial lipid
bilayers depends on FKBP12.6. J Gen Physiol, 2023. 155(8).
[56]. Shi, Y., Y. Wang and H. Wei, Dantrolene : From Malignant Hyperthermia to Alzheimer's
Disease. CNS Neurol Disord Drug Targets, 2019. 18(9): p. 668-676.
[57]. Schmeckpeper, J., et al., RyR2 inhibition with dantrolene is antiarrhythmic, prevents further
pathological remodeling, and improves cardiac function in chronic ischemic heart disease. J Mol Cell
Cardiol, 2023. 181: p. 67-78.
[58]. Choi, R.H., X. Koenig and B.S. Launikonis, Dantrolene requires Mg(2+) to arrest malignant
hyperthermia. Proc Natl Acad Sci U S A, 2017. 114(18): p. 4811-4815.
[59]. Fujii, S., et al., RyR2-targeting therapy prevents left ventricular remodeling and ventricular
tachycardia in post-infarction heart failure. J Mol Cell Cardiol, 2023. 178: p. 36-50.
[60]. Ryanodex--A New Dantrolene Formulation for Malignant Hyperthermia. Med Lett Drugs
Ther, 2015. 57(1472): p. 100.
[61]. Ward, A., M.O. Chaffman and E.M. Sorkin, Dantrolene. A review of its pharmacodynamic
and pharmacokinetic properties and therapeutic use in malignant hyperthermia, the neuroleptic
malignant syndrome and an update of its use in muscle spasticity. Drugs, 1986. 32(2): p. 130-68.
[62]. Lerman, J., M.E. McLeod and H.A. Strong, Pharmacokinetics of intravenous dantrolene in
children. Anesthesiology, 1989. 70(4): p. 625-9.
[63]. Dykes, M.H., Evaluation of a muscle relaxant: dantrolene sodium (Dantrium). JAMA, 1975.
231(8): p. 862-4.
[64]. Lietman, P.S., R.H. Haslam and J.R. Walcher, Pharmacology of dantrolene sodium in children.
Arch Phys Med Rehabil, 1974. 55(8): p. 388-92.
[65]. Krause, T., et al., Dantrolene--a review of its pharmacology, therapeutic use and new
developments. Anaesthesia, 2004. 59(4): p. 364-73.
[66]. Kheder, A. and K.P. Nair, Spasticity: pathophysiology, evaluation and management. Pract
Neurol, 2012. 12(5): p. 289-98.
[67]. Tormoehlen, L.M. and D.E. Rusyniak, Neuroleptic malignant syndrome and serotonin
syndrome. Handb Clin Neurol, 2018. 157: p. 663-675.
[68]. Rosenberg, M.R. and M. Green, Neuroleptic malignant syndrome. Review of response to
therapy. Arch Intern Med, 1989. 149(9): p. 1927-31.
[69]. Cameron-Burr, K.T., R.A. Bola and E.A. Kiyatkin, Dantrolene sodium fails to reverse robust
brain hyperthermia induced by MDMA and methamphetamine in rats. Psychopharmacology (Berl),
2023. 240(4): p. 785-795.
[70]. Laitano, O., K.O. Murray and L.R. Leon, Overlapping Mechanisms of Exertional Heat Stroke
and Malignant Hyperthermia: Evidence vs. Conjecture. Sports Med, 2020. 50(9): p. 1581-1592.
[71]. Jiang, B., et al., Could dantrolene be explored as a repurposed drug to treat COVID-19 patients
by restoring intracellular calcium homeostasis? Eur Rev Med Pharmacol Sci, 2020. 24(19): p. 10228-
10238.
Journal Pre-proof
[72]. Wei, H., G. Liang and R.M. Vera, Dantrolene repurposed to treat sepsis or septic shock and
COVID-19 patients. Eur Rev Med Pharmacol Sci, 2021. 25(7): p. 3136-3144.
[73]. Hopp, S.C., et al., Calcium dysregulation via L-type voltage-dependent calcium channels and
ryanodine receptors underlies memory deficits and synaptic dysfunction during chronic
neuroinflammation. J Neuroinflammation, 2015. 12: p. 56.
[74]. Hopp, S.C., et al., Differential neuroprotective and anti-inflammatory effects of L-type voltage
dependent calcium channel and ryanodine receptor antagonists in the substantia nigra and locus
coeruleus. J Neuroimmune Pharmacol, 2015. 10(1): p. 35-44.
[75]. Osipchuk, N.C., A.M. Soulika and A.F. Fomina, Modulation of Ryanodine Receptors Activity
Alters the Course of Experimental Autoimmune Encephalomyelitis in Mice. Front Physiol, 2021. 12: p.
770820.
[76]. Wu, Z., et al., Long-term dantrolene treatment reduced intraneuronal amyloid in aged
Alzheimer triple transgenic mice. Alzheimer Dis Assoc Disord, 2015. 29(3): p. 184-191.
[77]. Mustaly-Kalimi, S., et al., Protein mishandling and impaired lysosomal proteolysis generated
through calcium dysregulation in Alzheimer's disease. Proc Natl Acad Sci U S A, 2022. 119(49): p.
e2211999119.
[78]. Zhang, H., et al., Calcium signaling, excitability, and synaptic plasticity defects in a mouse
model of Alzheimer's disease. J Alzheimers Dis, 2015. 45(2): p. 561-80.
[79]. Chen, X., et al., Dantrolene is neuroprotective in Huntington's disease transgenic mouse model.
Mol Neurodegener, 2011. 6: p. 81.
[80]. Oikonomou, K.D., et al., Calcium dysregulation and compensation in cortical pyramidal
neurons of the R6/2 mouse model of Huntington's disease. J Neurophysiol, 2021. 126(4): p. 1159-1171.
[81]. Carvalho, D.S., et al., What are the Evolutionary Origins of Mitochondria? A Complex
Network Approach. PLoS One, 2015. 10(9): p. e0134988.
[82]. DELUCA, H.F. and G.W. ENGSTROM, Calcium uptake by rat kidney mitochondria. Proc
Natl Acad Sci U S A, 1961. 47(11): p. 1744-50.
[83]. Pivovarova, N.B., et al., Depolarization-induced mitochondrial Ca accumulation in
sympathetic neurons: spatial and temporal characteristics. J Neurosci, 1999. 19(15): p. 6372-84.
[84]. Montero, M., et al., Chromaffin-cell stimulation triggers fast millimolar mitochondrial Ca2+
transients that modulate secretion. Nat Cell Biol, 2000. 2(2): p. 57-61.
[85]. Starkov, A.A., The molecular identity of the mitochondrial Ca2+ sequestration system. FEBS
J, 2010. 277(18): p. 3652-63.
[86]. Duchen, M.R., Mitochondria in health and disease: perspectives on a new mitochondrial
biology. Mol Aspects Med, 2004. 25(4): p. 365-451.
[87]. Wang, C., et al., Role of intracellular calcium stores in cell death from oxygen-glucose
deprivation in a neuronal cell line. J Cereb Blood Flow Metab, 2002. 22(2): p. 206-14.
[88]. Zhai, Q., et al., Nanorepairers Rescue Inflammation-Induced Mitochondrial Dysfunction in
Mesenchymal Stem Cells. Adv Sci (Weinh), 2022. 9(4): p. e2103839.
[89]. Qiu, L.L., et al., Dysregulation of BDNF/TrkB signaling mediated by
NMDAR/Ca(2+)/calpain might contribute to postoperative cognitive dysfunction in aging mice. J
Neuroinflammation, 2020. 17(1): p. 23.
[90]. Choi, D.W., Ionic dependence of glutamate neurotoxicity. J Neurosci, 1987. 7(2): p. 369-79.
[91]. Muehlschlegel, S. and J.R. Sims, Dantrolene: mechanisms of neuroprotection and possible
clinical applications in the neurointensive care unit. Neurocrit Care, 2009. 10(1): p. 103-15.
Journal Pre-proof
[92]. Walters, G.C. and Y.M. Usachev, Mitochondrial calcium cycling in neuronal function and
neurodegeneration. Front Cell Dev Biol, 2023. 11: p. 1094356.
[93]. Orrenius, S., B. Zhivotovsky and P. Nicotera, Regulation of cell death: the calcium-apoptosis
link. Nat Rev Mol Cell Biol, 2003. 4(7): p. 552-65.
[94]. Triantafilou, K., et al., The complement membrane attack complex triggers intracellular Ca2+
fluxes leading to NLRP3 inflammasome activation. J Cell Sci, 2013. 126(Pt 13): p. 2903-13.
[95]. Frandsen, A. and A. Schousboe, Dantrolene prevents glutamate cytotoxicity and Ca2+ release
from intracellular stores in cultured cerebral cortical neurons. J Neurochem, 1991. 56(3): p. 1075-8.
[96]. Nakayama, R., et al., Effects of dantrolene on extracellular glutamate concentration and
neuronal death in the rat hippocampal CA1 region subjected to transient ischemia. Anesthesiology, 2002.
96(3): p. 705-10.
[97]. Duzenli, S., K. Bakuridze and A. Gepdiremen, The effects of ruthenium red, dantrolene and
nimodipine, alone or in combination, in NMDA induced neurotoxicity of cerebellar granular cell culture
of rats. Toxicol In Vitro, 2005. 19(5): p. 589-94.
[98]. Makarewicz, D., E. Zieminska and J.W. Lazarewicz, Dantrolene inhibits NMDA-induced
45Ca uptake in cultured cerebellar granule neurons. Neurochem Int, 2003. 43(4-5): p. 273-8.
[99]. Hayashi, T., et al., Effect of dantrolene on KCl- or NMDA-induced intracellular Ca2+ changes
and spontaneous Ca2+ oscillation in cultured rat frontal cortical neurons. J Neural Transm (Vienna),
1997. 104(8-9): p. 811-24.
[100]. Gursoy, I.D., et al., Investigation of the Possible Protective Effects of Ketamine and
Dantrolene on the Hippocampal Apoptosis and Spatial Learning in Rats Exposed to Repeated
Electroconvulsive Seizures as a Model of Status Epilepticus. Turk Neurosurg, 2020. 30(6): p. 871-884.
[101]. Martins, B.C., et al., Association of riluzole and dantrolene improves significant recovery after
acute spinal cord injury in rats. Spine J, 2018. 18(3): p. 532-539.
[102]. Ansari, N., et al., Interaction of 2-APB, dantrolene, and TDMT with IP3R and RyR modulates
ER stress-induced programmed cell death I and II in neuron-like PC12 cells: an experimental and
computational investigation. J Biomol Struct Dyn, 2014. 32(8): p. 1211-30.
[103]. Leng, F. and P. Edison, Neuroinflammation and microglial activation in Alzheimer disease:
where do we go from here? Nat Rev Neurol, 2021. 17(3): p. 157-172.
[104]. Hoffmann, A., et al., Elevation of basal intracellular calcium as a central element in the
activation of brain macrophages (microglia): suppression of receptor-evoked calcium signaling and
control of release function. J Neurosci, 2003. 23(11): p. 4410-9.
[105]. Wu, T., et al., Nitrogen-doped graphene quantum dots induce ferroptosis through disrupting
calcium homeostasis in microglia. Part Fibre Toxicol, 2022. 19(1): p. 22.
[106]. Klegeris, A., et al., Functional ryanodine receptors are expressed by human microglia and
THP-1 cells: Their possible involvement in modulation of neurotoxicity. J Neurosci Res, 2007. 85(10):
p. 2207-15.
[107]. Liu, N., et al., TRPV4 contributes to ER stress and inflammation: implications for Parkinson's
disease. J Neuroinflammation, 2022. 19(1): p. 26.
[108]. Kurum, T., E. Tatli and M. Yuksel, Effects of carvedilol on plasma levels of pro-inflammatory
cytokines in patients with ischemic and nonischemic dilated cardiomyopathy. Tex Heart Inst J, 2007.
34(1): p. 52-9.
[109]. Fairless, R., S.K. Williams and R. Diem, Dysfunction of neuronal calcium signalling in
neuroinflammation and neurodegeneration. Cell Tissue Res, 2014. 357(2): p. 455-62.
Journal Pre-proof
[110]. Ducreux, S., et al., Effect of ryanodine receptor mutations on interleukin-6 release and
intracellular calcium homeostasis in human myotubes from malignant hyperthermia-susceptible
individuals and patients affected by central core disease. J Biol Chem, 2004. 279(42): p. 43838-46.
[111]. Hisatsune, J., et al., Molecular characterization of Helicobacter pylori VacA induction of IL-
8 in U937 cells reveals a prominent role for p38MAPK in activating transcription factor-2, cAMP
response element binding protein, and NF-kappaB activation. J Immunol, 2008. 180(7): p. 5017-27.
[112]. Hotchkiss, R.S., et al., Calcium antagonists decrease plasma and tissue concentrations of
tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-1 alpha in a mouse model of endotoxin.
Shock, 1995. 3(5): p. 337-42.
[113]. Fischer, D.R., et al., Dantrolene reduces serum TNFalpha and corticosterone levels and muscle
calcium, calpain gene expression, and protein breakdown in septic rats. Shock, 2001. 15(3): p. 200-7.
[114]. Teleanu, D.M., et al., An Overview of Oxidative Stress, Neuroinflammation, and
Neurodegenerative Diseases. Int J Mol Sci, 2022. 23(11).
[115]. Todorova, V.K., et al., Dantrolene Attenuates Cardiotoxicity of Doxorubicin Without
Reducing its Antitumor Efficacy in a Breast Cancer Model. Transl Oncol, 2020. 13(2): p. 471-480.
[116]. Keles, I., et al., Protective effects of dantrolene and methylprednisolone against spinal cord
injury-induced early oxidative damage in rabbit bladder: A comparative experimental study. Adv Clin
Exp Med, 2019. 28(12): p. 1697-1704.
[117]. Lu, Y.C., et al., ER-Dependent Ca++-mediated Cytosolic ROS as an Effector for Induction of
Mitochondrial Apoptotic and ATM-JNK Signal Pathways in Gallic Acid-treated Human Oral Cancer
Cells. Anticancer Res, 2016. 36(2): p. 697-705.
[118]. Jing, G., et al., Erbin protects against sepsis-associated encephalopathy by attenuating
microglia pyroptosis via IRE1alpha/Xbp1s-Ca(2+) axis. J Neuroinflammation, 2022. 19(1): p. 237.
[119]. Wang, S., et al., The mechanisms of NLRP3 inflammasome/pyroptosis activation and their
role in Parkinson's disease. Int Immunopharmacol, 2019. 67: p. 458-464.
Journal Pre-proof
... This can be further aggravated by excessive Ca 2+ release from the primary intracellular Ca 2+ store endoplasmic reticulum (ER) via over activation of inositol trisphosphate receptors (InsP3R) [7,8]. This upstream Ca 2+ dysregulation results in downstream multiple AD pathologies [9], including amyloid [10,11] and tau [12,13] pathologies, mitochondria dysfunction [14,15], pathological reactive oxygen species (ROS) production and lipid peroxidation [16,17], impairment of autophagy [18,19], neuroinflammation [20][21][22], impaired neurogenesis [18,23], disruption of synaptic structure and function [18,24], and eventually cognitive dysfunction [25][26][27]. Importantly, an increasing number of studies suggest that downstream inflammation and associated cell/neuron death pyroptosis play critical role in AD pathology as etiology for both dementia [28,29] and depression behavior [28,30], and could be effective therapeutic target [31]. ...
... Lithium, a first-line drug for depression and mania in bipolar disorder, is also neuroprotective against many neurodegenerative diseases [53,54], including AD [55][56][57]. Although multiple mechanisms have been proposed for lithium treatment on depression behaviors and cognitive dysfunction in AD, the ability of lithium to ameliorate upstream Ca 2+ dysregulation by inhibiting NMDAR [58] and suppressing InsP3R [59], as well as addressing multiple downstream pathologies [9]-including oxidative stress, inflammation and pyroptosis, and synapse and neuronal destruction-is considered the primary mechanism by which lithium may alleviate both cognitive dysfunction and depression behaviors in AD and other neurodegenerative diseases [60][61][62][63]. However, major concerns or limitations for lithium in bipolar disorder patients is its narrow therapeutic window, proneness to side effects/toxicity, and intolerance in patients with chronic use [64][65][66]. ...
... This increase leads to downstream mitochondrial dysfunction, oxidative stress, and subsequent activation of the NRLP3 inflammasome complex and caspase-1. This cascade also leads to an elevation of cytokines IL-1β and IL-18, cleavage of NSDMD into an ion channel N-terminal to release cytotoxic cytokines, and ultimately, programmed cell death by pyroptosis [9]. Although lithium has been demonstrated to be neuroprotective by inhibition of inflammation, its effects and mechanisms on inflammation associated pyroptosis has not been well investigated. ...
Preprint
Full-text available
Background Alzheimer’s disease (AD) is a devastating neurodegenerative disease (AD) and has no treatment that can cure or halt the disease progression. This study explored the therapeutic potential of lithium salt dissolved in Ryanodex formulation vehicle (RFV) and delivered to the brain by intranasal application. We first compared lithium concentrations in the brain and blood of wild-type mice following intranasal or oral administration of lithium chloride (LiCl) dissolved in either RFV or water. The beneficial and side effects of intranasal versus oral LiCl in RFV in these mice were assessed and potential mechanisms underlying the efficacy of anti-inflammation and anti-pyroptosis in the brains were also investigated in both wild-type (WT) and 5XFAD Alzheimer’s Disease (AD) mice brains. Methods For the study of brain versus blood lithium concentrations, WT B6SJLF1/J mice at 2 months of age were treated with intranasal or oral LiCl (3 mmol/kg) dissolved in RFV or in water. Brain and blood lithium concentrations were measured at various times after drugs administration. Brain/blood lithium concentration ratios were then determined. For studying therapeutic efficacy versus side effects and their underlying mechanisms, 5XFAD and WT B6SJLF1/J mice were treated with intranasal LiCl (3 mmol/kg) daily, Monday to Friday each week, in RFV beginning at 2 or 9 months of age with a 12-week treatment duration. Animal behaviors were assessed for depression (tail suspension), cognition (fear conditioning and Y maze), olfaction (buried food test), and motor functions (rotarod) at the age of 5 and 12 months. Blood and brain tissue were harvested from these mice at 13 months. Blood biomarkers for the functions of thyroid (thyroid stimulating hormone, TSH) and kidney (creatinine) were measured using ELISA. Changes in protein expression levels of the endoplasmic reticulum Ca ²⁺ release channels type 1 InsP 3 receptors (InsP 3 R-1), malondialdehyde (MDA)-modified proteins and 4-hydroxy-2-nonenal (4-HNE), pyroptosis regulatory proteins (NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, N-terminal of Gasdermin D (GSDMD)), cytotoxic (IL-1β, IL-18, IL-6, TNF-α) and cytoprotective (IL-10) cytokines and synapse proteins (PSD-95, synapsin-1) were determined using immunoblotting. Mouse body weights were monitored regularly. Results Compared to oral LiCl in RFV nanoparticles, intranasal treatment of WT mice with LiCl in RFV markedly decreased blood concentrations at the time frame of 30-120 minutes. The ratio of brain/blood lithium concentration after Intranasal lithium chloride in RFV significantly increased, in comparison to those after oral administration lithium chloride in RFV or intranasal administration of lithium chloride in water. Intranasal lithium chloride in RFV inhibited both memory loss and depressive behavior in adult and aged 5XFAD mice. Additionally intranasal treatment of aged 5XFAD mice with LiCl in RFV effectively suppressed the increases in InsP 3 R-1, intracellular oxidative stress markers (4-HNE-bound and MDA-modified proteins), pyroptosis activation proteins (NLRP3, cleaved caspase-1, N-terminal GSDMD) and cytotoxic cytokines (IL-1β, IL-6, TNF-α), but reversed the down-regulation of cytoprotective cytokine IL-10. Intranasal LiCl in RFV also alleviated the loss of the postsynaptic synapse protein PSD-95, but not synapsin-1, in aged 5XFAD mice. Blood level of the kidney function marker creatinine was significantly increased in 5XFAD than in WT mice in an age-dependent manner and this elevation was abolished by intranasal delivery of LiCl in RFV. Intranasal LiCl in RFV for 12 weeks in both WT or 5XFAD mice did not affect blood biomarkers for thyroid function, nor did it affect smell or muscle function or body weight. Conclusion Intranasal administration of LiCl in RFV significantly decreased lithium blood concentrations and increased brain/blood lithium concentration ratio, in comparison to its oral administration. Intranasal administration of LiCl in RFV robustly protected against both memory loss and depressive-like behavior, while had no side effects concerning thyroid and kidney toxicity in 5XFAD mice. These lithium-induced beneficial effects were strongly associated with lithium’s suppression of InsP 3 R-1 Ca ²⁺ channel receptor increase, pathological neuroinflammation and activation of the pyroptosis pathway, as well as the loss of some synaptic proteins. Intranasal delivery of lithium salt in RFV could become an effective and potent inhibitor of pathological inflammation/pyroptosis in the CNS and serve as a new treatment for both AD-associated dementia and depression with minimal unwanted side effects including peripheral organ toxicity.
... Although mechanisms unclear, increasing evidence suggest that disruption of intracellular Ca 2+ homeostasis and associated pathologies including inflammation and synapse dysfunction play critical role of pathologies in Alzheimer's disease (AD) 1 . The abnormally increased glutamate and associated excitotoxicity via overactivation of N-methyl-D-aspartate (NMDA) receptors (NMDAR) result in the overactivation of ryanodine (RyRs) and InsP3 (InsP3R) receptors located on membrane of endoplasmic reticulum (ER) and associated excessive Ca 2+ release from ER, leading to depletion of ER Ca 2+ and pathological elevation of cytosol and mitochondrial Ca 2+ concentrations, detrimental to cell survival 2,3 . ...
Preprint
Full-text available
Background: This study investigates the effects of intranasal dantrolene nanoparticles on inflammation and programmed cell death by pyroptosis in 5XFAD Alzheimers Disease (AD) mice. Methods: 5XFAD and wild type (WT) B6SJLF1/J mice were treated with intranasal dantrolene nanoparticles (5 mg/kg), daily, Monday to Friday, for 12 weeks continuously, starting at 9 months of age. Blood and brain were harvested at 13 months of age, one month after completion of 12 weeks intranasal dantrolene nanoparticle treatment. Blood biomarkers function of liver (Alanine transaminase, ALT), kidney (Creatinine), and thyroid (TSH: Thyroid-stimulating hormone) were measured using ELISA. The changes of whole brain tissue proteins on Ca2+ release channels on membrane of endoplasmic reticulum (type 2 ryanodine and type 1 InsP3 receptors, RyR-2 and InsP3R-1), lipid peroxidation byproduct malondialdehyde (MDA)-modified proteins, 4-HNE, pyroptosis regulatory proteins (NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, full length or N-terminal of Gasdermin D (GSDMD), cytotoxic (IL-1, IL-18, IL-6, TNF-α) and cytoprotective (IL-10) cytokines, astrogliosis (GFAP), microgliosis (IBA-1) and synapse proteins (PSD-95, Synapsin-1) were determined using immunoblotting. Body weights were monitored regularly. Results: Intranasal dantrolene nanoparticles significantly inhibited the increase of RyR-2 and InsP3R-1 proteins, MDA-modified proteins, 4-NHE, pyroptosis regulatory proteins (NLRP3, cleaved caspase-1, N-terminal GSDMD), cytotoxic cytokine (IL-1, IL-18, IL-6, TNF-), biomarkers for astrogliosis (GFAP) and microgliosis (IBA-1), and the decrease of cytoprotective cytokine (IL-10) and synaptic proteins (PSD-95, synpasin-1). Intranasal dantrolene nanoparticles for 12 weeks did not affect blood biomarkers for function of liver, kidney, and thyroid, not did it change body weight significantly. Conclusion: Intranasal dantrolene nanoparticles significantly inhibit the increase of RyR-2 and InsP3R-1 Ca2+ channel receptor proteins, ameliorate activation of the pyroptosis pathway and pathological inflammation, and the associated loss of synapse proteins. Intranasal dantrolene nanoparticles for three months did not affect liver, kidney and thyroid functions or cause other side effects.
Article
Full-text available
Background Alzheimer’s disease (AD) is a devastating neurodegenerative disease (AD) and has no treatment that can cure or halt the disease progression. This study explored the therapeutic potential of lithium salt dissolved in Ryanodex formulation vehicle (RFV) and delivered to the brain by intranasal application. We first compared lithium concentrations in the brain and blood of wild-type mice following intranasal or oral administration of lithium chloride (LiCl) dissolved in either RFV or water. The beneficial and side effects of intranasal versus oral LiCl in RFV in these mice were assessed and potential mechanisms underlying the efficacy of anti-inflammation and anti-pyroptosis in the brains were also investigated in both wild-type and 5XFAD Alzheimer’s Disease (AD) mice brains. Methods For the study of brain versus blood lithium concentrations, wild-type (WT) B6SJLF1/J mice at 2 months of age were treated with intranasal or oral LiCl (3 mmol/kg) dissolved in RFV or in water. Brain and blood lithium concentrations were measured at various times after drugs administration. Brain/blood lithium concentration ratios were then determined. For studying therapeutic efficacy versus side effects and their underlying mechanisms, 5XFAD and WT B6SJLF1/J mice were treated with intranasal LiCl (3 mmol/kg) daily, Monday to Friday each week, in RFV beginning at 2 or 9 months of age with a 12-week treatment duration. Animal behaviors were assessed for depression (tail suspension), cognition (fear conditioning and Y maze), olfaction (buried food test), and motor functions (rotarod) at the age of 5 and 12 months. Blood and brain tissue were harvested from these mice at 13 months. Blood biomarkers for the functions of thyroid (thyroid stimulating hormone, TSH) and kidney (creatinine) were measured using ELISA. Changes in protein expression levels of the endoplasmic reticulum Ca²⁺ release channels type 1 InsP3 receptors (InsP3R-1), malondialdehyde (MDA)-modified proteins and 4-hydroxy-2-nonenal (4-HNE), pyroptosis regulatory proteins (NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, N-terminal of Gasdermin D (GSDMD)), cytotoxic (IL-1β, IL-18, IL-6, TNF-α) and cytoprotective (IL-10) cytokines and synapse proteins (PSD-95, synapsin-1) were determined using immunoblotting. Mouse body weights were monitored regularly. Results Compared to oral LiCl in RFV nanoparticles, intranasal treatment of WT mice with LiCl in RFV markedly decreased blood concentrations at the time range of 30–120 min. The ratio of brain/blood lithium concentration after intranasal lithium chloride in RFV significantly increased, in comparison to those after oral administration lithium chloride in RFV or intranasal administration of lithium chloride in water. Intranasal lithium chloride in RFV inhibited both memory loss and depressive behavior in adult and aged 5XFAD mice. Additionally intranasal treatment of aged 5XFAD mice with LiCl in RFV effectively suppressed the increases in InsP3R-1, intracellular oxidative stress markers (4-HNE-bound and MDA-modified proteins), pyroptosis activation proteins (NLRP3, cleaved caspase-1, N-terminal GSDMD) and cytotoxic cytokines (IL-1β, IL-6, TNF-α), but reversed the down-regulation of cytoprotective cytokine IL-10. Intranasal LiCl in RFV also alleviated the loss of the postsynaptic synapse proteins PSD-95, but not synapsin-1, in aged 5XFAD mice. Blood level of the kidney function marker creatinine was significantly increased in 5XFAD than in WT mice in an age-dependent manner and this elevation was abolished by intranasal delivery of LiCl in RFV. Intranasal LiCl in RFV for 12 weeks in both WT or 5XFAD mice did not affect blood biomarkers for thyroid function, nor did it affect smell or muscle function or body weight. Conclusion Intranasal administration of LiCl in RFV significantly decreased lithium blood concentrations and increased brain/blood lithium concentration ratio, in comparison to its oral administration. Intranasal administration of LiCl in RFV robustly protected against both memory loss and depressive-like behavior, while had no side effects concerning thyroid and kidney toxicity in 5XFAD mice. These lithium-induced beneficial effects were strongly associated with lithium’s suppression of InsP3R-1 Ca²⁺ channel receptor increase, pathological neuroinflammation and activation of the pyroptosis pathway, as well as the loss of the synaptic protein PSD-95. Intranasal delivery of lithium salt in RFV could become an effective and potent inhibitor of pathological inflammation/pyroptosis in the CNS and serve as a new treatment for both AD-associated dementia and depression with minimal unwanted side effects including peripheral organ toxicity. Graphical Abstract Intranasal delivery of lithium salt in Ryanodex Formulation Vehicle (RFV) robustly protected against both cognitive dysfunction and depression-like behavior 5XFAD mice, with no side effects or organ toxicity on thyroid and kidney functions. These lithium-induced beneficial effects were strongly associated with lithium’s suppression of pathological elevation type I InsP3 receptors (InsP3R-1) Ca2+ channel receptor proteins, oxidative stress, pathological neuroinflammation and activation of the pyroptosis pathway (Increased proteins of NLRP3, cleaved caspase-1, N-terminal GSDMD, IL-1β and IL-18). Intranasal delivery of lithium salt in RFV could become an effective drug in treating AD.
Article
Full-text available
Neuroinflammation is a common pathological feature of amyotrophic lateral sclerosis (ALS). Although scientific evidence to date does not allow defining neuroinflammation as an ALS trigger, its role in exacerbating motor neuron (MNs) degeneration and disease progression is attracting research interest. Activated CNS (Central Nervous System) glial cells, proinflammatory peripheral and infiltrated T lymphocytes and monocytes/macrophages, as well as the immunoreactive molecules they release, represent the active players for the role of immune dysregulation enhancing neuroinflammation. The crosstalk between the peripheral and CNS immune cells significantly correlates with the survival of ALS patients since the modification of peripheral macrophages can downregulate inflammation at the periphery along the nerves and in the CNS. As putative vehicles for misfolded protein and inflammatory mediators between cells, extracellular vesicles (EVs) have also drawn particular attention in the field of ALS. Both CNS and peripheral immune cells release EVs, which are able to modulate the behavior of neighboring recipient cells; unfortunately, the mechanisms involved in EVs-mediated communication in neuroinflammation remain unclear. This review aims to synthesize the current literature regarding EV-mediated cell-to-cell communication in the brain under ALS, with a particular point of view on the role of peripheral macrophages in responding to inflammation to understand the biological process and exploit it for ALS management.
Article
Full-text available
Dantrolene is a neutral hydantoin that is clinically used as a skeletal muscle relaxant to prevent overactivation of the skeletal muscle calcium release channel (RyR1) in response to volatile anesthetics. Dantrolene has aroused considerable recent interest as a lead compound for stabilizing calcium release due to overactive cardiac calcium release channels (RyR2) in heart failure. Previously, we found that dantrolene produces up to a 45% inhibition RyR2 with an IC50 of 160 nM, and that this inhibition requires the physiological association between RyR2 and CaM. In this study, we tested the hypothesis that dantrolene inhibition of RyR2 in the presence of CaM is modulated by RyR2 phosphorylation at S2808 and S2814. Phosphorylation was altered by incubations with either exogenous phosphatase (PP1) or kinases; PKA to phosphorylate S2808 or endogenous CaMKII to phosphorylate S2814. We found that PKA caused selective dissociation of FKBP12.6 from the RyR2 complex and a loss of dantrolene inhibition. Rapamycin-induced FKBP12.6 dissociation from RyR2 also resulted in the loss of dantrolene inhibition. Subsequent incubations of RyR2 with exogenous FKBP12.6 reinstated dantrolene inhibition. These findings indicate that the inhibitory action of dantrolene on RyR2 depends on RyR2 association with FKBP12.6 in addition to CaM as previously found.
Article
Full-text available
RationaleHyperthermia induced by psychomotor stimulants may cause leakage of the blood-brain barrier, vasogenic edema, and lethality in extreme cases. Current treatments such as whole-body cooling are only symptomatic and a clear need to develop pharmacological interventions exists. Dantrolene sodium, a peripheral muscle relaxant used in the treatment of malignant hyperthermia, has been proposed as potentially effective to treat MDMA-hyperthermia in emergency rooms. However, debate around its efficacy for this indication persists.Objectives To investigate dantrolene as a treatment for illicit hyperthermia induced by psychomotor stimulant drugs, we examined how Ryanodex®, a concentrated formulation of dantrolene sodium produced by Eagle Pharmaceuticals, influences 3,4-methylenedioxymethamphetamine (MDMA)- and methamphetamine (METH)-induced hyperthermia in awake freely moving rats. We injected rats with moderate doses of MDMA (9 mg/kg) and METH (9 mg/kg) and administered Ryanodex® intravenously (6 mg/kg) after the development of robust hyperthermia (>2.5 °C) mimicking clinical acute intoxication. We conducted simultaneous temperature recordings in the brain, temporal muscle, and skin to determine the basic mechanisms underlying temperature responses. To assess the efficacy of dantrolene in attenuating severe hyperthermia, we administered MDMA to rats maintained in a warm ambient environment (29 °C), conditions which produce robust brain and body hyperthermia (>40 °C) and lethality.ResultsDantrolene failed to attenuate MDMA- and METH-induced hyperthermia, though locomotor activity was significantly reduced. All animals maintained at warm ambient temperatures that received dantrolene during severe drug-induced hyperthermia died within or soon after the recording session.Conclusions Our results suggest that dantrolene sodium formulations are not mechanistically suited to treat MDMA- and METH-induced hyperthermia.
Article
Full-text available
Mitochondria are essential for proper cellular function through their critical roles in ATP synthesis, reactive oxygen species production, calcium (Ca²⁺) buffering, and apoptotic signaling. In neurons, Ca²⁺ buffering is particularly important as it helps to shape Ca²⁺ signals and to regulate numerous Ca²⁺-dependent functions including neuronal excitability, synaptic transmission, gene expression, and neuronal toxicity. Over the past decade, identification of the mitochondrial Ca²⁺ uniporter (MCU) and other molecular components of mitochondrial Ca²⁺ transport has provided insight into the roles that mitochondrial Ca²⁺ regulation plays in neuronal function in health and disease. In this review, we discuss the many roles of mitochondrial Ca²⁺ uptake and release mechanisms in normal neuronal function and highlight new insights into the Ca²⁺-dependent mechanisms that drive mitochondrial dysfunction in neurologic diseases including epilepsy, Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. We also consider how targeting Ca²⁺ uptake and release mechanisms could facilitate the development of novel therapeutic strategies for neurological diseases.
Article
Full-text available
Impairments in neural lysosomal- and autophagic-mediated degradation of cellular debris contribute to neuritic dystrophy and synaptic loss. While these are well-characterized features of neurodegenerative disorders such as Alzheimer’s disease (AD), the upstream cellular processes driving deficits in pathogenic protein mishandling are less understood. Using a series of fluorescent biosensors and optical imaging in model cells, AD mouse models and human neurons derived from AD patients, we reveal a previously undescribed cellular signaling cascade underlying protein mishandling mediated by intracellular calcium dysregulation, an early component of AD pathogenesis. Increased Ca ²⁺ release via the endoplasmic reticulum (ER)-resident ryanodine receptor (RyR) is associated with reduced expression of the lysosome proton pump vacuolar-ATPase (vATPase) subunits (V1B2 and V0a1), resulting in lysosome deacidification and disrupted proteolytic activity in AD mouse models and human-induced neurons (HiN). As a result of impaired lysosome digestive capacity, mature autophagosomes with hyperphosphorylated tau accumulated in AD murine neurons and AD HiN, exacerbating proteinopathy. Normalizing AD-associated aberrant RyR-Ca ²⁺ signaling with the negative allosteric modulator, dantrolene (Ryanodex), restored vATPase levels, lysosomal acidification and proteolytic activity, and autophagic clearance of intracellular protein aggregates in AD neurons. These results highlight that prior to overt AD histopathology or cognitive deficits, aberrant upstream Ca ²⁺ signaling disrupts lysosomal acidification and contributes to pathological accumulation of intracellular protein aggregates. Importantly, this is demonstrated in animal models of AD, and in human iPSC-derived neurons from AD patients. Furthermore, pharmacological suppression of RyR-Ca ²⁺ release rescued proteolytic function, revealing a target for therapeutic intervention that has demonstrated effects in clinically-relevant assays.
Article
Full-text available
Background Microglia pyroptosis-mediated neuroinflammation is thought to be the crucial pathogenesis of sepsis-associated encephalopathy (SAE). Erbin has been reported to be associated with various inflammatory diseases. However, the role of Erbin in SAE and the relationship between Erbin and microglia pyroptosis are unknown. In this study, we investigated the promising role and underlying molecular mechanism of Erbin in the regulation of microglia pyroptosis. Methods WT and Erbin knockout mice underwent cecum ligation perforation (CLP) to induce SAE. Primary mouse microglia and BV2 cells were treated with LPS/nigericin in vitro. Behavioral tests were performed to evaluate cognitive function. Nissl staining and transmission electron microscopy were used to assess histological and structural lesions. ELISA and qPCR were carried out to detect neuroinflammation. Western blot and immunofluorescence were used to analyze protein expression. Flow cytometry and confocal microscopy were utilized to observe the Ca²⁺ changes in the cytoplasm and endoplasmic reticulum (ER). To further explore the underlying mechanism, STF083010 was administered to block the IRE1α/Xbp1s pathway. Results Erbin deletion resulted in more pronounced neuronal damage and cognitive impairment in mice that underwent CLP. Erbin knockout promoted microglial pyroptosis and inflammatory cytokines secretion in vivo and in vitro, which was mediated by activation of the IRE1α/Xbp1s. Treatment with the selective inhibitor STF083010 significantly inhibited IRE1α/Xbp1s pathway activity, decreased intracytoplasmic Ca²⁺, attenuated microglial pyroptosis, reduced pro-inflammatory cytokine secretion, lessened neuronal damage, and improved cognitive function. Conclusions In SAE, Erbin inhibits IRE1/Xbp1s pathway activity and reduces the ER Ca²⁺ influx to the cytoplasm, reducing microglial pyroptosis.
Article
Full-text available
Oxidative stress has been linked with a variety of diseases, being involved in the debut and/or progress of several neurodegenerative disorders. This review intends to summarize some of the findings that correlate the overproduction of reactive oxygen species with the pathophysiology of Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. Oxidative stress was also noted to modify the inflammatory response. Even though oxidative stress and neuroinflammation are two totally different pathological events, they are linked and affect one another. Nonetheless, there are still several mechanisms that need to be understood regarding the onset and the progress of neurodegenerative diseases in order to develop efficient therapies. As antioxidants are a means to alter oxidative stress and slow down the symptoms of these neurodegenerative diseases, the most common antioxidants, enzymatic as well as non-enzymatic, have been mentioned in this paper as therapeutic options for the discussed disorders.
Article
Diastolic Ca2+ leak due to cardiac ryanodine receptor (RyR2) hyperactivity has been widely documented in chronic ischemic heart disease (CIHD) and may contribute to ventricular tachycardia (VT) risk and progressive left-ventricular (LV) remodeling. Here we test the hypothesis that targeting RyR2 hyperactivity can suppress VT inducibility and progressive heart failure in CIHD by the RyR2 inhibitor dantrolene. METHODS AND RESULTS: CIHD was induced in C57BL/6 J mice by left coronary artery ligation. Four weeks later, mice were randomized to either acute or chronic (6 weeks via implanted osmotic pump) treatment with dantrolene or vehicle. VT inducibility was assessed by programmed stimulation in vivo and in isolated hearts. Electrical substrate remodeling was assessed by optical mapping. Ca2+ sparks and spontaneous Ca2+ releases were measured in isolated cardiomyocytes. Cardiac remodeling was quantified by histology and qRT-PCR. Cardiac function and contractility were measured using echocardiography. Compared to vehicle, acute dantrolene treatment reduced VT inducibility. Optical mapping demonstrated reentrant VT prevention by dantrolene, which normalized the shortened refractory period (VERP) and prolonged action potential duration (APD), preventing APD alternans. In single CIHD cardiomyocytes, dantrolene normalized RyR2 hyperactivity and prevented spontaneous intracellular Ca2+ release. Chronic dantrolene treatment not only reduced VT inducibility but also reduced peri-infarct fibrosis and prevented further progression of LV dysfunction in CIHD mice. CONCLUSIONS: RyR2 hyperactivity plays a mechanistic role for VT risk, post-infarct remodeling, and contractile dysfunction in CIHD mice. Our data provide proof of concept for the anti-arrhythmic and anti-remodeling efficacy of dantrolene in CIHD.
Article
Background: Dantrolene binds to the Leu601-Cys620 region of the N-terminal domain of cardiac ryanodine receptor (RyR2), which corresponds to the Leu590-Cys609 region of the skeletal ryanodine receptor, and suppresses diastolic Ca2+ leakage through RyR2. Objective: We investigated whether the chronic administration of dantrolene prevented left ventricular (LV) remodeling and ventricular tachycardia (VT) after myocardial infarction (MI) by the same mechanism with the mutation V3599K of RyR2, which indicated that the inhibition of diastolic Ca2+ leakage occurred by enhancing the binding affinity of calmodulin (CaM) to RyR2. Methods and results: A left anterior descending coronary artery ligation MI model was developed in mice. Wild-type (WT) were divided into four groups: sham-operated mice (WT-Sham), sham-operated mice treated with dantrolene (WT-Sham-DAN), MI mice (WT-MI), and MI mice treated with dantrolene (WT-MI-DAN). Homozygous V3599K RyR2 knock-in (KI) mice were divided into two groups: sham-operated mice (KI-Sham) and MI mice (KI-MI). The mice were followed for 12 weeks. Survival was significantly higher in the WT-MI-DAN (73%) and KI-MI groups (70%) than the WT-MI group (40%). Echocardiography, pathological tissue, and epinephrine-induced VT studies showed that LV remodeling and VT were prevented in the WT-MI-DAN and KI-MI groups compared to the WT-MI group. An increase in diastolic Ca2+ spark frequency and a decrease in the binding affinity of CaM to the RyR2 were observed at 12 weeks after MI in the WT-MI group, although significant improvements in these values were observed in the WT-MI-DAN and KI-MI groups. Conclusions: Pharmacological or genetic stabilization of RyR2 tetrameric structure improves survival after MI by suppressing LV remodeling and proarrhythmia.
Article
Huntington's disease is the most frequent autosomal dominant neurodegenerative disorder; however, no disease-modifying interventions are available for patients with this disease. The molecular pathogenesis of Huntington's disease is complex, with toxicity that arises from full-length expanded huntingtin and N-terminal fragments of huntingtin, which are both prone to misfolding due to proteolysis; aberrant intron-1 splicing of the HTT gene; and somatic expansion of the CAG repeat in the HTT gene. Potential interventions for Huntington's disease include therapies targeting huntingtin DNA and RNA, clearance of huntingtin protein, DNA repair pathways, and other treatment strategies targeting inflammation and cell replacement. The early termination of trials of the antisense oligonucleotide tominersen suggest that it is time to reflect on lessons learned, where the field stands now, and the challenges and opportunities for the future.