Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
9
result(s) for
"Choi, Zoe Yuen-Kiu"
Sort by:
LRRK2, GBA and their interaction in the regulation of autophagy: implications on therapeutics in Parkinson's disease
by
Malki, Yasine
,
Leung, Chi-Ting
,
Choi, Zoe Yuen-Kiu
in
Alzheimer's disease
,
Apoptosis
,
Autophagy
2022
Mutations in leucine-rich repeat kinase 2 (
LRRK2
) and glucocerebrosidase (
GBA
) represent two most common genetic causes of Parkinson’s disease (PD). Both genes are important in the autophagic-lysosomal pathway (ALP), defects of which are associated with α-synuclein (α-syn) accumulation. LRRK2 regulates macroautophagy
via
activation of the mitogen activated protein kinase/extracellular signal regulated protein kinase (MAPK/ERK) kinase (MEK) and the calcium-dependent adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathways. Phosphorylation of Rab GTPases by LRRK2 regulates lysosomal homeostasis and endosomal trafficking. Mutant LRRK2 impairs chaperone-mediated autophagy, resulting in α-syn binding and oligomerization on lysosomal membranes. Mutations in
GBA
reduce glucocerebrosidase (GCase) activity, leading to glucosylceramide accumulation, α-syn aggregation and broad autophagic abnormalities.
LRRK2
and
GBA
influence each other: GCase activity is reduced in
LRRK2
mutant cells, and LRRK2 kinase inhibition can alter GCase activity in
GBA
mutant cells. Clinically,
LRRK2
G2019S mutation seems to modify the effects of
GBA
mutation, resulting in milder symptoms than those resulting from
GBA
mutation alone. However, dual mutation carriers have an increased risk of PD and earlier age of onset compared with single mutation carriers, suggesting an additive deleterious effect on the initiation of PD pathogenic processes. Crosstalk between
LRRK2
and
GBA
in PD exists, but its exact mechanism is unclear. Drugs that inhibit LRRK2 kinase or activate GCase are showing efficacy in pre-clinical models. Since LRRK2 kinase and GCase activities are also altered in idiopathic PD (iPD), it remains to be seen if these drugs will be useful in disease modification of iPD.
Journal Article
LRRK2 mutant knock-in mouse models: therapeutic relevance in Parkinson's disease
by
Malki, Yasine
,
Leung, Chi-Ting
,
Choi, Zoe Yuen-Kiu
in
Animals
,
Autophagy
,
Biomedical and Life Sciences
2022
Mutations in the leucine-rich repeat kinase 2 gene (
LRRK2
) are one of the most frequent genetic causes of both familial and sporadic Parkinson’s disease (PD). Mounting evidence has demonstrated pathological similarities between
LRRK2
-associated PD (
LRRK2
-PD) and sporadic PD, suggesting that LRRK2 is a potential disease modulator and a therapeutic target in PD.
LRRK2
mutant knock-in (KI) mouse models display subtle alterations in pathological aspects that mirror early-stage PD, including increased susceptibility of nigrostriatal neurotransmission, development of motor and non-motor symptoms, mitochondrial and autophagy-lysosomal defects and synucleinopathies. This review provides a rationale for the use of
LRRK2
KI mice to investigate the LRRK2-mediated pathogenesis of PD and implications from current findings from different
LRRK2
KI mouse models, and ultimately discusses the therapeutic potentials against LRRK2-associated pathologies in PD.
Journal Article
In vivo overexpression of synaptogyrin‐3 promotes striatal synaptic dopamine uptake in LRRK2R1441G mutant mouse model of Parkinson's disease
by
Malki, Yasine
,
Li, Lingfei
,
Kung, Michelle Hiu‐Wai
in
Accreditation
,
Alzheimer's disease
,
Antibodies
2023
Background Leucine‐rich repeat kinase 2 (LRRK2) mutation is a common genetic risk factor of Parkinson's disease (PD). Presynaptic dysfunction is an early pathogenic event associated with dopamine (DA) dysregulation in striatum of the brain. DA uptake activity of DA uptake transporter (DAT) affects synaptic plasticity and motor and non‐motor behavior. Synaptogyrin‐3 (SYNGR3) is part of the synaptogyrin family, especially abundant in brain. Previous in vitro studies demonstrated interaction between SYNGR3 and DAT. Reduced SYNGR3 expression was observed in human PD brains with unclear reasons. Methods Here, we further explored whether inducing SYNGR3 expression can influence (i) cellular DA uptake using differentiated human SH‐SY5Y neuronal cells, (ii) striatal synaptosomal DA uptake in a mutant LRRK2R1441G knockin mouse model of PD, and (iii) innate rodent behavior using the marble burying test. Results Young LRRK2 mutant mice exhibited significantly lower SYNGR3 levels in striatum compared to age‐matched wild‐type (WT) controls, resembling level in aged WT mice. SYNGR3 is spatially co‐localized with DAT at striatal presynaptic terminals, visualized by immuno‐gold transmission electron microscopy and immunohistochemistry. Their protein–protein interaction was confirmed by co‐immunoprecipitation. Transient overexpression of SYNGR3 in differentiated SH‐SY5Y cells increased cellular DA uptake activity without affecting total DAT levels. Inducing SYNGR3 overexpression by adeno‐associated virus‐7 (AAV7) injection in vivo into striatum increased ex vivo synaptosomal DA uptake in LRRK2 mutant mice and improved their innate marble burying behavior. Conclusion Brain SYNGR3 expression may be an important determinant to striatal DA homeostasis and synaptic function. Our preliminary behavioral test showed improved innate behavior after SYNGR3 overexpression in LRRK2 mutant mice, advocating further studies to determine the influence of SYNGR3 in the pathophysiology of DA neurons in PD.
Journal Article
Long-term oral glucocerebrosidase activator reduces soluble α-synuclein oligomer accumulation in Parkinsonian LRRK2 mutant mouse brain
2025
Brain accumulation of toxic soluble α-synuclein (α-syn) oligomers represents a prodromal marker of synucleinopathies in Parkinson’s disease (PD), contributing to progressive nigrostriatal neurodegeneration. Dysfunction in beta-glucocerebrosidase (GCase) and leucine-rich repeat kinase 2 (LRRK2) mutation are genetic risks for developing synucleinopathies. However, whether pharmacological GCase activation ameliorated synucleinopathies in LRRK2-PD was unexplored. Here, we showed that long-term treatment of ambroxol (ABX), a brain-penetrant GCase activator, reduced α-syn oligomer accumulation in aged mutant LRRK2
R1441G
mouse striatum. Acute ABX treatment (50 µM) increased cellular GCase enzymatic activity and reduced Ser129-α-syn phosphorylation in human SH-SY5Y cells and mutant LRRK2 mouse fibroblasts, independent to LRRK2 kinase activity. Real-time DQ-BSA assay revealed lysosomal dysfunction in mutant MEFs, which was partially attenuated by ABX treatment. Lysosomal stress by bafilomycin-A1 induced endogenous GCase activity in wildtype (WT) MEFs, which was not observed in the LRRK2 mutant. Single gavage of ABX (400 mg/kg) in aged mice achieved peak drug level in serum and brain within 6 h post-administration. Ad libitum feeding of ABX (in food pellets) over 18 weeks (average dose: 45.9 mg/kg/day) elevated brain GCase activity in both WT and mutant striatum without affecting body weight. This regimen significantly reduced α-syn oligomer level in mutant striatum to a comparable physiological level in age-matched WT without altering total α-syn and Ser129-phosphorylation levels. This is the first study demonstrating reduced α-syn oligomer accumulation by chronic treatment of GCase activator in aged mouse brains vulnerable to PD, suggesting early intervention to alter progression of synucleinopathies as a key determinant of clinical outcomes of PD.
Journal Article
Long-term inhibition of mutant LRRK2 hyper-kinase activity reduced mouse brain α-synuclein oligomers without adverse effects
by
Malki, Yasine
,
Mak, Judith Choi-Wo
,
Choi, Zoe Yuen-Kiu
in
631/378/1689/1718
,
631/80
,
692/617/375/1718
2022
Parkinson’s disease (PD) is characterized by dopaminergic neurodegeneration in nigrostriatal and cortical brain regions associated with pathogenic α-synuclein (αSyn) aggregate/oligomer accumulation. LRRK2 hyperactivity is a disease-modifying therapeutic target in PD. However, LRRK2 inhibition may be associated with peripheral effects, albeit with unclear clinical consequences. Here, we significantly reduced αSyn oligomer accumulation in mouse striatum through long-term LRRK2 inhibition using GNE-7915 (specific brain-penetrant LRRK2 inhibitor) without causing adverse peripheral effects. GNE-7915 concentrations in wild-type (WT) mouse sera and brain samples reached a peak at 1 h, which gradually decreased over 24 h following a single subcutaneous (100 mg/kg) injection. The same dose in young WT and LRRK2
R1441G
mutant mice significantly inhibited LRRK2 kinase activity (Thr73-Rab10 and Ser106-Rab12 phosphorylation) in the lung, which dissipated by 72 h post-injection. 14-month-old mutant mice injected with GNE-7915 twice weekly for 18 weeks (equivalent to ~13 human years) exhibited reduced striatal αSyn oligomer and cortical pSer129-αSyn levels, correlating with inhibition of LRRK2 hyperactivity in brain and lung to WT levels. No GNE-7915-treated mice showed increased mortality or morbidity. Unlike reports of abnormalities in lung and kidney at acute high doses of LRRK2 inhibitors, our GNE-7915-treated mice did not exhibit swollen lamellar bodies in type II pneumocytes or abnormal vacuolation in the kidney. Functional and histopathological assessments of lung, kidney and liver, including whole-body plethysmography, urinary albumin-creatinine ratio (ACR), serum alanine aminotransferase (ALT) and serum interleukin-6 (inflammatory marker) did not reveal abnormalities after long-term GNE-7915 treatment. Long-term inhibition of mutant LRRK2 hyper-kinase activity to physiological levels presents an efficacious and safe disease-modifying therapy to ameliorate synucleinopathy in PD.
Journal Article
Loss of mitochondrial Ca2+ response and CaMKII/ERK activation by LRRK2R1441G mutation correlate with impaired depolarization-induced mitophagy
by
Malki, Yasine
,
Choi, Zoe Yuen-Kiu
,
Ramsden, David B.
in
Autophagy (Cytology)
,
Biology Beyond Boundaries: Mitochondrial and Cell Death in Translational Research
,
Biomedical and Life Sciences
2024
Background
Stress-induced activation of ERK/Drp1 serves as a checkpoint in the segregation of damaged mitochondria for autophagic clearance (mitophagy). Elevated cytosolic calcium (Ca
2+
) activates ERK, which is pivotal to mitophagy initiation. This process is altered in Parkinson’s disease (PD) with mutations in leucine-rich repeat kinase 2 (LRRK2), potentially contributing to mitochondrial dysfunction. Pathogenic LRRK2 mutation is linked to dysregulated cellular Ca
2+
signaling but the mechanism involved remains unclear.
Methods
Mitochondrial damages lead to membrane depolarization. To investigate how LRRK2 mutation impairs cellular response to mitochondrial damages, mitochondrial depolarization was induced by artificial uncoupler (FCCP) in wild-type (WT) and LRRK2
R1441G
mutant knockin (KI) mouse embryonic fibroblasts (MEFs). The resultant cytosolic Ca
2+
flux was assessed using live-cell Ca
2+
imaging. The role of mitochondria in FCCP-induced cytosolic Ca
2+
surge was confirmed by co-treatment with the mitochondrial sodium-calcium exchanger (NCLX) inhibitor. Cellular mitochondrial quality and function were evaluated by Seahorse™ real-time cell metabolic analysis, flow cytometry, and confocal imaging. Mitochondrial morphology was visualized using transmission electron microscopy (TEM). Activation (phosphorylation) of stress response pathways were assessed by immunoblotting.
Results
Acute mitochondrial depolarization induced by FCCP resulted in an immediate cytosolic Ca
2+
surge in WT MEFs, mediated predominantly via mitochondrial NCLX. However, such cytosolic Ca
2+
response was abolished in LRRK2 KI MEFs. This loss of response in KI was associated with impaired activation of Ca
2+
/calmodulin-dependent kinase II (CaMKII) and MEK, the two upstream kinases of ERK. Treatment of LRRK2 inhibitor did not rescue this phenotype indicating that it was not caused by mutant LRRK2 kinase hyperactivity. KI MEFs exhibited swollen mitochondria with distorted cristae, depolarized mitochondrial membrane potential, and reduced mitochondrial Ca
2+
store and mitochondrial calcium uniporter (MCU) expression. These mutant cells also exhibited lower cellular ATP: ADP ratio albeit higher basal respiration than WT, indicating compensation for mitochondrial dysfunction. These defects may hinder cellular stress response and signals to Drp1-mediated mitophagy, as evident by impaired mitochondrial clearance in the mutant.
Conclusions
Pathogenic LRRK2
R1441G
mutation abolished mitochondrial depolarization-induced Ca
2+
response and impaired the basal mitochondrial clearance. Inherent defects from LRRK2 mutation have weakened the cellular ability to scavenge damaged mitochondria, which may further aggravate mitochondrial dysfunction and neurodegeneration in PD.
Journal Article
Transcriptional Regulation of the Synaptic Vesicle Protein Synaptogyrin-3 (SYNGR3) Gene: The Effects of NURR1 on Its Expression
by
Malki, Yasine
,
Li, Lingfei
,
Choi, Zoe Yuen-Kiu
in
Alzheimer's disease
,
Binding sites
,
Dopamine
2022
Synaptogyrin-3 (SYNGR3) is a synaptic vesicular membrane protein. Amongst four homologues (SYNGR1 to 4), SYNGR1 and 3 are especially abundant in the brain. SYNGR3 interacts with the dopamine transporter (DAT) to facilitate dopamine (DA) uptake and synaptic DA turnover in dopaminergic transmission. Perturbed SYNGR3 expression is observed in Parkinson’s disease (PD). The regulatory elements which affect SYNGR3 expression are unknown. Nuclear-receptor-related-1 protein (NURR1) can regulate dopaminergic neuronal differentiation and maintenance via binding to NGFI-B response elements (NBRE). We explored whether NURR1 can regulate SYNGR3 expression using an in silico analysis of the 5′-flanking region of the human SYNGR3 gene, reporter gene activity and an electrophoretic mobility shift assay (EMSA) of potential cis-acting sites. In silico analysis of two genomic DNA segments (1870 bp 5′-flanking region and 1870 + 159 bp of first exon) revealed one X Core Promoter Element 1 (XCPE1), two SP1, and three potential non-canonical NBRE response elements (ncNBRE) but no CAAT or TATA box. The longer segment exhibited gene promoter activity in luciferase reporter assays. Site-directed mutagenesis of XCPE1 decreased promoter activity in human neuroblastoma SH-SY5Y (↓43.2%) and human embryonic kidney HEK293 cells (↓39.7%). EMSA demonstrated NURR1 binding to these three ncNBRE. Site-directed mutagenesis of these ncNBRE reduced promoter activity by 11–17% in SH-SY5Y (neuronal) but not in HEK293 (non-neuronal) cells. C-DIM12 (Nurr1 activator) increased SYNGR3 protein expression in SH-SY5Y cells and its promoter activity using a real-time luciferase assay. As perturbed vesicular function is a feature of major neurodegenerative diseases, inducing SYNGR3 expression by NURR1 activators may be a potential therapeutic target to attenuate synaptic dysfunction in PD.
Journal Article
In vivo overexpression of synaptogyrin‐3 promotes striatal synaptic dopamine uptake in LRRK2 R1441G mutant mouse model of Parkinson's disease
2023
Leucine-rich repeat kinase 2 (LRRK2) mutation is a common genetic risk factor of Parkinson's disease (PD). Presynaptic dysfunction is an early pathogenic event associated with dopamine (DA) dysregulation in striatum of the brain. DA uptake activity of DA uptake transporter (DAT) affects synaptic plasticity and motor and non-motor behavior. Synaptogyrin-3 (SYNGR3) is part of the synaptogyrin family, especially abundant in brain. Previous in vitro studies demonstrated interaction between SYNGR3 and DAT. Reduced SYNGR3 expression was observed in human PD brains with unclear reasons.
Here, we further explored whether inducing SYNGR3 expression can influence (i) cellular DA uptake using differentiated human SH-SY5Y neuronal cells, (ii) striatal synaptosomal DA uptake in a mutant LRRK2
knockin mouse model of PD, and (iii) innate rodent behavior using the marble burying test.
Young LRRK2 mutant mice exhibited significantly lower SYNGR3 levels in striatum compared to age-matched wild-type (WT) controls, resembling level in aged WT mice. SYNGR3 is spatially co-localized with DAT at striatal presynaptic terminals, visualized by immuno-gold transmission electron microscopy and immunohistochemistry. Their protein-protein interaction was confirmed by co-immunoprecipitation. Transient overexpression of SYNGR3 in differentiated SH-SY5Y cells increased cellular DA uptake activity without affecting total DAT levels. Inducing SYNGR3 overexpression by adeno-associated virus-7 (AAV7) injection in vivo into striatum increased ex vivo synaptosomal DA uptake in LRRK2 mutant mice and improved their innate marble burying behavior.
Brain SYNGR3 expression may be an important determinant to striatal DA homeostasis and synaptic function. Our preliminary behavioral test showed improved innate behavior after SYNGR3 overexpression in LRRK2 mutant mice, advocating further studies to determine the influence of SYNGR3 in the pathophysiology of DA neurons in PD.
Journal Article
Loss of mitochondrial Ca 2+ response and CaMKII/ERK activation by LRRK2 R1441G mutation correlate with impaired depolarization-induced mitophagy
by
Malki, Yasine
,
Ramsden, David B
,
Choi, Zoe Yuen-Kiu
in
Animals
,
Calcium - metabolism
,
Calcium-Calmodulin-Dependent Protein Kinase Type 2 - genetics
2024
Stress-induced activation of ERK/Drp1 serves as a checkpoint in the segregation of damaged mitochondria for autophagic clearance (mitophagy). Elevated cytosolic calcium (Ca
) activates ERK, which is pivotal to mitophagy initiation. This process is altered in Parkinson's disease (PD) with mutations in leucine-rich repeat kinase 2 (LRRK2), potentially contributing to mitochondrial dysfunction. Pathogenic LRRK2 mutation is linked to dysregulated cellular Ca
signaling but the mechanism involved remains unclear.
Mitochondrial damages lead to membrane depolarization. To investigate how LRRK2 mutation impairs cellular response to mitochondrial damages, mitochondrial depolarization was induced by artificial uncoupler (FCCP) in wild-type (WT) and LRRK2
mutant knockin (KI) mouse embryonic fibroblasts (MEFs). The resultant cytosolic Ca
flux was assessed using live-cell Ca
imaging. The role of mitochondria in FCCP-induced cytosolic Ca
surge was confirmed by co-treatment with the mitochondrial sodium-calcium exchanger (NCLX) inhibitor. Cellular mitochondrial quality and function were evaluated by Seahorse™ real-time cell metabolic analysis, flow cytometry, and confocal imaging. Mitochondrial morphology was visualized using transmission electron microscopy (TEM). Activation (phosphorylation) of stress response pathways were assessed by immunoblotting.
Acute mitochondrial depolarization induced by FCCP resulted in an immediate cytosolic Ca
surge in WT MEFs, mediated predominantly via mitochondrial NCLX. However, such cytosolic Ca
response was abolished in LRRK2 KI MEFs. This loss of response in KI was associated with impaired activation of Ca
/calmodulin-dependent kinase II (CaMKII) and MEK, the two upstream kinases of ERK. Treatment of LRRK2 inhibitor did not rescue this phenotype indicating that it was not caused by mutant LRRK2 kinase hyperactivity. KI MEFs exhibited swollen mitochondria with distorted cristae, depolarized mitochondrial membrane potential, and reduced mitochondrial Ca
store and mitochondrial calcium uniporter (MCU) expression. These mutant cells also exhibited lower cellular ATP: ADP ratio albeit higher basal respiration than WT, indicating compensation for mitochondrial dysfunction. These defects may hinder cellular stress response and signals to Drp1-mediated mitophagy, as evident by impaired mitochondrial clearance in the mutant.
Pathogenic LRRK2
mutation abolished mitochondrial depolarization-induced Ca
response and impaired the basal mitochondrial clearance. Inherent defects from LRRK2 mutation have weakened the cellular ability to scavenge damaged mitochondria, which may further aggravate mitochondrial dysfunction and neurodegeneration in PD.
Journal Article