ArticlePDF Available

Myeloid cell-specific loss of NPC1 in mice recapitulates microgliosis and neurodegeneration in patients with Niemann-Pick type C disease

Authors:
  • German Center for Neurodegenerative Diseases (DZNE), Germany, Munich

Abstract

Niemann-Pick type C (NPC) disease is an inherited lysosomal storage disorder mainly driven by mutations in the NPC1 gene, causing lipid accumulation within late endosomes/lysosomes and resulting in progressive neurodegeneration. Although microglial activation precedes neuronal loss, it remains elusive whether loss of the membrane protein NPC1 in microglia actively contributes to NPC pathology. In a mouse model with depletion of NPC1 in myeloid cells, we report severe alterations in microglial lipidomic profiles, including the enrichment of bis(monoacylglycero)phosphate, increased cholesterol, and a decrease in cholesteryl esters. Lipid dyshomeostasis was associated with microglial hyperactivity, marked by an increase in translocator protein 18 kDa (TSPO). These hyperactive microglia initiated a pathological cascade resembling NPC-like phenotypes, including a shortened life span, motor impairments, astrogliosis, neuroaxonal pathology, and increased neurofilament light chain (NF-L), a neuronal injury biomarker. As observed in the mouse model, patients with NPC showed increased NF-L in the blood and microglial hyperactivity, as visualized by TSPO-PET imaging. Reduced TSPO expression in blood-derived macrophages of patients with NPC was measured after N -acetyl- l -leucine treatment, which has been recently shown to have beneficial effects in patients with NPC, suggesting that TSPO is a potential marker to monitor therapeutic interventions for NPC. Conclusively, these results demonstrate that myeloid dysfunction, driven by the loss of NPC1, contributes to NPC disease and should be further investigated for therapeutic targeting and disease monitoring.
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
1 of 14
NEURODEGENERATION
Myeloid cell–specific loss of NPC1 in mice recapitulates
microgliosis and neurodegeneration in patients with
Niemann- Pick type C disease
Lina Dinkel1†, Selina Hummel2†, Valerio Zenatti1, Mariagiovanna Malara1, Yannik Tillmann1,
Alessio Colombo1, Laura Sebastian Monasor1, Jung H. Suh3, Todd Logan3, Stefan Roth4,
Lars Paeger1, Patricia Hoelner5,6, Oliver Bludau5, Andree Schmidt1,6,7, Stephan A. Müller1,7,
Martina Schierer1,8, Brigitte Nuscher9, Jasenka Rudan Njavro1, Matthias Prestel1,
Laura M. Bartos2, Karin Wind- Mark1,2, Luna Slemann2, Leonie Hoermann2, Sebastian T. Kunte2,
Johannes Gnörich2, Simon Lindner2, Mikael Simons1,4,8,10, Jochen Herms1,8,11, Dominik Paquet4,8,
Stefan F. Lichtenthaler1,7,8, Peter Bartenstein2, Nicolai Franzmeier4,8,12, Arthur Liesz4,8,
Antje Grosche5, Tatiana Bremova- Ertl13,14, Claudia Catarino15, Skadi Beblo16, Caroline Bergner17,
Susanne A. Schneider13, Michael Strupp13, Gilbert Di Paolo3,
Matthias Brendel1,2,8*, Sabina Tahirovic1*
Niemann- Pick type C (NPC) disease is an inherited lysosomal storage disorder mainly driven by mutations in the NPC1
gene, causing lipid accumulation within late endosomes/lysosomes and resulting in progressive neurodegeneration.
Although microglial activation precedes neuronal loss, it remains elusive whether loss of the membrane protein NPC1
in microglia actively contributes to NPC pathology. In a mouse model with depletion of NPC1 in myeloid cells, we re-
port severe alterations in microglial lipidomic proles, including the enrichment of bis(monoacylglycero)phosphate,
increased cholesterol, and a decrease in cholesteryl esters. Lipid dyshomeostasis was associated with microglial hy-
peractivity, marked by an increase in translocator protein 18 kDa (TSPO). These hyperactive microglia initiated a path-
ological cascade resembling NPC- like phenotypes, including a shortened life span, motor impairments, astrogliosis,
neuroaxonal pathology, and increased neurolament light chain (NF- L), a neuronal injury biomarker. As observed in
the mouse model, patients with NPC showed increased NF- L in the blood and microglial hyperactivity, as visualized
by TSPO- PET imaging. Reduced TSPO expression in blood- derived macrophages of patients with NPC was measured
after N- acetyl- l- leucine treatment, which has been recently shown to have benecial eects in patients with NPC,
suggesting that TSPO is a potential marker to monitor therapeutic interventions for NPC. Conclusively, these results
demonstrate that myeloid dysfunction, driven by the loss of NPC1, contributes to NPC disease and should be further
investigated for therapeutic targeting and disease monitoring.
INTRODUCTION
Niemann- Pick type C (NPC) disease is an inherited metabolic lyso-
somal storage disorder that manifests with progressive neurodegen-
eration. NPC is caused by autosomal recessive mutations in the NPC1
(95%) or NPC2 (5%) gene. e encoded proteins reside in late endo-
somal (LE)/lysosomal compartments and jointly regulate cholesterol
egress (1). It has been postulated that the small luminal protein NPC2
transfers cholesterol to the transmembrane protein NPC1, which expor ts
cholesterol out of LE/lysosomes (24). Disease- causing mutations
in NPC1 lead to disturbances in cellular lipid metabolism, result-
ing in storage of adverse lipids, including unesteried cholesterol or
glycosphingolipids (58).
NPC manifests mostly in early childhood with a complex, heterog-
enous, and oen severe clinical phenotype. Neurological symptoms of
NPC include ataxia, dystonia, supranuclear saccade and gaze palsy,
epileptic seizures, and cognitive impairment, leading to premature
death (1,9,10). Pathological hallmarks include neurobrillary tangles,
axonal swellings, neuroinammation, and hypomyelination (1, 1114).
Molecular mechanisms such as aberrant endolysosomal tracking
and autophagy seem to contribute to NPC pathology (1519). e
neurodegenerative aberrations are region specic, encompassing vul-
nerable Purkinje cells in the cerebellum and retinal pathology. Retinal
1German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich,
Germany. 2Department of Nuclear Medicine, LMU University Hospital, LMU Munich,
81377 Munich, Germany. 3Denali Therapeutics Inc., South San Francisco, CA 94080, USA.
4Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU
Munich, 81377 Munich, Germany. 5Department of Physiological Genomics, Biomed-
ical Center (BMC), Faculty of Medicine, Ludwig- Maximilians- Universität München,
82152 Planegg- Martinsried, Germany. 6Graduate School of Systemic Neurosciences,
Ludwig Maximilian University, 82152 Planegg- Martinsried, Germany. 7Neuropro-
teomics School of Medicine and Health, Klinikum rechts der Isar, Technical Univer-
sity of Munich, 81675 Munich, Germany. 8Munich Cluster for Systems Neurology
(SyNergy), 81377 Munich, Germany. 9Metabolic Biochemistry, Biomedical Center
(BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany. 10Institute of
Neuronal Cell Biology (TUM- NZB), Technical University of Munich, 80802 Munich,
Germany. 11Center for Neuropathology and Prion Research, Ludwig- Maximilians-
University München, 81377 Munich, Germany. 12Department of Psychiatry and
Neurochemistry, University of Gothenburg, Sahlgrenska Academy, Institute of
Neuroscience and Physiology, SE- 413 90 Mölndal and Gothenburg, Sweden. 13De-
partment of Neurology, LMU University Hospital, LMU Munich, 81377 Munich,
Germany. 14Department of Neurology, University Hospital Bern, 3010 Bern, Switzer-
land. 15Friedrich Baur Institute, Department of Neurology, LMU University Hospital,
LMU Munich, 80336 Munich, Germany. 16Center for Pediatric Research Leipzig, De-
partment of Women and Child Health, Hospital for Children and Adolescents, Uni-
versity Hospital Leipzig; Leipzig University Center for Rare Diseases, 04103 Leipzig,
Germany. 17Department of Neurology, University Hospital Leipzig, 04103 Leipzig,
Germany.
*Corresponding author. Email: matthias. brendel@ med. uni- muenchen. de (M.B.); sa-
bina. tahirovic@ dzne. de (S.T.)
†These authors contributed equally to this work.
Copyright © 2024 The
Authors, some rights
reserved; exclusive
licensee American
Association for the
Advancement of
Science. No claim to
original U.S.
Government Works
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
2 of 14
axonal degeneration has been shown to correlate with motor symp-
toms and disease progression (20,21).
Currently, there is no cure for NPC. In many countries, the gan-
glioside synthesis inhibitor miglustat is the rst approved treatment
with potential to ameliorate disease progression (22). In recent clinical
trials, it was shown that the modied amino acid N- acetyl- - leucine
(NALL) improved clinical signs and symptoms in patients with NPC
(23,24). Besides therapeutics, there is also an unmet need to accel-
erate the diagnosis of NPC and enable the precise monitoring of the
therapeutic benets (25).
Early- onset human NPC disease is well recapitulated in the whole-
body Npc1 knockout mouse model (here abbreviated as Npc1/) that
provides an excellent tool for preclinical studies (26,27). is model
displays prominent neuronal pathology, such as axonal spheroids
and region- specic neurodegeneration, particularly evident in the
cerebellum (Purkinje cell loss), thalamus, and retina (21,2830).
Further pathological hallmarks of NPC are recapitulated in mice,
such as demyelination, and severe microgliosis and astrogliosis, which
are emerging as active players in disease pathology (16,3134). Ben-
ecial eects of NALL treatment have also been reported in Npc1/
mice, including reduced lipid storage and improvements in energy
metabolism and behavioral skills associated with reduced microglial
activation (35).
Cell type–specic depletion of NPC1 and rescue experiments
identied a cell- autonomous role of NPC1 in neurons and oligo-
dendrocytes, whereas its cell- autonomous role in astrocytes is still
debated (36). Our recent work began to address the role of NPC1
in microglia and demonstrated the requirement of NPC1 for en-
dolysosomal lipid tracking. We showed an accumulation of lipid-
enriched substrates such as myelin debris within LE/multivesicular
bodies (MVBs) of Npc1/ microglia and demonstrated that mi-
croglial molecular and functional alterations occur before neuro-
degeneration (16).
It is now unknown how neuroinammation and microglial malfu nc-
tion contribute to neurodegeneration in NPC. To monitor neuro-
inammation in vivo, translocator protein 18 kDa (TSPO) is commonly
used as a positron emission tomography (PET) ligand for visual-
izing microglial activation (37). Although the mitochondrial pro-
tein TSPO is also expressed by endothelial cells and astrocytes,
upon inammatory and neurodegenerative conditions, it oen be-
comes enriched in microglia, suggesting its suitability for monitor-
ing microglial activation (38,39). Increased neuroinammation in
the white matter of patients with NPC has been detected by TSPO-
PET imaging, but no association with disease severity was ob-
served (13), questioning whether microglial activation contributes
to NPC disease progression.
Here, by exploring a mouse model with a depletion of NPC1
in myeloid cells, we showed that NPC1 regulates microglial lipid
homeostasis and that its loss is sucient to trigger microglial acti-
vation, followed by astrogliosis and neuroaxonal pathology. e
neuroaxonal pathology is also supported by the increase of its
blood surrogate marker neurolament light chain (NF- L) in the
mouse model and in patients with NPC. Our work shows a patho-
logical cross- talk between microglia, astrocytes, and neurons, sup-
porting the contribution of these various brain cells to NPC
disease. We further reveal that microglial dysfunction, recapit-
ulated in blood- derived macrophages of patients with NPC,
was ameliorated after a 6- week treatment with NALL, as deter-
mined by reduced TSPO expression and a trend toward reduced
phagocytosis. ese ndings suggest that microglia actively con-
tribute to NPC disease and that blood- derived macrophages show
similar pathological alterations, oering an ideal tool to monitor
therapeutic interventions.
RESULTS
Loss of NPC1 in myeloid cells compromises endolysosomal
tracking and microglial lipid metabolism
To analyze a cell- autonomous pathomolecular function of NPC1
in microglia, we used our recently generated conditional mouse
model Npc1ox/ox; Cx3cr1Cre (Cre+), which depletes NPC1 in mi-
croglia, and other Cx3cr1- expressing myeloid cells (16,40,41). In
contrast with the whole- body Npc1 knockout (Npc1/) that reaches
humane end points at the age of approximately 3 months (42), the
myeloid- specic model reaches humane end points at the age of 11 to
12 months (g. S1, A and B). To characterize a key cellular pheno-
type in NPC, namely, enhanced phagocytosis and impairment in
lipid tracking, we cultured Cre and Cre+ microglia in the pres-
ence of pHrodo- labeled myelin. Cre+ microglia displayed signi-
cantly (P < 0.001) enhanced phagocytic uptake of myelin (Fig. 1, A
and B). Cre microglia were able to process myelin into lipid drop-
lets, as judged by positive staining for neutral lipids (Nile Red) at
the cell periphery (Fig. 1, C and D). In contrast, lipid droplets were
almost completely absent at the cell periphery of Cre+ microglia.
Electron microscopy conrmed impairments in myelin turnover
into lipid droplets and its accumulation in LE/MVBs in Cre+ mi-
croglia (Fig. 1E). e lipid accumulation within LE/MVBs was
further corroborated by the nding that the number of lysosomes
was decreased in Cre+ microglia under this myelin- fed condition
(Fig. 1, E and F).
Previous work has demonstrated altered lipidomic proles in
NPC at the level of the whole brain (43), but lipid proles were not
analyzed in specic brain cell types. To characterize the lipid pro-
le of microglia from Cre and Cre+ mice at early stages (2 months)
of disease pathology, we performed a lipidomic analysis using
liquid chromatography–mass spectrometry (LC- MS) (table S1).
We detected increased cholesterol and decreased cholesteryl esters
(CEs) in Cre+ microglia (Fig. 1, G to I), in accordance with the
absence of Nile Red–positive lipid droplets observed in vitro (Fig.
1, C and D). Multiple bis(monoacylglycero)phosphate (BMP) spe-
cies, which are lipids enriched in MVBs and linked to cholesterol
dyshomeostasis in NPC (4446), were prominently increased in
Cre+ microglia (Fig. 1, G and H). Further, we detected ceramide
derivates such as lactosylceramide, myelin- enriched lipids like sul-
fatides, and gangliosides among the most up- regulated lipids in
Cre+ microglia (Fig. 1, G and H). In addition to CEs, we detected
reduced coenzyme Q10, phospholipids, cholesterol sulphate, and
di- and triglycerides (Fig. 1, G and I). Signicantly increased cho-
lesterol (P < 0.05) and a trend toward an increase in GM2 were
also quantied by immunocytochemistry of cultured Cre+ microg-
lia (Fig. 1, J and K).
To estimate the potential contribution of the compromised brain
environment in NPC to the lipidomic prole of microglia, we per-
formed a lipidomic analysis in Npc1/ mice at 2 months of age,
corresponding to a late pathological stage with severe neurode-
generation (fig. S2A and table S2). The lipidomic profile of
Npc1/ microglia and Cre+ microglia showed a high overlap, in-
cluding an increase in cholesterol and BMP species (g. S2, A to C).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
3 of 14
We further investigated the lipid prole of microglia from early
pathological stages (1 week) of Npc1/ mice, which resembled
the lipidomic prole of Npc1/ microglia at late pathological stages,
underscoring increased cholesterol and BMP species and decreased
CE (g. S2D and table S3). is is in line with an increased LE/
MVB area, whereas the number of lysosomes was not changed (g.
S2, E to G). Several CE species showed the highest reduction
at the early pathological stage, suggesting that CEs were more
abundant during development. Our results demonstrate that lipid
changes, as well as enrichment of LE/MVBs in microglia, occur
very early in NPC disease and are driven by the cell- autonomous
function of NPC1.
TSPO- PET imaging reveals strong microglial activation in
mice lacking NPC1 in myeloid cells
Next, we investigated whether lipid tracking defects are associated
with microglial activation and neuroinammation. To visualize
microglial activation, we performed TSPO- PET imaging at early
(2 months), intermediate (7 months), and late (11 months) stages of
disease pathology in Cre and Cre+ mice. We detected an increase in
Cre
-
Cre
+
0
20
40
60
80
100
Lipid droplet
-
positive cells (%)
GM2 Filipin
ABC
D
CD68 GM2 filipin
Cre+
Cre
-
1200× 5000×
G
J
CD68
Cre+
Cre
-
H
Coenzyme Q10CE(18:1)
CholesterolBMP(18:1/18:1
)
LacCer(d18:1/18:0) GM2(d38:1)
PS(16:0_22:6) TG(18:0_36:2)
I
E
Cre
-
Cre+
Myelin
F
Cre
-
+ CD
Cre+ + CD
K
Cre
-
Cre+
CD
--
++
Myelin
Nile Red
CD68
my.Nile CD68 Red
Cre+
Cre
-
H
**
*
**
*
Cre
-
Cre
+
0.00
0.02
0.04
0.06
0.08
0.10
Lysosomes pe
0
100
200
300
Mean intensityper cell (%)
Cre
-
Cre+
0
1
2
3
4
5
Cre
-
Cre+
0.0
0.5
1.0
1.5
2.0
2.5
Cre
-
Cre+
0
1
2
3
4
5
Cre
-
Cre+
-
10
-
8
-
6
-
4
-
2
0
Cre
-
Cre
+
-
6
-
4
-
2
0
2
Cre
-
Cre
+
0
1
2
3
4
Cre
-
Cre
+
-
5
-
4
-
3
-
2
-
1
0
Cre
-
Cre
+
-
4
-
2
0
2
4
RUV-adjusted area ratios (log2)
RUV-adjusted area ratios (log2)
FilipinGM2
0
2000
4000
6000
Mean intensitypercell(a.u.)
Cre
-
Cre
+
ns
Down
Up
Stable
Fig. 1. Loss of NPC1 impairs myelin tracking
and lipid homeostasis in microglia. (A) Analysis
of phagocytic uptake of pHrodo- labeled myelin
in cultured Cre (n = 3) and Cre+ (n = 3) microglia.
Cytochalasin D (CD) was used as a negative control.
Scale bars, 200 μm. (B) Quantication of the uo-
rescent mean intensity of pHrodo 6 hours after feed-
ing. Values were normalized to Cre controls and
are depicted as means ± SEM, and a one- way analysi s
of variance (ANOVA) with Tukey’s multiple compari-
son test was performed (***P < 0.001). (C) Immu-
nocytochemical analysis of myelin process ing in
cultured Cre and Cre+ microglia. Microglia were
fed with uorescently labeled myelin (my., green), and
the presence of lipid droplets (Nile Red, magenta)
was analyzed after 48 hours. Cells were immunos-
tained using CD68 (white). A representative microg-
lial cell is depicted, and one lipid droplet is marked
with an arrow. Hoechst (H) was used as the nuclear
stain (cyan). Scale bars, 10 μm. (D) Quantication of
lipid droplet–positive cells in myelin- fed Cre (n =
3) and Cre+ (n = 3) microglia. Data are presented as
means ± SEM. A two- tailed unpaired Student’s t test
was performed (***P < 0.001). (E) Representative
electron microscopy images of myelin- fed Cre
and Cre+ microglia 48 hours after feeding. Myelin
accumulation is shown by yellow asterisks, and
lysosomes are indicated by red arrows. Dierent
identities of lipid droplets are visualized by blue
and green arrows. Scale bars, 2 μm (1200×) and 1 μm
(5000×). (F) Quantication of lysosomal number
in myelin- fed Cre+ (n = 3) and Cre microglia (n =
3). Data are presented as means ± SEM, and a two-
tailed unpaired Student’s t test was performed (*P <
0.05). (G) Lipidomic analysis of acutely isolated
microglia from 2- month- old Cre (n = 5) and Cre+
mice (n = 5). Signicantly (adjusted P value < 0.05)
increased (up) or decreased (down) lipids are de-
picted in red and blue, respectively. Lipids that are
not signicantly changed (adjusted P value > 0.05)
are depicted in gray (stable). The dotted line repre-
sents a P value of 0.05. (H and I) Selected up-
regulated (H) and down- regulated (I) lipids from
the lipidomic analysis of Cre and Cre+ microglia.
The log2- transformed RUV (removal of unwanted
variables)–adjusted area ratios are depicted as
means ± SEM (the nonadjusted P value is depicted,
*P < 0.05, **P < 0.01, ***P < 0.001). (J) Cultured
Cre (n = 3) and Cre+ (n = 3) microglia isolated from 2- month- old mice were immunostained against CD68 (magenta) and GM2 lipid (green). Cholesterol was stained using
lipin (cyan). Scale bars, 10 μm. (K) Quantication of the uorescent mean intensity [arbitrary unit (a.u.)] of lipin and GM2 was measured using ImageJ and is represented
as means ± SEM. A two- tailed unpaired Student’s t test was performed for each lipid (ns, P > 0.05; *P < 0.05).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
4 of 14
TSPO- PET signals in the midbrains and brainstems of Cre+ mice at
intermediate disease stages (7 months) and across all analyzed brain
regions at late stages of disease pathology (Fig. 2, A and B). A com-
parison of TSPO- PET signals in Cre+ and Cre mice revealed the
midbrain and brainstem as hotspot regions of microglial activation
upon loss of NPC1 (Fig. 2C). To corroborate the microglial contribu-
tion to increased TSPO- PET signals, we used our recently established
single- cell radiotracing technology (scRadiotracing) (47). Cre mice
showed enriched TSPO tracer uptake in microglia compared with
astrocytes (Fig. 2D), arguing for microglia- specic uptake of the
TSPO- PET tracer. Moreover, microglia isolated from the Cre+ mice
showed a 1.7- fold increased TSPO tracer uptake per cell compared
with the Cre mice (Fig. 2D), implying that the increased TSPO- PET
imaging signals not only were a consequence of microgliosis but also
indicated higher TSPO per cell. ese results were further supported
by immunohistochemical analysis, revealing a signicantly (P <
0.001) increased expression of TSPO in Iba1- positive microglia of
Cre+ mice at the age of 12 months that was not observed at 2 months
(Fig. 2, E to H). In contrast to microglia, we did not detect changes in
TSPO expression in astrocytes (g. S3, A and B).
Next, we asked whether microglial activation occurs also at ear-
lier time points by performing time- dependent immunohistochem-
ical analysis using microglial LE/lysosomal marker CD68. ese
experiments determined a signicantly (P < 0.001) increased CD68
coverage in Cre+ mice already at an early stage (Fig. 2I and g. S4, A
and B), which was further enhanced at intermediate and late patho-
logical stages in a region- specic manner, with the midbrain and
brainstem being most aected (Fig. 2J and g. S4, C to F). Similar
time- and region- dependent phenotypic changes in microglia were
also evident by increased Iba1 coverage in Cre+ mice (g. S4, G and
H). Along with enhanced microglial activity, phagocytic cups were
prominent in Cre+ mice at late stages, providing in vivo evidence of
increased phagocytosis (g. S4, I and J). us, microglial activation
and alterations in the endolysosomal pathway can be captured in
Cre+ mice already at early pathological stages, whereas alterations of
TSPO become evident at intermediate and late stages of pathology.
Observed regional vulnerabilities in the Cre+ mice dier from the
Npc1/ mice, which showed the highest CD68 immunoreactivity in
the cerebellum (g. S5), with a well- described neurodegeneration of
Purkinje cells (29).
To further study the dierential vulnerability, we compared the
cerebellar with the cerebral (brain without cerebellum) proteome
in Cre and Cre+ mice at late pathological stages. e number of
false discovery rate (FDR)–adjusted signicantly altered proteins
was higher in the cerebrum (273 proteins) compared with the
cerebellum (36 proteins) (g. S6A and table S4). Both proteomes
included an enrichment of complement factors (C1QA, C1QB,
C1QC, and C4B), lysosomal proteins (HEXA, HEXB, MAN2B1,
CTSZ, CTSH, and NAGA), disease- associated microglia (DAM)
proteins (LGALS3, APOE, and GRN), and astrocytic glial brillary
acidic protein (GFAP) in Cre+ mice. e microglial homeostatic
protein P2RY12, together with proteins associated with neuro-
nal function (SLC6A5, SYT2, and KCNC3), were significantly
decreased (FDR- adjusted P < 0.05) in the Cre+ cerebral proteome
(g. S6A). e underlying altered molecular pathways are involved
in regulation of immune responses and development and prolif-
eration of glial cells (g. S6B). ese results suggest that microglial
loss of NPC1 has profound eects on brain cell homeostasis espe-
cially in the cerebrum.
Microglia- to- astrocyte propagation of pathological insults
in mice lacking NPC1 in myeloid cells
Prominent microglial pathology upon loss of NPC1 may have direct
consequences on the homeostasis of other brain cells. Microglia- to-
astrocyte propagation of pathological insults is a well- supported
phenomenon (48), and the brain proteome of Cre+ mice revealed
phenotypic changes in astrocytes (g. S6, A and B). To track poten-
tial changes in astrocytes in vivo, we explored monoamine oxidase B
(MAOB)–PET as an astrocyte imaging tool using the tracer [18F]
uorodeprenyl- D2 (49). We observed an increase in the MAOB-
PET signal in all analyzed brain regions at intermediate and late
pathological stages in Cre+ mice (Fig. 3, A and B), with the midbrain
and brainstem being the hotspot regions of MAOB- positive astro-
cytic reactivity (Fig. 3C).
e regional overlap of the highest TSPO- PET (Fig. 2C) and
MAOB- PET signal (Fig. 3C) suggests that astrocyte reactivity
may be a response to pathological alterations in microglia. Using
scRadiotracing technology, we conrmed that astrocytes are a ma-
jor cellular source of the increased MAOB- PET signal (Fig. 3D). In
contrast to microglia, cellular uptake of the MAOB radiotracer was
comparable in the astrocyte- enriched fraction from Cre and Cre+
mice (Fig. 3D), suggesting that the increased MAOB- PET signal is
a consequence of astrogliosis. e astrocytic response was further
veried by Western blot and immunohistochemical analyses of GFAP,
a common astrocytic marker (50). Western blot analysis revealed a
trend toward increased GFAP immunoreactivity in Cre+ mice al-
ready at early pathological stages, and signicantly increased GFAP
was detected at intermediate (P < 0.05) and late (P < 0.01) patho-
logical stages (Fig. 3, E and F). Similar results were also obtained
by immunohistochemical analysis (Fig. 3, G and H). Despite the
prominent alterations in microglia and astrocytes, we did not de-
tect any obvious dierence in myelination at late pathological stages
in Cre+ mice by looking at the oligodendrocyte marker 2ʹ, 3ʹ -cyclic
nucleotide phosphodiesterase (CNPase) (g. S7), suggesting that
microglial NPC1 depletion is not a major driver ʹof myelination de-
fects observed in NPC (16).
Region- specic neurodegeneration in mice lacking NPC1 in
myeloid cells
Neurodegeneration in NPC is best illustrated by the loss of Purkinje cells
in the cerebellum as well as retinal degeneration (21,29). To determine
the impact of myeloid cell–specic loss of NPC1 on neuronal function,
we rst analyzed the morphology of the Purkinje cell layer by immuno-
histochemistry using the marker calbindin. We revealed an intact
Purkinje cell layer even at late pathological stages in Cre+ mice (Fig. 4A).
Total dendrite length of Purkinje cells and branching pattern quantied
by Sholl analysis showed no dierences between Cre and Cre+ mice (Fig.
4, B to E). is result supports the hypothesis that neurodegeneration of
Purkinje cells observed in Npc1/ mice is likely driven by the loss of the
neuronal (40) rather than microglial NPC1 function. Next, we examined
neurodegeneration in the well- stratied retinal tissue. Although reti-
nal layer thickness was not signicantly altered at late pathological
stages in Cre+ mice (Fig. 4, F and G), we observed a trend toward a
reduction in calretinin- positive neurons and a signicantly (P < 0.01)
reduced number of protein kinase Cα–positive neuronal popula tions
in the inner nuclear layer (Fig. 4H). Similar to other analyzed brain
regions, the retina had increased CD68 immunoreactivity (g. S8A).
To characterize neuronal function across dierent pathological
stages and brain regions, we determined glucose uptake by applying
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
5 of 14
Cre
-
Cre+
0
20
40
60
80
TSPO perIba1-positive cell )
ns
Cre
-
Cre
+
0
100
200
300
400
500
TSPO perIba1-positive cell(µm³)
0
10
-
6
10
-
6
10
-
6
Cellular TSPO tracer uptake
(%ID * weight/cell)
Cre+
Cre
-
Microglia-
enriched
fraction
Astrocyte-
enriched
fraction
ns
F
CD68
Cre
-
12
1
2
Cre
+
34
43
Ctx Ctx
bs bs
123
4
Cre
+
CD68
Cre
-
Ctx Ctx
bs bs
12 43
A
Cre+
Cre
-
)VUS( TEP-OPST
1.0
shtnom 2
7 months11 months
0.1
B
C
11 months
DE
TSPO-PET (SUV)
2 months
Midbrain
GH
12 months
Midbrain
Iba1TSPO
Cre
-
Cre+
IJ
2 months 12 months
Iba1TSPO
Cre
-
Cre+
2 months 12 months
TSPO Iba1
TSPO Iba1
0% 80%
Cre+ vs. Cre
-
TSPO-PET (SUV)
Cortex Midbrain Brainstem Cerebellum
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
Age (months)
G*A,T = 1.962
ns
2711
ns
ns
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
Age (months)
G*A,T = 2.001
ns
2711
ns
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
Age (months)
G*A,T = 1.894
ns
2711
ns
ns Cre+
Cre
-
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
Age (months)
G*A,T = 2.574
P = 0.0134
2711
ns
Fig. 2. TSPO- PET reveals region- specic microglial hyperactivity. (A) TSPO- PET images of Cre and Cre+ mouse brains imaged at 2, 7, and 11 months of age. The shown sagit-
tal brain sections represent group average images of the standardized uptake value (SUV) projected on a standard MRI T1w atlas. (B) Quantication of individual TSPO- PET SUVs
derived from the cortex, midbrain, brainstem, and cerebellum. Dotted lines represent linear associations between age and TSPO- PET quantication with their 95% condence
interval (lled area). The genotype × age (G * A) interaction was calculated. A multiple linear regression was performed for statistical analysis with the following sample sizes:
Cre n = 8 and Cre+ n = 5 for 2 months, Cre n = 10 and Cre+ n = 8 for 7 months, and Cre n = 12 and Cre+ n = 6 for 11 months (ns, P > 0.05; *P < 0.05; **P < 0.01; ***P
< 0.001). (C) Percentage dierence in TSPO- PET (SUV) between Cre+ and Cre mice at 11 months of age. (D) Quantication of cellular TSPO tracer uptake in microglia- and
astrocyte- enriched fractions of 12- month- old mice by scRadiotracing. Data are presented as means ± SEM, and a one- way ANOVA with Tukey’s multiple comparison test was
performed (ns, P > 0.05; **P < 0.01; ***P < 0.001) with the following sample sizes: Cre n = 8 and Cre+ n = 6 (microglia- enriched fraction), and Cre n = 8 and Cre+ n = 5 (astrocyte-
enriched fraction). (E) Immunohistochemical analysis of TSPO (green) in Iba1- positive cells (magenta) in the midbrains of Cre and Cre+ mice at 2 months of age. Scale bars,
50 μm. (F) Quantication of TSPO volume per Iba1- positive cell in Cre (n = 3) and Cre+ (n = 3) mice at 2 months of age. Data are presented as means ± SEM, and a two- tailed
unpaired Student’s t test was performed (ns, P > 0.05). (G) Immunohistochemical analysis of TSPO (green) and Iba1 (magenta) in the midbrains of Cre and Cre+ mice at 12 months
of age. Scale bars, 50 μm. (H) Quantication of TSPO volume per Iba1- positive cell of Cre (n = 3) and Cre+ (n = 3) mice at 12 months of age. Data are presented as means ± SEM,
and a two- tailed unpaired Student’s t test was performed (***P < 0.001). (I and J) Sagittal brain sections of 2- (I) and 12 (J)- month- old Cre and Cre+ mice were immunostained
against CD68 (white). Numbers 1 to 4 represent zoom- ins from the respective tile scans. Scale bars, 2000 μm (tile scans) and 500 μm (zoom- ins). Ctx, cortex; bs, brainstem.
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
6 of 14
2- deoxy- 2- [18F]uoro- D- glucose (FDG)–PET imaging. Reduced glu-
cose metabolism is used as a surrogate marker of neuronal dysfunction
and neurodegeneration (51). e FDG- PET signal was signicantly
(P < 0.05) reduced at intermediate (cortex and midbrain) or late (brain-
stem) pathological stages in Cre+ mice (Fig. 5, A and B), illustrating a
widespread neuronal dysfunction upon loss of NPC1 in myeloid cells.
Next, we assessed NF- L in the serum, a well- accepted biomarker for
neuroaxonal injury (52,53). In line with the reduced FDG- PET, we
detected signicantly increased NF- L in sera at intermediate (P < 0.01)
and late (P < 0.001) pathological stages in Cre+ mice (Fig. 5C). Im-
munohistochemical analysis of neurolament heavy chain (NF200)
revealed a signicant (P < 0.001) accumulation at intermediate patho-
logical stages in the midbrains and brainstems of Cre+ mice (Fig. 5, D
and E), which indicates axonal swelling, a well- described neuronal pa-
thology in NPC disease (31,34). Neuronal defects were further sup-
ported by accumulation of the autophagy marker p62 in NeuN- positive
cells in the cortex, midbrain, and brainstem (Fig. 5, F and G), which
was not detected in the cerebellum (g. S8B). To test whether defects
caused by activated microglia lacking NPC1 have consequences on be-
havior, we assessed motor performance using a rotarod test. Cre+ mice
showed reduced latency to fall at intermediate pathological stages
compared with Cre mice (Fig. 5H).
0
10
-
10
10
-
10
10
-
10
10
-
10
Cellular MAOB tracer uptake
(%ID * weight/cell)
Cre+
Cre
-
ns
ns
ns
Astrocyte-
enriched
fraction
Astrocyte-
depleted
fraction
0
2
4
6
8
10
GFAP fold change
ns
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Age (months)
MAOB-PET (SUV)
G * A
T = 1.897
ns
2711
ns
A
D
MAOB-PET (SUV)
1.0
0.1
Cre+
Cre
-
shtnom 2
7 months
11 months
Cortex Midbrain Brainstem
B
C
0% 80%MAOB-PET (SUV)
Cre+ vs. Cre
-
11 months
E
F
GFAP
50 kDa -
37 kDa -
Cre
-
Cre+
37 kDa - GAPDH
GFAP
GAPDH
GFAP
GAPDH
50 kDa -
37 kDa -
37 kDa -
shtnom 2
shtnom 7
shtnom 21
2 months
50 kDa -
37 kDa -
37 kDa -
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Age (months)
G * A
T = 3.038
P = 0.005
2711
ns
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Age (months)
G * A
T = 2.227
P = 0.03
2711
ns
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Age (months)
G * A
T = 1.628
ns
2711
ns
Cerebellum
Cre+
Cre
-
G
Cre
-
Cre+
GFAP
H
CortexMidbrainBrainstem
7 months
Cre
-
Cre+
12 months
Cre
-
Cre+
2 months 7 months 12 months
Cortex Midbrain Brainstem
0.0
0.2
0.4
0.6
0.8
1.0
1.2
GFAP coverage (%)
ns
ns
Cortex Midbrain Brainstem
0
2
4
6
8
10
12
GFAP coverage (%)
Cortex Midbrain Brainstem
0
5
10
15
20
GFAP coverage (%)
Fig. 3. NPC1- decient microglia cause astro-
gliosis. (A) MAOB- PET images of Cre and Cre+
mouse brains at 2, 7, and 11 months of age. The
shown sagittal brain sections represent group
average images of SUVs projected on a stan-
dard MRI T1w atlas. (B) Quantication of indi-
vidual MAOB- PE T SUVs derived from the cortex,
midbrain, brainstem, and cerebellum. Dotted
lines represent linear associations between age
and MAOB- PET quantication with their 95%
condence interval (lled area). The genotype ×
age (G * A) interaction was calculated. A multi-
ple linear regression was performed for statisti-
cal analysis with the following sample sizes for
the cortex, midbrain, and brainstem: Cre n = 5
and Cre+ n = 4 for 2 months, Cre n = 5 and Cre+
n = 6 for 7 months, and Cre n = 6 and Cre+ n =
5 for 11 months. For the cerebellum, the sample
sizes were Cre n = 5 and Cre+ n = 4 for 2 months,
Cre n = 6 and Cre+ n = 6 for 7 months, and Cre n =
5 and Cre+ n = 5 for 11 months (ns, P > 0.05; *P <
0.05; **P < 0.01; ***P < 0.001). (C) Percentage
dierence in MAOB- PET (SUVs) between Cre
and Cre+ mice at 11 months of age. (D) Quanti-
cation of cellular MAOB- PET tracer uptake in
astrocyte- enriched and astrocyte- depleted frac-
tions of 10- month- old mice by scRadiotracing.
A one- way ANOVA with Tukey’s multiple com-
parison test was performed with n = 4 for Cre
and Cre+ samples (ns, P > 0.05; **P < 0.01).
(E) Western blot showing the expression of GFAP
in Cre (n = 3) and Cre+ (n = 3) mouse brain
tissue harvested at 2, 7, and 11 months of
age. GAPDH (glyceraldehyde- 3- phosphate de-
hydrogenase) was used as a loading control.
(F) Fold change of GFAP quantied by densitom-
etry (MultiGauge). Values were normalized to
Cre and are depicted as means ± SEM. A two-
tailed unpaired Student’s t test was performed
for each age (ns, P > 0.05; *P < 0.05; **P < 0.01).
(G) Immunohistochemical analysis of GFAP in
the cortices, midbrains, and brainstems of 2- ,
7- , and 12- month- old Cre and Cre+ mice. Scale
bars, 100 μm. (H) Quantication of GFAP cover-
age in the cortices, midbrains, and brainstems
of Cre (n = 3) and Cre+ (n = 3) mice at 2, 7, and
12 months of age. Data are presented as means
± SEM, and a two- way ANOVA with Šidák’s mul-
tiple comparisons test was performed (ns, P >
0.05; **P < 0.01; ***P < 0.001).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
7 of 14
Human iPSC- derived NPC1- decient microglia display
increased phagocytic function but are not neurotoxic
We have shown that loss of NPC1 in myeloid cells is sucient to
cause neuronal pathology in the brain, but whether microglia can
directly trigger neurodegeneration remains unclear. erefore, us-
ing CRISPR- Cas9, we generated a human- induced pluripotent stem
cell (hiPSC) line carrying the most common patient NPC1 mutation
I1061T (g. S9A) and established cocultures of microglia carrying
the NPC1 mutation (iMG NPC) and hiPSC- derived wild- type neu-
rons (iN WT). Successful generation of iN [70 days in vitro (DIV)]
and iMG (28 DIV) was monitored by the reduction in pluripotency
(g. S9B) and induction of neuronal (g. S9C) and microglial markers
(g. S9D), respectively. iMG NPC (28 DIV) showed no dierence in
NPC1 mRNA compared to iMG WT (g. S9E), but substantial re-
duction in NPC1 protein (g. S9, F and G) was reported for this
mutation (54). As shown for Cre+ microglia, iMG NPC (16 DIV)
displayed increased phagocytic uptake of myelin (g. S9, H and I).
To test whether iMG NPC are neurotoxic, we rst dierentiated
iMG and iN in monoculture for 16 and 60 DIV, respectively, and
then cocultured either iMG NPC or iMG WT with iN WT for an
additional 8 days and monitored their survival by quantifying the
neuronal marker MAP2. No obvious dierences were observed in
MAP2 coverage, number of MAP2- positive cells, or MAP2 protein
in cocultures of iN WT and iMG NPC compared to iMG WT (g. S9,
J to M). is result implies that more complex neuronal- glial inter-
actions and long- term microglial deficiency in vivo may be re-
sponsible for neuronal defects rather than a direct microglia- driven
neurotoxicity.
Emerging clinical tools for NPC disease monitoring
As a proof of concept that microglial activation is of relevance for
human NPC pathology, we performed TSPO- PET imaging (37) in
0
40
80
120
160
200
240
280
0
10
20
30
40
50
60
Radius (µm)
#Intersections
Shollintersections
Cre
-
Cre+
Cre
-
Cre
+
0
50
100
150
200
250
Retinal thickness (µm)
ns
Cre
-
Cre+
0
2000
4000
6000
8000
10,000
Area under the curve
ns
GCL INLINL
Calretinin
+
PKC
+
Cre
-
Hoechst calbindinCalbindin
AC
12 months
Cre+
BDE
FGH
Retinal section (12 months)
Calretinin PKC
GCL
INL
ONL
IPL
OPL
Calretinin
Cre
-
PKCDAPI Calretinin PKC
GCL
INL
ONL
IPL
OPL
Cre+
Calretinin
PKC
DAPI
ILM
OLM
ILM
OLM
0
10
20
30
Cellcountper scanfield
ns ns
Cre
-
Cre+
0
2000
4000
6000
8000
10,000
Total dendritelength(µm)
ns
Cre
-
Cre+
Fig. 4. NPC1- decient microglia drive region- specic neurodegeneration. (A) Immunohistochemical analysis of Purkinje cells (calbindin, green) in 12- month- old Cre
and Cre+ mice. Hoechst was used as the nuclear stain (cyan). Scale bars, 500 μm. (B to E) Morphological characterization of Purkinje cells in 7- month- old Cre and Cre+
mice. Three- dimensional reconstructions of single Purkinje cells from Cre and Cre+ mice and after biocytin- backll and subsequent labeling with streptavidin–Alexa
Fluor 647 (red insets). Scale bars, 30 μm (B). Analysis of the total length of the dendritic tree of Purkinje cells from Cre (n = 4 cells from two mice) and Cre+ (n = 4 cells from
one mouse) animals. Data are presented as means ± SEM, and a two- tailed unpaired Student’s t test was performed (ns, P > 0.05) (C). Sholl analysis depicting the mean
number of intersections along the distance to the soma (D). Analysis of the area under the curve obtained from (D). Data are presented as means ± SEM, and a two- tailed
unpaired Student’s t test was performed (ns, P > 0.05) (E). (F to H) Analysis of retinal morphology in 12- month- old Cre and Cre+ mice. Retinal sections were stained for
calretinin (magenta) and PKCα (green). DAPI was used as the nuclear stain (cyan). Grayscale images represent single channels for calretinin and PKCα. The dashed line
represents the borders of the ILM and OLM. Scale bars, 20 μm (F). The retinal thickness from ILM to OLM (G) or the number of calretinin- or PKCα- positive cells (H) was
quantied. Data are presented as means ± SEM, and simple nonparametric, unpaired Mann- Whitney t test was performed with the following sample sizes: n = 5 for Cre
samples, and n = 7 for Cre+ samples (ns, P > 0.05; **P < 0.01). ILM, internal limiting membrane; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer;
OPL, outer plexiform layer; ONL, outer nuclear layer; OLM, outer limiting membrane.
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
8 of 14
7 months
Midbrain
A
1.7
FDG-PET (SUV)
Cre
+
Cre
-
shtnom 2
7 months
11 months
0.4
B
Cortex Midbrain Brainstem
C
Cre
-
Cre+
NF200
Cortex Brainstem
F
Cerebellum
DE
GH
Cortex
p62 NeuN p62 NeuN
Cre
-
Cre+
7 months
Midbrain Brainstem
p62 NeuN p62 NeuN p62 NeuN p62 NeuN
0
500
1000
1500
2000
NF-L in serum(pg/ml)
Cre
-
Cre+
ns
ns
2months7months 12 months
Cortex Midbrain Brainstem
0
10
20
30
40
NF200aggregates perimage
Cre
-
Cre
+
ns
ns
Cre
-
Cre+
0
50
100
150
200
250
Latency tofall(s)
0.0
0.4
0.8
1.2
1.6
2.0
2.4
Age (months)
FDG-PET (SUV)
G * A
T = 2.362
P = 0.02
2711
ns
Age (months)
G * A
T = 2.522
P = 0.02
2711
ns ns
Age (months)
G * A
T = 2.250
P = 0.03
2711
ns ns
Age (months)
G * A
T = 1.674
ns
2711
ns
ns
Cortex Midbrain Brainstem
0
20
40
60
80
100
p62/NeuN-positivecells (%)
Cre
-
Cre+
ns
0.0
0.4
0.8
1.2
1.6
2.0
2.4
0.0
0.4
0.8
1.2
1.6
2.0
2.4
0.0
0.4
0.8
1.2
1.6
2.0
2.4
Fig. 5. NPC1- decient microglia drive neuronal dysfunction. (A) FDG- PET images of Cre and Cre+ mouse brains imaged at 2, 7, and 11 months of age. The shown
sagittal brain sections display group average images of SUVs projected on a standard MRI T1w atlas. (B) Quantication of individual FDG- PET SUVs derived from the cortex,
midbrain, brainstem, and cerebellum. Dotted lines represent linear associations between age and FDG- PET quantication with their 95% condence interval (lled area).
The G * A interaction was calculated. A multiple linear regression was performed for statistical analysis with the following sample sizes: Cre n = 13, Cre+ n = 5 for 2
months, Cre n = 13, Cre+ n = 7 for 7 months, and Cre n = 8, Cre+ n = 5 for 11 months (ns, P > 0.05; *P < 0.05). (C) NF- L enzyme- linked immunosorbent assay (ELISA) of
blood sera from 2- , 7- , and 12- month- old Cre (n = 4) and Cre+ (n = 4) mice. Data are presented as means ± SEM, and a two- way ANOVA with Bonferroni’s multiple com-
parison correction was performed (ns, P > 0.05; **P < 0.01; ***P < 0.001). (D) Immunohistochemical analysis using an antibody against NF200 (white) at 7 months of age.
NF200 aggregates in Cre+ mice are marked with magenta arrows. Scale bars, 50 μm. (E) Quantication of NF200 aggregates in Cre (n = 3) and Cre+ (n = 3) mice in the
cortices, midbrains, and brainstems at 7 months of age. Data are presented as means ± SEM, and a two- way ANOVA with Tukey’s multiple comparisons test was performed
(ns, P > 0.05; **P < 0.01; ***P < 0.001). (F) Immunohistochemical analysis using an antibody against p62 (green) and the pan–neuronal marker NeuN (magenta) in Cre
and Cre+ mice at 7 months of age. Scale bars, 50 μm. (G) Quantication of p62/NeuN- positive cells relative to the total number of NeuN- positive cells in the cortices,
midbrains, and brainstems at 7 months of age. Data are presented as means ± SEM, and a two- way ANOVA with Tukey’s multiple comparisons test was performed (ns,
P > 0.05; *P < 0.05; **P < 0.01; ***P < 0.001). (H) Motor function and coordination of 7- month- old Cre (n = 20) and Cre+ (n = 17) mice were tested using the rotarod. Data
are presented as means ± SEM, and a two- tailed unpaired Student’s t test was performed (***P < 0.001).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
9 of 14
ve patients with NPC. We detected a profound up- regulation of the
TSPO- PET signal across dierent brain regions with pronounced
signal elevations in the insula, brainstem, and cerebellum (Fig. 6, A
and B). Moreover, these regional TSPO- PET signals were higher in
patients with NPC compared with patients with Alzheimer’s disease
(AD) or 4- repeat tauopathies, supporting its utility as a clinical tool
for NPC (Fig. 6B).
Next, we explored the translational potential of peripheral myeloid
cells for NPC disease monitoring. We showed that rodent Npc1/
bone marrow–derived macrophages recapitulate major hallmarks of
NPC1- decient microglia, including cholesterol accumulation, up-
regulation of intraluminal vesicle (ILV)–resident CD63, increased
phagocytic uptake, lipid tracking defects, and alterations in choles-
terol metabolism (g. S10, A to H, and table S5). is is in line with
our previous characterization of blood- derived macrophages in pa-
tients with NPC that also recapitulated functional hallmarks of NPC
(16). us, blood- derived macrophages may provide a potential clini-
cal tool for therapeutic monitoring. Preclinical studies suggested that
NALL treatment of Npc1/ mice ameliorates disease progression and
is associated with reduced microglial activation (35). NALL treatment
showed benecial eects in an NPC clinical trial (24), which prompted
us to test whether blood- derived macrophages of patients with NPC
may be altered upon NALL treatment. To this end, we performed an
ex vivo assay for Aβ plaque clearance—a surrogate marker to measure
increased phagocytosis in NPC (16). A 6- week treatment of patients
with NALL displayed a trend toward reduced phagocytic hyperactiv-
ity in all three longitudinally analyzed patients (Fig. 6C). Subsequently,
we compared the proteomic signatures of patient macrophages before
and aer a 6- week treatment with NALL (table S6). We measured a
35% reduction in TSPO in blood macrophages aer treatment of pa-
tients with NALL (Fig. 6D), pointing to a benecial eect of the drug
on ameliorating hyperactivation of myeloid cells. Collectively, these
data underscore the importance of brain microglia and peripheral
myeloid cells in the course of NPC disease and document a potential
for TSPO as a monitoring biomarker in clinical trials.
Next, we explored the potential of blood- based markers that
show promise in other neurodegenerative diseases, namely, the neu-
ronal injury markers NF- L and ubiquitin C- terminal hydrolase L1
(UCH- L1) and the astrocytic marker GFAP (55,56). Plasma concen-
trations of NF- L, UCH- L1, and GFAP were signicantly (P < 0.05)
higher in patients with NPC compared with their age- matched controls
(Fig. 6E), favoring their potential as tools for disease monitoring.
In summary, we envisage the utility of markers capable of capturing
microglial (TSPO), astrocytic (GFAP), and neuronal (NF- L and UCH-
L1) pathology as a promising avenue to explore in NPC. It remains to
be tested whether concentrations of NF- L, UCH- L1, and GFAP may be
sensitive to treatment, as we could show for TSPO, underscoring their
potential for disease monitoring in future clinical trials.
DISCUSSION
Microglia are strongly engaged in NPC pathology, and their altera-
tions occur before neurodegeneration, suggesting that they may
play an active role during disease progression (16,31, 32). We ex-
plored a myeloid NPC1 depletion mouse model as a tool to study
the consequences of microglial hyperactivity on brain homeostasis
and delineate the contribution of microglia to NPC pathology. Loss
of NPC1 in myeloid cells was sucient to trigger astrogliosis, neu-
ronal pathology, and region- specic neurodegeneration, supporting
neuronal- glial cross- talk in NPC (36). No neurotoxicity could be de-
tected aer 8 days of coculturing iPSC- derived iN WT and iMG
NPC, suggesting that long- term microglial deciency in vivo may be
responsible for neuronal defects. Furthermore, our ndings from the
mouse model translated well to human patients with NPC as illus-
trated by the increased TSPO and accumulation of the neuroaxonal
injury biomarker NF- L and astrocytic marker GFAP in the blood.
Compared with Npc1/ mice with a fast- progressing neurode-
generation and early lethality, pathological changes upon loss of
NPC1 in myeloid cells occur slowly, and mice survive for almost a
year. We show that hallmarks of the NPC pathology, including axonal
swelling, NF- L increase, microgliosis, astrogliosis, and motor de-
fects, occur upon loss of NPC1 in myeloid cells. However, loss of
NPC1 in myeloid cells was not sucient to trigger Purkinje cell de-
generation, reduced myelination, or early lethality. us, our study
is in line with described cell- autonomous roles of NPC1 in other cell
types that together contribute to the wide range of clinical NPC phe-
notypes (36).
NPC is described as a lysosomal storage disorder in which,
among others, unesteried low- density lipoprotein–derived choles-
terol accumulates in lamellar inclusions of LE/lysosomes (1,57). We
showed that loss of NPC1 causes a lipid tracking dysfunction and
that myelin accumulation predominates in LE/MVBs, suggesting that
the delivery of the endolysosomal cargo rather than the catalytic
capacity of lysosomes is impaired in NPC (16). e lipidomic and pro-
teomic data align with this hypothesis because NPC1- decient microg-
lia strongly accumulate BMPs and CD63, which are predominately
found in ILVs of MVBs (58), favoring MVBs as the organellar
hotspot of NPC pathology in microglia. Increased di- 22:6- BMP was
detected in urine from patients with NPC (59). Apart from defects
in BMP homeostasis, cholesterol does not reach the endoplasmic
reticulum (ER) in NPC disease and is therefore not processed into
CEs, which is in line with the reduction in lipid droplet formation
(16). is is fully supported by the reduced CEs in NPC1- decient
microglia and defects in cholesterol esterication reported in pa-
tients with NPC (60). Besides the role of NPC1 as a cholesterol trans-
porter (61), it is proposed that NPC1 also steers the cholesterol
transport from LE/MVBs to the ER by regulating membrane contact
sites (62), and defects in membrane contact sites may contribute to
broad lipid dyshomeostasis in NPC. We detected GM2 accumula-
tion in the same intracellular compartment as cholesterol in cultured
Cre+ microglia, supporting that tracking of other lipids may be im-
paired, leading to their accumulation in LE/MVBs and additional
pathologic burden (63,64). Furthermore, severe lipid accumulation
in NPC microglia may be facilitated by increased phagocytic activity
toward extracellular substrates such as cell debris and extracellular
lipids. We demonstrated that loss of NPC1 in microglia results in
increased phagocytosis in vivo as reected by the detection of
phagocytic cups in Cre+ mice, resembling defects reported during
development in NPC (65). Phagocytic cups are typically found dur-
ing development (66), which is a period of active microglia- mediated
synaptic pruning (67). In line with these ndings, we have reported
increased synaptic materials in microglia of Npc1/ and Cre+ mice
as well as increased key regulators of synaptic pruning such as C3
and C1q (16). It has been proposed that excessive synaptic pruning
may be responsible for triggering schizophrenia- like symptoms (68),
which occur in patients with NPC but are more common in late- onset
disease. us, the microglial contribution may be even more promi-
nent in patients who display a late- onset NPC disease and present
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
10 of 14
with psychiatric symptoms (9). It has been shown that C1q is not a
major cause for neurodegeneration in NPC (69), arguing that en-
hanced synaptic pruning of NPC microglia is not directly responsible
for neurodegeneration. Because phagocytic cups were more promi-
nent in Cre+ mice at late pathological stages, microglia may be rather
engaged in removing already dysfunctional synapses or neurons. Con-
sidering the severe defects in lipid processing and phagocytic hyper-
activity of NPC1- depleted microglia, it is clear that microglia are
exposed to a pathological feed- forward loop by continuously accu-
mulating substrates from their environment.
Noninvasive approaches like PET imaging are emerging as im-
portant clinical tools in monitoring neurodegenerative processes
in vivo but oen lack information regarding cellular identity. Using
scRadiotracing (47), we showed that even under nonpathological
conditions, the TSPO tracer is mainly taken up by microglia, con-
rming its cell specicity. Furthermore, the TSPO- PET signal is
aberrantly increased in NPC1- depleted microglia, and this is not
only a result of microgliosis as recently suggested in human cells
(70). Increased TSPO- PET signals in Cre+ mice was in line with the
immunohistochemical analysis of CD68. However, in contrast with
TSPO, CD68 immunohistochemistry detected microglial hyperac-
tivation already at the age of 2 months, stressing that LE/lysosomal al-
terations are among the rst pathological changes in microglia of Cre+
mice. Of interest are also regional dierences in microglial activation,
with the strongest microglial hyperactivity detected in the mid-
brains and brainstems of Cre+ mice. We hypothesize that early and
broad microglial activation is triggered by cell- autonomous dys-
function of NPC1 that is further potentiated by pathological insults
NF-L
0
5
10
15
20
25
Concentration (pg/ml)
NPCAD4RT HC
0
1
2
3
4
TSPO-PET (SUVr)
NPCAD4RT HC
0
1
2
3
TSPO-PET (SUVr)
C
5
NPC #1
NPC #2
NPC #3
NPC #1
NPC #2
NPC #3
D
A
40 years
58 years
36 years27 years 34 years
Insula Brainstem Cerebellum
B
NPCAD4RT HC
0
1
2
3
TSPO-PET (SUVr)
E
GFAP
0
50
100
150
200 CTRL
NPC
UCH-L1
0
50
100
150
200
Ba
seline
NALL
0
20
40
60
80
100
Phagocytosed plaques (%)
ns
Basel
ine
NALL
-
1.0
-
0.8
-
0.6
-
0.4
-
0.2
0.0
Log2 LFQNALL/baseline (TSPO)
TEP-OPST
SUVr z score vs. HC
5.0
1.5
Fig. 6. Potential markers for therapeutic monitoring in NPC. (A) Axial and sagittal sections showing the z- scores of global mean scaled SUVs of TSPO- PET in patients
with NPC (n = 5) compared with age- matched (to the oldest patient with NPC) healthy controls (HC, n = 27). (B) Quantitative comparison of global mean scaled TSPO- PET
signals in the insulae, brainstems, and cerebella of patients with NPC (NPC, n = 5), Alzheimer’s disease (AD, n = 12), 4- repeat tauopathy (4RT, n = 12), and a subset of old
HC (n = 12). One- way ANOVA with Tukey post hoc test was performed (*P < 0.05; **P < 0.01; ***P < 0.001). (C) Ex vivo Aβ phagocytic assay of blood- derived macrophages
from patients with NPC (NPC #1 to 3) before (baseline) and after NALL treatment (NALL). Values are presented as means ± SEM. A two- tailed paired Student’s t test was
performed (ns, P > 0.05). (D) Analysis of TSPO in macrophages from patients with NPC treated with NALL. Macrophages from patients with NPC (NPC1 #1 to 3) were ana-
lyzed before (baseline) and after a 6- week NALL treatment via LC- MS/MS. Label- free quantication (LFQ) intensities were normalized on the individual baseline LFQ inten-
sity. Values are plotted in log2 scale. Data are presented as means ± SEM, and a one- tailed Student’s t test was performed (*P < 0.05). (E) Plasma concentrations of NF- L,
UCH- L1, and GFAP in plasma samples from patients with NPC (n = 8) and age- matched controls (CTRL, n = 8) were measured by ELISA. Data are presented as means ±
SEM, and a two- tailed unpaired Student’s t test was performed (*P < 0.05).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
11 of 14
in astrocytes or neurons, contributing to region- specic microglial
pathology at later stages. e regional overlap of TSPO- PET and
MAOB- PET signals supports pathological cross- talk between mi-
croglia and astrocytes. A similar pathological cross- talk of brain
cells is described in microglial regulator colony stimulating factor
1R (CSF1R)–related leukoencephalopathy, a disease in which mi-
croglia are primarily aected and cause prominent axonal spheroid
pathology (71). Modeling Csf1r deciency in mice results in axonal
spheroids, astrogliosis, and region- specic neurodegeneration, with
the thalamus being a pathological hotspot (72,73). However, further
mechanistic studies are warranted to understand how global mi-
croglial dysfunction may provoke selective regional vulnerabilities.
Of translational relevance is also a reduction in the FDG- PET sig-
nal in Cre+ mice that is a phenotype occurring already in patients car-
rying a heterozygous NPC1 mutation (74), supporting early neuronal
dysfunction that precedes neurodegeneration. Similar to our results,
activated microglia were capable of inducing non–cell- autonomous
inhibition of neuronal autophagy (75), underscoring the pathological
role of microglia- to- neuron cross- talk in neurodegeneration.
We show that microglial hyperactivity in NPC is reected by a
massive increase in TSPO- PET signals and increased microglial
phagocytic function. We furthermore found a lowering of phago-
cytic activity and TSPO in blood- derived macrophages from patients
with NPC aer NALL treatment, a drug that emerges as a benecial
therapeutic for NPC (24). ese new data underscore the potential of
monitoring phagocytic alterations as a response to therapy and pro-
vides a functional correlate for microglial hyperactivation measured
by the increased TSPO expression. is work suggests the translational
potential for TSPO as a monitoring biomarker in NPC and suggests
that hyperactive microglia lost their supportive functions, leading to
the observed astrogliosis, neuronal dysfunction, and, eventually, neu-
rodegeneration. Furthermore, as now witnessed in other neurodegen-
erative diseases such as AD (76) or Parkinsons disease (77), biological
disease classication develops toward a valuable tool in clinical routine.
Biomarkers capable of capturing microglial (TSPO), astrocytic (GFAP),
and neuronal (NF- L and UCH- L1) (MAN) pathology could be
combined and tested for their utility for disease monitoring in NPC
clinical trials.
One limitation of our study is that we cannot exclude the contribu-
tion of NPC1 loss in Cx3cr1- expressing peripheral myeloid cells to dis-
ease phenotypes in mice. An additional limitation is the low number of
NALL- treated patients. Aqneursa (NALL) has just been approved by
the US Food and Drug Administration for NPC (https://www.fda.gov/
news- events/press announcements/fda-approves-new-drug-treat-
niemann-pick-disease-type-c), which is a major clinical advancement
enabling future studies to link benecial clinical outcomes with bio-
markers capturing reduced MAN pathology in NPC.
MATERIALS AND METHODS
Study design
e aim of this study was to delineate the role of NPC1 in myeloid
cells and the contribution of neuroinammation to neurode generation.
Our study includes characterization of mice with myeloid cell–
specic NPC1 depletion, using primary cell culture, PET imaging,
and immunohistological, proteomic, and lipidomic analyses comple-
mented with analysis of patients with NPC and hiPSC- derived
models. For animal experiments, sample sizes were determined a
priori by applying G*power soware (V3.1.9.7) and on the basis of
our previous experimental data in rodent models of neurodegenera-
tion and neuroinammation. e animals were assigned to experi-
mental groups randomly. e P value for a priori calculation had
been set to 0.05 and power to 0.8. Analysis of all experiments was
performed blinded and with a randomized order. All animal experi-
ments complied with the ARRIVE guidelines and the German ani-
mal welfare law and were approved by the government of Upper
Bavaria (license numbers ROB- 55.2- 2532.Vet_02- 17- 075 and ROB-
55.2- 2532.Vet_02- 22- 125). All experiments using human material
were authorized by the local Ethics Committee of the University of
Munich (ethics- application: 20- 350) and University of Leipzig (ethics-
application: 370- 21- ek) and executed according to the 1964 Decla-
ration of Helsinki. PET acquisition (ethics- applications: 17- 569,
17- 755) was approved by the local institutional ethics committee
(LMU Munich) and the German radiation protection authorities
(BfS- application: Z 5- 22464/2017- 047- K- G). All participants pro-
vided written informed consent before the PET scans. Consider-
ing that NPC is a rare disease, all human patient data that we were
able to collect during the duration of the study were included. No
samples were excluded from the study. e n numbers of indepen-
dently performed experiments are reported in the gure legends.
Detailed Materials and Methods are outlined in the Supplementa-
ry Materials.
Statistical analysis
Measured values from LC- MS lipidomic analysis were log2 trans-
formed. Dierences between Npc1+/+ or Cre and Npc1/ or Cre+
in selected lipids were estimated using a robust linear model adjust-
ing for covariates of sex, age, and the interaction between age and
genotype in the model (78). e P values were adjusted for multiple
comparisons using the Benjamin- Hochberg procedure (adjusted
P value). Statistical analysis of the LC- MS proteomic data was per-
formed as described in the respective section. For each experiment,
the used sample size and statistical test are denoted in the gure
legends. A P value of P < 0.05 was considered as signicant [not
signicant (ns) = P > 0.05; *P < 0.05; **P < 0.01; ***P < 0.001].
Supplementary Materials
The PDF le includes:
Materials and Methods
Figs. S1 to S10
References (80101)
Other Supplementary Material for this manuscript includes the following:
Data les S1 and S2
MDAR Reproducibility checklist
REFERENCES AND NOTES
1. M. T. Vanier, Niemann- Pick disease type C. Orphanet J. Rare Dis. 5, 16 (2010).
2. M. T. Vanier, G. Millat, Niemann- Pick disease type C. Clin. Genet. 64, 269–281 (2003).
3. D. E. Sleat, J. A. Wiseman, M. El- Banna, S. M. Price, L. Verot, M. M. Shen, G. S. Tint,
M. T. Vanier, S. U. Walkley, P. Lobel, Genetic evidence for nonredundant functional
cooperativity between NPC1 and NPC2 in lipid transport. Proc. Natl. Acad. Sci. U.S.A. 101,
5886–5891 (2004).
4. X. Li, P. Saha, J. Li, G. Blobel, S. R. Pfeer, Clues to the mechanism of cholesterol transfer
from the structure of NPC1 middle lumenal domain bound to NPC2. Proc. Natl. Acad. Sci.
U.S.A. 113, 10079–10084 (2016).
5. E. Lloyd- Evans, F. M. Platt, Lipids on trial: The search for the oending metabolite in
Niemann- Pick type C disease. Trac 11, 419–428 (2010).
6. M. T. Vanier, Lipid changes in Niemann- Pick disease type C brain: Personal experience
and review of the literature. Neurochem. Res. 24, 481–489 (1999).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
12 of 14
7. D. te Vruchte, E. Lloyd- Evans, R. J. Veldman, D. C. A. Neville, R. A. Dwek, F. M. Platt,
W. J. van Blitterswijk, D. J. Sillence, Accumulation of glycosphingolipids in Niemann-
Pick C disease disrupts endosomal transport. J. Biol. Chem. 279, 26167–26175
(2004).
8. S. Boenzi, G. Catesini, E. Sacchetti, F. Tagliaferri, C. Dionisi- Vici, F. Deodato,
Comprehensive- targeted lipidomic analysis in Niemann- Pick C disease. Mol. Genet.
Metab. 134, 337–343 (2021).
9. O. Bonnot, C. S. Gama, E. Mengel, M. Pineda, M. T. Vanier, L. Watson, M. Watissee,
B. Schwierin, M. C. Patterson, Psychiatric and neurological symptoms in patients with
Niemann- Pick disease type C (NP- C): Findings from the International NPC Registry. World
J. Biol. Psychiatry 20, 310–319 (2019).
10. M. C. Patterson, P. Clayton, P. Gissen, M. Anheim, P. Bauer, O. Bonnot, A. Dardis,
C. Dionisi- Vici, H. H. Klunemann, P. Latour, C. M. Lourenco, D. S. Ory, A. Parker, M. Pocovi,
M. Strupp, M. T. Vanier, M. Walterfang, T. Marquardt, Recommendations for the detection
and diagnosis of Niemann- Pick disease type C: An update. Neurol. Clin. Pract. 7, 499–511
(2017).
11. S. U. Walkley, K. Suzuki, Consequences of NPC1 and NPC2 loss of function in mammalian
neurons. Biochim. Biophys. Acta 1685, 48–62 (2004).
12. S. M. Cologna, C. V. M. Cluzeau, N. M. Yanjanin, P. S. Blank, M. K. Dail, S. Siebel, C. L. Toth,
C. A. Wassif, A. P. Lieberman, F. D. Porter, Human and mouse neuroinammation markers
in Niemann- Pick disease, type C1. J. Inherit. Metab. Dis. 37, 83–92 (2014).
13. M. Walterfang, M. A. Di Biase, V. L. Cropley, A. M. Scott, G. O’Keefe, D. Velakoulis,
K. Pathmaraj, U. Ackermann, C. Pantelis, Imaging of neuroinammation in adult
Niemann- Pick type C disease: A cross- sectional study. Neurology 94, e1716–e1725
(2020).
14. T. Kodachi, S. Matsumoto, M. Mizuguchi, H. Osaka, N. Kanai, E. Nanba, K. Ohno,
T. Yamagata, Severe demyelination in a patient with a late infantile form of Niemann- Pick
disease type C. Neuropathology 37, 426–430 (2017).
15. B. M. Castellano, A. M. Thelen, O. Moldavski, M. Feltes, R. E. N. van der Welle,
L. Mydock- McGrane, X. Jiang, R. J. van Eijkeren, O. B. Davis, S. M. Louie, R. M. Perera,
D. F. Covey, D. K. Nomura, D. S. Ory, R. Zoncu, Lysosomal cholesterol activates mTORC1 via
an SLC38A9- Niemann- Pick C1 signaling complex. Science 355, 1306–1311 (2017).
16. A. Colombo, L. Dinkel, S. A. Muller, L. Sebastian Monasor, M. Schierer,
L. Cantuti- Castelvetri, J. Konig, L. Vidatic, T. Bremova- Er tl, A. P. Lieberman, S. Hecimovic,
M. Simons, S. F. Lichtenthaler, M. Strupp, S. A. Schneider, S. Tahirovic, Loss of NPC1
enhances phagocytic uptake and impairs lipid tracking in microglia. Nat. Commun. 12,
1158 (2021).
17. M. J. Elrick, T. Yu, C. Chung, A. P. Lieberman, Impaired proteolysis underlies autophagic
dysfunction in Niemann- Pick type C disease. Hum. Mol. Genet. 21, 4876–4887
(2012).
18. G. Liao, Y. Yao, J. Liu, Z. Yu, S. Cheung, A. Xie, X. Liang, X. Bi, Cholesterol accumulation is
associated with lysosomal dysfunction and autophagic stress in Npc1/ mouse brain.
Am. J. Pathol. 171, 962–975 (2007).
19. S. Sar kar, B. Carroll, Y. Buganim, D. Maetzel, A. H. Ng, J. P. Cassady, M. A. Cohen,
S. Chakraborty, H. Wang, E. Spooner, H. Ploegh, J. Gsponer, V. I. Korolchuk, R. Jaenisch,
Impaired autophagy in the lipid- storage disorder Niemann- Pick type C1 disease. Cell Rep.
5, 1302–1315 (2013).
20. J. Havla, M. Moser, C. Sztatecsny, A. S. Lotz- Havla, E. M. Maier, B. Hizli, R. Schinner,
T. Kumpfel, M. Strupp, T. Bremova- Er tl, S. A. Schneider, Retinal axonal degeneration in
Niemann- Pick type C disease. J. Neurol. 267, 2070–2082 (2020).
21. T. Claudepierre, M. Paques, M. Simonutti, I. Buard, J. Sahel, R. A. Maue, S. Picaud,
F. W. Pfrieger, Lack of Niemann- Pick type C1 induces age- related degeneration in the
mouse retina. Mol. Cell. Neurosci. 43, 164–176 (2010).
22. M. Pineda, M. Walterfang, M. C. Patterson, Miglustat in Niemann- Pick disease type C
patients: A review. Orphanet J. Rare Dis. 13, 140 (2018).
23. T. Bremova- Ertl, J. Claassen, T. Foltan, J. Gascon- Bayarri, P. Gissen, A. Hahn, A. Hassan,
A. Hennig, S. A. Jones, M. Kolnikova, K. Martakis, J. Raethjen, U. Ramaswami, R. Sharma,
S. A. Schneider, Ecacy and safety of N- acetyl- L- leucine in Niemann- Pick disease type C.
J. Neurol. 269, 1651–1662 (2022).
24. T. Bremova- Ertl, U. Ramaswami, M. Brands, T. Foltan, M. Gautschi, P. Gissen, F. Gowing,
A. Hahn, S. Jones, R. Kay, M. Kolnikova, L. Arash- Kaps, T. Marquardt, E. Mengel, J. H. Park,
S. Reichmannova, S. A. Schneider, S. Sivananthan, M. Walterfang, P. Wibawa, M. Strupp,
K. Martakis, Trial of N- acetyl- l- leucine in Niemann- Pick disease type C. N. Engl. J. Med.
390, 421–431 (2024).
25. X. Jiang, D. S. Ory, Advancing diagnosis and treatment of Niemann- Pick C disease
through biomarker discovery. Explor. Neuroprotective Ther. 1, 146–158 (2021).
26. S. K . Loftus, J. A. Morris, E. D. Carstea, J. Z. Gu, C. Cummings, A. Brown, J. Ellison, K. Ohno,
M. A. Rosenfeld, D. A. Tagle, P. G. Pentchev, W. J. Pavan, Murine model of Niemann- Pick C
disease: Mutation in a cholesterol homeostasis gene. Science 277, 232–235 (1997).
27. P. G. Pentchev, A. E. Gal, A. D. Booth, F. Omodeo- Sale, J. Fouks, B. A. Neumeyer, J. M. Quirk,
G. Dawson, R. O. Brady, A lysosomal storage disorder in mice characterized by a dual
deciency of sphingomyelinase and glucocerebrosidase. Biochim. Biophys. Acta 619,
669–679 (1980).
28. Y. Higashi, S. Murayama, P. G. Pentchev, K. Suzuki, Cerebellar degeneration in the
Niemann- Pick type C mouse. Acta Neuropathol. 85, 175–184 (1993).
29. D. C. German, E. M. Quintero, C. L. Liang, B. Ng, S. Punia, C. Xie, J. M. Dietschy, Selective
neurodegeneration, without neurobrillary tangles, in a mouse model of Niemann- Pick
C disease. J. Comp. Neurol. 433, 415–425 (2001).
30. S. Ohara, Y. Ukita, H. Ninomiya, K. Ohno, Degeneration of cholecystokinin-
immunoreactive aerents to the VPL thalamus in a mouse model of Niemann- Pick
disease type C. Brain Res. 1022, 244–246 (2004).
31. S. N. Pressey, D. A. Smith, A. M. S. Wong, F. M. Platt, J. D. Cooper, Early glial activation,
synaptic changes and axonal pathology in the thalamocortical system of Niemann- Pick
type C1 mice. Neurobiol. Dis. 45, 1086–1100 (2012).
32. M. Baudr y, Y. Yao, D. Simmons, J. Liu, X. Bi, Postnatal development of inammation in a
murine model of Niemann- Pick type C disease: Immunohistochemical observations of
microglia and astroglia. Exp. Neurol. 184, 887–903 (2003).
33. A. Ber nardo, C. De Nuccio, S. Visentin, A. Martire, L. Minghetti, P. Popoli, A. Ferrante,
Myelin defects in Niemann- Pick type C disease: Mechanisms and possible therapeutic
perspectives. Int. J. Mol. Sci. 22, 8858 (2021).
34. X. Yan, F. Yang, J. Lukas, M. Witt, A. Wree, A. Rolfs, J. Luo, Hyperactive glial cells contribute
to axonal pathologies in the spinal cord of Npc1 mutant mice. Glia 62, 1024–1040 (2014).
35. E. K aya, D. A. Smith, C. Smith, L. Morris, T. Bremova- Ertl, M. Cor tina- Borja, P. Fineran,
K. J. Morten, J. Poulton, B. Boland, J. Spencer, M. Strupp, F. M. Platt, Acetyl- leucine slows
disease progression in lysosomal storage disorders. Brain Commun. 3, fcaa148 (2021).
36. M. M alara, M. Prestel, S. Tahirovic, Endo- lysosomal dysfunction and neuronal- glial
crosstalk in Niemann- Pick type C disease. Philos. Trans. R. Soc. Lond. B Biol. Sci. 379,
20220388 (2024).
37. L. Zhang, K. Hu, T. Shao, L. Hou, S. Zhang, W. Ye, L. Josephson, J. H. Meyer, M. R. Zhang,
N. Vasdev, J. Wang, H. Xu, L. Wang, S. H. Liang, Recent developments on PET radiotracers
for TSPO and their applications in neuroimaging. Acta Pharm. Sin. B 11, 373–393 (2021).
38. E. Nutma, K . Ceyzeriat, S. Amor, S. Tsartsalis, P. Millet, D. R. Owen, V. Papadopoulos,
B. B. Tournier, Cellular sources of TSPO expression in healthy and diseased brain. Eur. J.
Nucl. Med. Mol. Imaging 49, 146–163 (2021).
39. T. R. Guilarte, A. N. Rodichkin, J. L. McGlothan, A. M. Acanda De La Rocha, D. J. Azzam,
Imaging neuroinammation with TSPO: A new perspective on the cellular sources and
subcellular localization. Pharmacol. Ther. 234, 108048 (2022).
40. M. J. Elrick , C. D. Pacheco, T. Yu, N. Dadgar, V. G. Shakkottai, C. Ware, H. L. Paulson,
A. P. Lieberman, Conditional Niemann- Pick C mice demonstrate cell autonomous
Purkinje cell neurodegeneration. Hum. Mol. Genet. 19, 837–847 (2010).
41. S. Yona, K. W. Kim, Y. Wolf, A. Mildner, D. Varol, M. Breker, D. Strauss- Ayali, S. Viukov,
M. Guilliams, A. Misharin, D. A. Hume, H. Perlman, B. Malissen, E. Zelzer, S. Jung, Fate
mapping reveals origins and dynamics of monocytes and tissue macrophages under
homeostasis. Immunity 38, 79–91 (2013).
42. D. Smith, K. L. Wallom, I. M. Williams, M. Jeyakumar, F. M. Platt, Benecial eects of
anti- inammator y therapy in a mouse model of Niemann- Pick disease type C1.
Neurobiol. Dis. 36, 242–251 (2009).
43. M. Fan, R. Sidhu, H. Fujiwara, B. Tortelli, J. Zhang, C. Davidson, S. U. Walkley, J. H. Bagel,
C. Vite, N. M. Yanjanin, F. D. Porter, J. E. Schaer, D. S. Ory, Identication of Niemann- Pick
C1 disease biomarkers through sphingolipid proling. J. Lipid Res. 54, 2800–2814 (2013).
44. T. Kobayashi, E. Stang, K. S. Fang, P. de Moerloose, R. G. Parton, J. Gruenberg, A lipid
associated with the antiphospholipid syndrome regulates endosome structure and
function. Nature 392, 193–197 (1998).
45. T. Kobayashi, M. H. Beuchat, M. Lindsay, S. Frias, R. D. Palmiter, H. Sakuraba, R. G. Parton,
J. Gruenberg, Late endosomal membranes rich in lysobisphosphatidic acid regulate
cholesterol transport. Nat. Cell Biol. 1, 113–118 (1999).
46. J. Chevallier, Z. Chamoun, G. Jiang, G. Prestwich, N. Sakai, S. Matile, R. G. Parton,
J. Gruenberg, Lysobisphosphatidic acid controls endosomal cholesterol levels. J. Biol.
Chem. 283, 27871–27880 (2008).
47. L. M. Bar tos, S. T. Kunte, P. Beumers, X. Xiang, K. Wind, S. Ziegler, P. Bartenstein, H. Choi,
D. S. Lee, C. Haass, L. V. Baumgarten, S. Tahirovic, N. L. Albert, S. Lindner, M. Brendel,
Single- cell radiotracer allocation via immunomagnetic sorting to disentangle PET signals
at cellular resolution. J. Nucl. Med. 63, 1459–1462 (2022).
48. M. Linnerbauer, M. A. Wheeler, F. J. Quintana, Astrocyte crosstalk in CNS inammation.
Neuron 108, 608–622 (2020).
49. A. Ballweg, C. Klaus, L. Vogler, S. Katzdobler, K. Wind, A. Zatcepin, S. I. Ziegler, B. Secgin,
F. Eckenweber, B. Bohr, A. Bernhardt, U. Fietzek, B. S. Rauchmann, S. Stoecklein, S. Quach,
L. Beyer, M. Scheifele, M. Simmet, E. Joseph, S. Lindner, I. Berg, N. Koglin, A. Mueller,
A. W. Stephens, P. Bartenstein, J. C. Tonn, N. L. Albert, T. Kumpfel, M. Kerschensteiner,
R. Perneczky, J. Levin, L. Paeger, J. Herms, M. Brendel, [(18)F]F- DED PET imaging of
reactive astrogliosis in neurodegenerative diseases: Preclinical proof of concept and
rst- in- human data. J. Neuroinammation 20, 68 (2023).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
13 of 14
50. A. M. Jurga, M. Paleczna, J. Kadluczka, K. Z. Kuter, Beyond the GFAP- astrocyte protein
markers in the brain. Biomolecules 11, 1361 (2021).
51. C. Bouter, P. Henniges, T. N. Franke, C. Irwin, C. O. Sahlmann, M. E. Sichler, N. Beindor,
T. A. Bayer, Y. Bouter, 18F- FDG- PET detects drastic changes in brain metabolism in the
Tg4- 42 model of Alzheimer’s disease. Front. Aging Neurosci. 10, 425 (2019).
52. A. Mullard, NfL makes regulatory debut as neurodegenerative disease biomarker. Nat.
Rev. Drug Discov. 22, 431–434 (2023).
53. F. Bavato, C. Barro, L. K. Schnider, J. Simren, H. Zetterberg, E. Seifritz, B. B. Quednow,
Introducing neurolament light chain measure in psychiatry: Current evidence,
opportunities, and pitfalls. Mol. Psychiatry 29, 2543–2559 (2024).
54. M. E. Gelsthorpe, N. Baumann, E. Millard, S. E. Gale, S. J. Langmade, J. E. Schaer, D. S. Ory,
Niemann- Pick type C1 I1061T mutant encodes a functional protein that is selected for
endoplasmic reticulum- associated degradation due to protein misfolding. J. Biol. Chem.
283, 8229–8236 (2008).
55. A. Abdelhak , C. Cordano, W. J. Boscardin, E. Caverzasi, J. Kuhle, B. Chan, J. M. Gelfand,
H. H. Yiu, F. C. Oertel, A. Beaudry- Richard, S. Condor Montes, J. R. Oksenberg,
A. Lario Lago, A. Boxer, J. C. Rojas- Martinez, F. M. Elahi, J. R. Chan, A. J. Green, Plasma
neurolament light chain levels suggest neuroaxonal stability following therapeutic
remyelination in people with multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 93,
972–977 (2022).
56. A. Abdelhak , M. Foschi, S. Abu- Rumeileh, J. K. Yue, L. D’Anna, A. Huss, P. Oeckl,
A. C. Ludolph, J. Kuhle, A. Petzold, G. T. Manley, A. J. Green, M. Otto, H. Tumani, Blood
GFAP as an emerging biomarker in brain and spinal cord disorders. Nat. Rev. Neurol. 18,
158–172 (2022).
57. V. Demais, A. Barthelemy, M. Perraut, N. Ungerer, C. Keime, S. Reibel, F. W. Pfrieger,
Reversal of pathologic lipid accumulation in npc1- decient neurons by drug- promoted
release of LAMP1- coated lamellar inclusions. J. Neurosci. 36, 8012–8025 (2016).
58. J. Gruenberg, Life in the lumen: The multivesicular endosome. Tra c 21, 76–93 (2020).
59. N. Liu, E. A. Tengstrand, L. Chourb, F. Y. Hsieh, Di- 22:6- bis(monoacylglycerol)phosphate:
A clinical biomarker of drug- induced phospholipidosis for drug development and safety
assessment. Toxicol. Appl. Pharmacol. 279, 467–476 (2014).
60. E. Mengel, B. Bembi, M. Del Toro, F. Deodato, M. Gautschi, S. Grunewald, S. Gronborg,
B. Heron, E. M. Maier, A. Roubertie, S. Santra, A. Tylki- Sz ymanska, S. Day, T. Symonds,
S. Hudgens, M. C. Patterson, C. Guldberg, L. Ingemann, N. H. T. Petersen, T. Kirkegaard,
C. Í. Dali, Clinical disease progression and biomarkers in Niemann- Pick disease type C:
A prospective cohort study. Orphanet J. Rare Dis. 15, 328 (2020).
61. S. R. Pfeer, NPC intracellular cholesterol transporter 1 (NPC1)- mediated cholesterol
export from lysosomes. J. Biol. Chem. 294, 1706–1709 (2019).
62. D. Hoglinger, T. Burgoyne, E. Sanchez- Heras, P. Hartwig, A. Colaco, J. Newton, C. E. Futter,
S. Spiegel, F. M. Platt, E. R. Eden, NPC1 regulates ER contacts with endocytic organelles to
mediate cholesterol egress. Nat. Commun. 10, 4276 (2019).
63. T. I. Chen, P. C. Hsu, N. C. Lee, Y. H. Liu, H. C. Wang, Y. H. Lu, Y. H. Chien, W. L. Hwu, Loss of
Flot2 expression in deep cerebellar nuclei neurons of mice with Niemann- Pick disease
type C. Heliyon 9, e18082 (2023).
64. M. Sato, S. Akaboshi, T. Katsumoto, M. Taniguchi, K. Higaki, T. Tai, H. Sakuraba, K. Ohno,
Accumulation of cholesterol and GM2 ganglioside in cells cultured in the presence of
progesterone: An implication for the basic defect in Niemann- Pick disease type C. Brain
Dev. 20, 50–52 (1998).
65. B. R. Boyle, S. E. Melli, R. S. Altreche, Z. M. Padron, F. A. K. Yousufzai, S. Kim, M. D. Vasquez,
D. M. Carone, B. R. Carone, I. Soto, NPC1 deciency impairs cerebellar postnatal
development of microglia and climbing ber renement in a mouse model of
Niemann- Pick disease type C. Development 147, dev189019 (2020).
66. J. W. VanRyzin, A. E. Marquardt, K. J. Argue, H. A. Vecchiarelli, S. E. Ashton, S. E. Arambula,
M. N. Hill, M. M. McCarthy, Microglial phagocytosis of newborn cells is induced by
endocannabinoids and sculpts sex dierences in juvenile rat social play. Neuron 102,
435–449.e6 (2019).
67. D. P. Schafer, E. K. Lehrman, A. G. Kautzman, R. Koyama, A. R. Mardinly, R. Yamasaki,
R. M. Ransoho, M. E. Greenberg, B. A. Barres, B. Stevens, Microglia sculpt postnatal
neural circuits in an activity and complement- dependent manner. Neuron 74, 691–705
(2012).
68. P. Boksa, Abnormal synaptic pruning in schizophrenia: Urban myth or reality?
J. Psychiatry Neurosci. 37, 75–77 (2012).
69. M. E. Lopez, A. D. Klein, M. P. Scott, Complement is dispensable for neurodegeneration in
Niemann- Pick disease type C. J. Neuroinammation 9, 216 (2012).
70. E. Nutma, N. Fancy, M. Weinert, S. Tsartsalis, M. C. Marzin, R. C. J. Muirhead, I. Falk,
M. Breur, J. de Bruin, D. Hollaus, R. Pieterman, J. Anink, D. Story, S. Chandran, J. Tang,
M. C. Trolese, T. Saito, T. C. Saido, K. H. Wiltshire, P. Beltran- Lobo, A. Phillips, J. Antel,
L. Healy, M. F. Dorion, D. A. Galloway, R. Y. Benoit, Q. Amosse, K. Ceyzeriat, A. M. Badina,
E. Kovari, C. Bendotti, E. Aronica, C. I. Radulescu, J. H. Wong, A. M. Barron, A. M. Smith,
S. J. Barnes, D. W. Hampton, P. van der Valk, S. Jacobson, O. W. Howell, D. Baker, M. Kipp,
H. Kaddatz, B. B. Tournier, P. Millet, P. M. M atthews, C. S. Moore, S. Amor, D. R. Owen,
Translocator protein is a marker of activated microglia in rodent models but not human
neurodegenerative diseases. Nat. Commun. 14, 5247 (2023).
71. N. Foulds, R. J. Pengelly, S. R. Hammans, J. A. R. Nicoll, D. W. Ellison, A. Ditcheld, S. Beck,
S. Ennis, Adult- onset leukoencephalopathy with axonal spheroids and pigmented glia
caused by a novel R782G mutation in CSF1R. Sci. Rep. 5, 10042 (2015).
72. D. A. D. Munro, N. Bestard- Cuche, C. McQuaid, A. Chagnot, S. K. Shabestari,
J. P. Chadarevian, U. Maheshwari, S. Szymkowiak, K. Morris, M. Mohammad, A. Corsinotti,
B. Bradford, N. Mabbott, R. J. Lennen, M. A. Jansen, C. Pridans, B. W. McColl, A. Keller,
M. Blurton- Jones, A. Montagne, A. Williams, J. Priller, Microglia protect against
age- associated brain pathologies. Neuron 112, 2732–2748.e8 (2024).
73. J. P. Chadarevian, J. Hasselmann, A. Lahian, J. K. Capocchi, A. Escobar, T. E. Lim, L. Le, C. Tu,
J. Nguyen, S. Kiani Shabestari, W. Carlen- Jones, S. Gandhi, G. Bu, D. A. Hume, C. Pridans,
Z. K. Wszolek, R. C. Spitale, H. Davtyan, M. Blurton- Jones, Therapeutic potential of human
microglia transplantation in a chimeric model of CSF1R- related leukoencephalopathy.
Neuron 112, 2686–2707.e8 (2024).
74. T. Bremova- Ertl, C. Sztatecsny, M. Brendel, M. Moser, B. Moller, D. A. Clevert, S. Beck- Wodl,
C. Kun- Rodrigues, J. Bras, A. Rominger, D. Ninov, M. Strupp, S. A. Schneider, Clinical,
ocular motor, and imaging prole of Niemann- Pick type C heterozygosity. Neurology 94,
e1702–e1715 (2020).
75. B. P. Festa, F. H. Siddiqi, M. Jimenez- Sanchez, H. Won, M. Rob, A. Djajadikerta,
E. Stamatakou, D. C. Rubinsztein, Microglial- to- neuronal CCR5 signaling regulates
autophagy in neurodegeneration. Neuron 111, 2021–2037.e12 (2023).
76. C. R. Jack Jr., J. S. Andrews, T. G. Beach, T. Buracchio, B. Dunn, A. Graf, O. Hansson, C. Ho,
W. Jagust, E. McDade, J. L. Molinuevo, O. C. Okonkwo, L. Pani, M. S. Rai, P. Scheltens,
E. Siemers, H. M. Snyder, R. Sperling, C. E. Teunissen, M. C. Carrillo, Revised criteria for
diagnosis and staging of Alzheimer’s disease: Alzheimer’s association workgroup.
Alzheimers Dement. 20, 5143–5169 (2024).
77. G. U. Hoglinger, C. H. Adler, D. Berg, C. Klein, T. F. Outeiro, W. Poewe, R. Postuma,
A. J. Stoessl, A. E. Lang, A biological classication of Parkinson’s disease: The SynNeurGe
research diagnostic criteria. Lancet Neurol. 23, 191–204 (2024).
78. T. Logan, M. J. Simon, A. Rana, G. M. Cherf, A. Srivastava, S. S. Davis, R. L. Y. Low, C.- L. Chiu,
M. Fang, F. Huang, A. Bhalla, C. Llapashtica, R. Prorok, M. E. Pizzo, M. E. K. Calvert,
E. W. Sun, J. Hsiao- Nakamoto, Y. Rajendra, K. W. Lexa, D. B. Srivastava, B. van Lengerich,
J. Wang, Y. Robles- Colmenares, D. J. Kim, J. Duque, M. Lenser, T. K. Earr, H. Nguyen,
R. Chau, B. Tsogtbaatar, R. Ravi, L. L. Skuja, H. Solanoy, H. J. Rosen, B. F. Boeve, A. L. Boxer,
H. W. Heuer, M. S. Dennis, M. S. Kariolis, K. M. Monroe, L. Przybyla, P. E. Sanchez,
R. Meisner, D. Diaz, K. R. Henne, R. J. Watts, A. G. Henry, K. Gunasekaran, G. Astarita,
J. H. Suh, J. W. Lewcock, S. L. DeVos, G. Di Paolo, Rescue of a lysosomal storage disorder
caused by Grn loss of function with a brain penetrant progranulin biologic. Cell 184,
4651–4668.e25 (2021).
79. Y. Perez- Riverol, J. Bai, C. Bandla, D. Garcia- Seisdedos, S. Hewapathirana,
S. Kamatchinathan, D. J. Kundu, A. Prakash, A. Frericks- Zipper, M. Eisenacher, M. Walzer,
S. Wang, A. Brazma, J. A. Vizcaino, The PRIDE database resources in 2022: A hub for mass
spectrometry- based proteomics evidences. Nucleic Acids Res. 50, D543–D552 (2022).
80. C. Palleis, J. Sauerbeck, L. Beyer, S. Harris, J. Schmitt, E. Morenas- R odriguez, A. Finze,
A. Nitschmann, F. Ruch- Rubinstein, F. Eckenweber, G. Biechele, T. Blume, Y. Shi,
E. Weidinger, C. Prix, K. Botzel, A. Danek, B. S. Rauchmann, S. Stocklein, S. Lindner,
M. Unterrainer, N. L. Albert, C. Wetzel, R. Rupprecht, A. Rominger, P. Bar tenstein, J. Herms,
R. Perneczky, C. Haass, J. Levin, G. U. Hoglinger, M. Brendel, In vivo assessment of
neuroinammation in 4- repeat tauopathies. Mov. Disord. 36, 883–894 (2021).
81. N. L. Alber t, M. Unterrainer, D. F. Fleischmann, S. Lindner, F. Vettermann, A. Brunegraf,
L. Vomacka, M. Brendel, V. Wenter, C. Wetzel, R. Rupprecht, J. C. Tonn, C. Belka,
P. Bartenstein, M. Niyazi, TSPO PET for glioma imaging using the novel ligand (18)
F- GE- 180: First results in patients with glioblastoma. Eur. J. Nucl. Med. Mol. Imaging 44,
2230–2238 (2017).
82. Z. I. Kolabas, L. B. Kuemmerle, R. Perneczky, B. Forstera, S. Ulukaya, M. Ali, S. Kapoor,
L. M. Bartos, M. Buttner, O. S. Caliskan, Z. Rong, H. Mai, L. Hoher, D. Jeridi, M. Molbay,
I. Khalin, I. K. Deligiannis, M. Negwer, K. Roberts, A. Simats, O. Caroglio, M. I. Todorov,
I. Horvath, F. Ozturk, S. Hummel, G. Biechele, A. Zatcepin, M. Unterrainer, J. Gnorich,
J. Roodselaar, J. Shrouder, P. Khosravani, B. Tast, L. Richter, L. Diaz- Marugan,
D. Kaltenecker, L. Lux, Y. Chen, S. Zhao, B. S. Rauchmann, M. Sterr, I. Kunze, K. Stanic,
V. W. Y. Kan, S. Besson- Girard, S. Katzdobler, C. Palleis, J. Schadler, J. C. Paetzold,
S. Liebscher, A. E. Hauser, O. Gokce, H. Lickert, H. Steinke, C. Benakis, C. Braun,
C. P. Martinez- Jimenez, K . Buerger, N. L. Albert, G. Hoglinger, J. Levin, C. Haass,
A. Kopczak, M. Dichgans, J. Havla, T. Kumpfel, M. Kerschensteiner, M. Schierer,
M. Simons, A. Liesz, N. Krahmer, O. A. Bayraktar, N. Franzmeier, N. Plesnila, S. Erener,
V. G. Puelles, C. Delbridge, H. S. Bhatia, F. Hellal, M. Elsner, I. Bechmann, B. Ondruschka,
M. Brendel, F. J. Theis, A. Erturk, Distinct molecular proles of skull bone marrow in health
and neurological disorders. Cell 186, 3706–3725.e29 (2023).
83. A. J. Song, R. D. Palmiter, Detecting and avoiding problems when using the Cre- lox
system. Trends Genet. 34, 333–340 (2018).
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
Dinkel et al., Sci. Transl. Med. 16, eadl4616 (2024) 4 December 2024
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE
14 of 14
84. J. Rudan Njavro, M. Vukicevic, E. Fiorini, L. Dinkel, S. A. Muller, A. Berghofer, C. Bordier,
S. Kozlov, A. Halle, K. Buschmann, A. Capell, C. Giudici, M. Willem, R. Feederle,
S. F. Lichtenthaler, C. Babolin, P. Montanari, A. Pfeifer, M. Kosco- Vilbois, S. Tahirovic,
Benecial eect of ACI- 24 vaccination on Aβ plaque pathology and microglial
phenotypes in an amyloidosis mouse model. Cells 12, 79 (2023).
85. X. Xiang, G. Werner, B. Bohrmann, A. Liesz, F. Mazaheri, A. Capell, R. Feederle, I. Knuesel,
G. Kleinberger, C. Haass, TREM2 deciency reduces the ecacy of immunotherapeutic
amyloid clearance. EMBO Mol. Med. 8, 992–1004 (2016).
86. J. Cox, M. Y. Hein, C. A. Luber, I. Paron, N. Nagaraj, M. Mann, Accurate proteome- wide
label- free quantication by delayed normalization and maximal peptide ratio extraction,
termed MaxLFQ. Mol. Cell. Proteomics 13, 2513–2526 (2014).
87. S. Tyanova, T. Temu, P. Sinitcyn, A. Carlson, M. Y. Hein, T. Geiger, M. Mann, J. Cox, The
Perseus computational platform for comprehensive analysis of (prote)omics data. Nat.
Methods 13, 731–740 (2016).
88. I. Weisheit, J. A. Kroeger, R. Malik, J. Klimmt, D. Crusius, A. Dannert, M. Dichgans,
D. Paquet, Detection of deleterious on- target eects after HDR- mediated CRISPR editing.
Cell Rep. 31, 107689 (2020).
89. F. A. Ran, P. D. Hsu, J. Wright, V. Agarwala, D. A. Scott, F. Zhang, Genome engineering
using the CRISPR- Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).
90. A. Danner t, J. Klimmt, C. Cardoso Goncalves, D. Crusius, D. Paquet, Reproducible and
scalable dierentiation of highly pure cortical neurons from human induced pluripotent
stem cells. STAR Protoc. 4, 102266 (2023).
91. E. M. Abud, R. N. Ramirez, E. S. Mar tinez, L. M. Healy, C. H. H. Nguyen,
S. A. Newman, A. V. Yeromin, V. M. Scarfone, S. E. Marsh, C. Fimbres, C. A. Caraway,
G. M. Fote, A. M. Madany, A. Agrawal, R. Kayed, K. H. Gylys, M. D. Cahalan, B. J. Cummings,
J. P. Antel, A. Mortazavi, M. J. Carson, W. W. Poon, M. Blurton- Jones, iPSC- derived human
microglia- like cells to study neurological diseases. Neuron 94, 278–293.e9 (2017).
92. A. Reifschneider, S. Robinson, B. van Lengerich, J. Gnorich, T. Logan, S. Heindl, M. A. Vogt,
E. Weidinger, L. Riedl, K. Wind, A. Zatcepin, I. Pesamaa, S. Haberl, B. Nuscher,
G. Kleinberger, J. Klimmt, J. K. Gotzl, A. Liesz, K. Burger, M. Brendel, J. Levin,
J. Diehl- Schmid, J. Suh, G. Di Paolo, J. W. Lewcock, K. M. Monroe, D. Paquet, A. Capell,
C. Haass, Loss of TREM2 rescues hyperactivation of microglia, but not lysosomal decits
and neurotoxicity in models of progranulin deciency. EMBO J. 41, e109108 (2022).
93. J. Schindelin, I. Arganda- Carreras, E. Frise, V. Kaynig, M. Longair, T. Pietzsch, S. Preibisch,
C. Rueden, S. Saalfeld, B. Schmid, J. Y. Tinevez, D. J. White, V. Hartenstein, K. Eliceiri,
P. Tomancak, A. Cardona, Fiji: An open- source platform for biological- image analysis. Nat.
Methods 9, 676–682 (2012).
94. E. R. Almajan, R. Richter, L. Paeger, P. Martinelli, E. Barth, T. Decker, N. G. Larsson,
P. Kloppenburg, T. Langer, E. I. Rugarli, AFG3L2 supports mitochondrial protein synthesis
and Purkinje cell survival. J. Clin. Invest. 122, 4048–4058 (2012).
95. S. Murru, S. Hess, E. Barth, E. R. Almajan, D. Schatton, S. Hermans, S. Brodesser, T. Langer,
P. Kloppenburg, E. I. Rugarli, Astrocyte- specic deletion of the mitochondrial m- AAA
protease reveals glial contribution to neurodegeneration. Glia 67, 1526–1541 (2019).
96. C. S. Hughes, S. Moggridge, T. Muller, P. H. Sorensen, G. B. Morin, J. Krijgsveld, Single- pot,
solid- phase - enhanced sample preparation for proteomics experiments. Nat. Protoc. 14,
68–85 (2019).
97. V. Demichev, C. B. Messner, S. I. Vernardis, K. S. Lilley, M. Ralser, DIA- NN: Neural networks
and interference correction enable deep proteome coverage in high throughput. Nat.
Methods 17, 41–44 (2020).
98. V. G. Tusher, R. Tibshirani, G. Chu, Signicance analysis of microarrays applied to the
ionizing radiation response. Proc. Natl. Acad. Sci. U.S.A. 98, 5116–5121 (2001).
99. S. Didusch, M. M adern, M. Hartl, M. Baccarini, amica: An interactive and user- friendly
web- platform for the analysis of proteomics data. BMC Genomics 23, 817 (2022).
100. F. Overho, M. Brendel, A. Jaworska, V. Korzhova, A. Delker, F. Probst, C. Focke,
F.- J. Gildehaus, J. Carlsen, K. Baumann, C. Haass, P. Bartenstein, J. Herms, A. Rominger,
Automated spatial brain normalization and hindbrain white matter reference tissue give
improved [(18)F]- Florbetaben PET quantitation in Alzheimer’s model mice. Front.
Neurosci. 10, 45 (2016).
101. Y. Ma, P. R. Hof, S. C. Grant, S. J. Blackband, R. Bennett, L. Slatest, M. D. McGuigan,
H. Benveniste, A three- dimensional digital atlas database of the adult C57BL/6J mouse
brain by magnetic resonance microscopy. Neuroscience 135, 1203–1215 (2005).
Acknowledgments: We thank C. Haass for the support with the NF- L analysis and S. Wagner
for support with PET imaging. Funding: This work was supported by an Alzheimer’s
Association grant through the AD Strategic Fund to S.T. (ADSF- 21- 831226- C), the DFG (DFG
Research Unit FOR2858–project numbers 403161218: GR 4403/7- 1 and GR 4403/7- 2 to A.G.; TA
551/3- 2 to S.T.; HE 2328/14- 1 to J.H.; PA 3450/4- 1 to L.P.; DFG Priority Programme SPP2395–
project numbers 500118375, TA 551/2- 1 to S.T. and HE 2328/13- 1 to J.H.), the Centers of
Excellence in Neurodegeneration (CoEN6005), the Bundesministerium für Bildung und
Forschung (BMBF, FKZ: 01ED2402A) under the aegis of JPND and by the Cure Alzheimer’s Fund
to S.F.L., the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under
Germany’s Excellence Strategy within the framework of the Munich Cluster for Systems
Neurology (EXC 2145 SyNergy–ID 390857198) to M. Schierer, M. Simons, J.H., S.F.L., N.F., A.L.,
and M.B. S.A.S. was supported by the LMU Clinician Scientist Excellence Programme. Author
contributions: L.D., S.T., and M.B. supervised the study. L.D., M.P., M.B., and S.T. wrote the
manuscript with the input of all coauthors. L.D., V.Z., and Y.T. isolated primary cells and
performed in vitro experiments. L.D. and J.R.N. collected brain tissue and performed
immunohistochemical analysis. L.D. and A.C. performed the phagocytosis assay with human
blood samples. J.H.S., T.L., and G.D.P. performed the lipidomic analysis. S.R. and A.L. performed
the rotarod test. L.P. and J.H. performed the Purkinje cell analysis. P.H., O.B., and A.G. performed
the retina analysis. A.S., S.A.M., and S.F.L. performed the proteomic analysis. L.D. and V.Z.
performed Western blot analysis. M.P. performed IPA analysis. M. Schierer and M. Simons
performed the EM analysis. S.H. performed small- animal PET with the help of L.S., L.H., and
S.T.K. P.B., N.F., J.G., and M.B. performed human PET and human PET analyses. L.M.B., K.W.- M.,
and S.T.K. performed scRadiotracing. S.L. synthesized all radiotracers. L.D., V.Z., M.M., L.S.M.,
M.P., and D.P. established and analyzed the hiPSC lines. B.N. performed mouse and human
ELISA. T.B.- E., C.C., S.B., C.B., S.A.S., M. Strupp, and M.B. recruited patients with NPC for blood
sampling and PET. Competing interests: M. Strupp is joint chief editor of the Journal of
Neurology, editor- in- chief of Frontiers of Neuro- otology, and section editor of F1000. He has
received speaker honoraria from Abbott, Auris Medical, Biogen, Eisai, Grünenthal, GSK,
Henning Pharma, Interacoustics, J&J, MSD, NeuroUpdate, Otometrics, Pierre- Fabre, TEVA, UCB,
and Viatris. He receives support for clinical studies from Decibel, USA; Cure within Reach, USA;
and Heel, Germany. He distributes “M- glasses” and “Positional vertigo App.” He acts as a
consultant for Abbott, AurisMedical, Bulbitec, Heel, IntraBio, Sensorion, and Vertify. He is an
investor, shareholder, and patent holder of IntraBio. T.B.- E. received speaker’s honoraria and
consultant fees from Actelion and Sano- Genz yme. She acts as a consultant for Azafaros and
machineMD. She received AB honoraria from Zevra and ScenicBio. She acts as a blinded video
rater for Intrabio. Her research has been supported by Baasch- Medicus Foundation, grant- no.
20211001 and InnoSuisse grant- no. 55424.1 IP- LS. M.B. received speaker honoraria from GE
HealthCare IBA, Roche, and Life Molecular Imaging and is an adviser for GE HealthCare, MIAC,
and Life Molecular Imaging, all outside the submitted work. The other authors declare that
they have no competing interests. Data and materials availability: The proteomics data have
been deposited with the ProteomeXchange Consortium via the PRIDE (79) partner repository
with the dataset identiers PXD056188 and PXD056249. C57BL/6- Npc1tm1.2Apl mice were
provided by A. Lieberman (University of Michigan, Ann Arbor, MI, USA) and B6.
Cx3cr1tm1.1(cre)Jung/N mice by S. Jung (Weizmann Institute of Science) under a material
transfer agreement. hiPSCs and the NPC1 depletion mouse model will be shared by S.T. upon
request under a material transfer agreement that will be provided by the DZNE Technology
Transfer Oce (techtransfer@ dzne. de). All data associated with this study are present in the
paper or the Supplementary Materials.
Submitted 19 October 2023
Resubmitted 12 July 2024
Accepted 11 November 2024
Published 4 December 2024
10.1126/scitranslmed.adl4616
Downloaded from https://www.science.org at Deutsches Zentrum fr Neurodegenerative Erkrankungen on December 05, 2024
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The National Institute on Aging and the Alzheimer's Association convened three separate work groups in 2011 and single work groups in 2012 and 2018 to create recommendations for the diagnosis and characterization of Alzheimer's disease (AD). The present document updates the 2018 research framework in response to several recent developments. Defining diseases biologically, rather than based on syndromic presentation, has long been standard in many areas of medicine (e.g., oncology), and is becoming a unifying concept common to all neurodegenerative diseases, not just AD. The present document is consistent with this principle. Our intent is to present objective criteria for diagnosis and staging AD, incorporating recent advances in biomarkers, to serve as a bridge between research and clinical care. These criteria are not intended to provide step‐by‐step clinical practice guidelines for clinical workflow or specific treatment protocols, but rather serve as general principles to inform diagnosis and staging of AD that reflect current science. Highlights We define Alzheimer's disease (AD) to be a biological process that begins with the appearance of AD neuropathologic change (ADNPC) while people are asymptomatic. Progression of the neuropathologic burden leads to the later appearance and progression of clinical symptoms. Early‐changing Core 1 biomarkers (amyloid positron emission tomography [PET], approved cerebrospinal fluid biomarkers, and accurate plasma biomarkers [especially phosphorylated tau 217]) map onto either the amyloid beta or AD tauopathy pathway; however, these reflect the presence of ADNPC more generally (i.e., both neuritic plaques and tangles). An abnormal Core 1 biomarker result is sufficient to establish a diagnosis of AD and to inform clinical decision making throughout the disease continuum. Later‐changing Core 2 biomarkers (biofluid and tau PET) can provide prognostic information, and when abnormal, will increase confidence that AD is contributing to symptoms. An integrated biological and clinical staging scheme is described that accommodates the fact that common copathologies, cognitive reserve, and resistance may modify relationships between clinical and biological AD stages.
Article
Full-text available
The recent introduction of new-generation immunoassay methods allows the reliable quantification of structural brain markers in peripheral matrices. Neurofilament light chain (NfL), a neuron-specific cytoskeletal component released in extracellular matrices after neuroaxonal impairment, is considered a promising blood marker of active brain pathology. Given its sensitivity to a wide range of neuropathological alterations, NfL has been suggested for the use in clinical practice as a highly sensitive, but unspecific tool to quantify active brain pathology. While large efforts have been put in characterizing its clinical profile in many neurological conditions, NfL has received far less attention as a potential biomarker in major psychiatric disorders. Therefore, we briefly introduce NfL as a marker of neuroaxonal injury, systematically review recent findings on cerebrospinal fluid and blood NfL levels in patients with primary psychiatric conditions and highlight the opportunities and pitfalls. Current evidence suggests an elevation of blood NfL levels in patients with major depression, bipolar disorder, psychotic disorders, anorexia nervosa, and substance use disorders compared to physiological states. However, blood NfL levels strongly vary across diagnostic entities, clinical stage, and patient subgroups, and are influenced by several demographic, clinical, and analytical factors, which require accurate characterization. Potential clinical applications of NfL measure in psychiatry are seen in diagnostic and prognostic algorithms, to exclude neurodegenerative disease, in the assessment of brain toxicity for different pharmacological compounds, and in the longitudinal monitoring of treatment response. The high inter-individual variability of NfL levels and the lack of neurobiological understanding of its release are some of the main current limitations. Overall, this primer aims to introduce researchers and clinicians to NfL measure in the psychiatric field and to provide a conceptual framework for future research directions.
Article
Full-text available
Niemann–Pick type C (NPC) disease is a rare progressive lysosomal lipid storage disorder that manifests with a heterogeneous spectrum of clinical syndromes, including visceral, neurological and psychiatric symptoms. This monogenetic autosomal recessive disease is largely caused by mutations in the NPC1 gene, which controls intracellular lipid homeostasis. Vesicle-mediated endo-lysosomal lipid trafficking and non-vesicular lipid exchange via inter-organelle membrane contact sites are both regulated by the NPC1 protein. Loss of NPC1 function therefore triggers intracellular accumulation of diverse lipid species, including cholesterol, glycosphingolipids, sphingomyelin and sphingosine. The NPC1-mediated dysfunction of lipid transport has severe consequences for all brain cells, leading to neurodegeneration. Besides the cell-autonomous contribution of neuronal NPC1, aberrant NPC1 signalling in other brain cells is critical for the pathology. We discuss here the importance of endo-lysosomal dysfunction and a tight crosstalk between neurons, oligodendrocytes, astrocytes and microglia in NPC pathology. We strongly believe that a cell-specific rescue may not be sufficient to counteract the severity of the NPC pathology, but targeting common mechanisms, such as endo-lysosomal and lipid trafficking dysfunction, may ameliorate NPC pathology. This article is part of a discussion meeting issue ‘Understanding the endo-lysosomal network in neurodegeneration’.
Article
Full-text available
Microglial activation plays central roles in neuroinflammatory and neurodegenerative diseases. Positron emission tomography (PET) targeting 18 kDa Translocator Protein (TSPO) is widely used for localising inflammation in vivo, but its quantitative interpretation remains uncertain. We show that TSPO expression increases in activated microglia in mouse brain disease models but does not change in a non-human primate disease model or in common neurodegenerative and neuroinflammatory human diseases. We describe genetic divergence in the TSPO gene promoter, consistent with the hypothesis that the increase in TSPO expression in activated myeloid cells depends on the transcription factor AP1 and is unique to a subset of rodent species within the Muroidea superfamily. Finally, we identify LCP2 and TFEC as potential markers of microglial activation in humans. These data emphasise that TSPO expression in human myeloid cells is related to different phenomena than in mice, and that TSPO-PET signals in humans reflect the density of inflammatory cells rather than activation state.
Article
Full-text available
Niemann-Pick disease type C (NPC) is caused by a deficiency of the NPC1 or NPC2 gene, leading to storages of unesterified cholesterol and sphingolipids. Cerebellar ataxia is a main symptom of NPC and the deep cerebellar nuclei (DCN) is the sole signal output of the cerebellum. In this study, we explored the pathological changes in DCN neurons of Npc1 knockout mice (Npc1-). We first demonstrated that DCN neurons of Npc1- mice had prominent ganglioside GM2 accumulation in the late endosomes but not in the lysosomes. More importantly, Flot2 expression, a marker for the lipid rafts, was lost. Single-nucleus RNA sequencing analysis revealed a generalized reduction in gene expression in DCN neurons, though Camk1d, encoding one of the Ca2+/calmodulin-dependent protein kinases (CaMKs), increased in expression. We treated Npc1- mice with CaMK inhibitor KN-93, but CaMK1D expression increased further. We also fed Npc1- mice with two medications for NPC. We found that miglustat, a sphingolipid synthesis inhibitor, increased the expression of Flot2. Moreover, N-acetyl l-leucine (NALL), an experimental medicine for NPC, recovered Flot2 expression. Therefore, our data suggest that in Npc1- mice, GM2 sequestration and the loss of lipid rafts lead to cell dysfunction and symptoms of NPC.
Article
Background: Niemann-Pick disease type C is a rare lysosomal storage disorder. We evaluated the safety and efficacy of N-acetyl-l-leucine (NALL), an agent that potentially ameliorates lysosomal and metabolic dysfunction, for the treatment of Niemann-Pick disease type C. Methods: In this double-blind, placebo-controlled, crossover trial, we randomly assigned patients 4 years of age or older with genetically confirmed Niemann-Pick disease type C in a 1:1 ratio to receive NALL for 12 weeks, followed by placebo for 12 weeks, or to receive placebo for 12 weeks, followed by NALL for 12 weeks. NALL or matching placebo was administered orally two to three times per day, with patients 4 to 12 years of age receiving weight-based doses (2 to 4 g per day) and those 13 years of age or older receiving a dose of 4 g per day. The primary end point was the total score on the Scale for the Assessment and Rating of Ataxia (SARA; range, 0 to 40, with lower scores indicating better neurologic status). Secondary end points included scores on the Clinical Global Impression of Improvement, the Spinocerebellar Ataxia Functional Index, and the Modified Disability Rating Scale. Crossover data from the two 12-week periods in each group were included in the comparisons of NALL with placebo. Results: A total of 60 patients 5 to 67 years of age were enrolled. The mean baseline SARA total scores used in the primary analysis were 15.88 before receipt of the first dose of NALL (60 patients) and 15.68 before receipt of the first dose of placebo (59 patients; 1 patient never received placebo). The mean (±SD) change from baseline in the SARA total score was -1.97±2.43 points after 12 weeks of receiving NALL and -0.60±2.39 points after 12 weeks of receiving placebo (least-squares mean difference, -1.28 points; 95% confidence interval, -1.91 to -0.65; P<0.001). The results for the secondary end points were generally supportive of the findings in the primary analysis, but these were not adjusted for multiple comparisons. The incidence of adverse events was similar with NALL and placebo, and no treatment-related serious adverse events occurred. Conclusions: Among patients with Niemann-Pick disease type C, treatment with NALL for 12 weeks led to better neurologic status than placebo. A longer period is needed to determine the long-term effects of this agent in patients with Niemann-Pick disease type C. (Funded by IntraBio; ClinicalTrials.gov number, NCT05163288; EudraCT number, 2021-005356-10.).
Article
With the hope that disease-modifying treatments could target the molecular basis of Parkinson's disease, even before the onset of symptoms, we propose a biologically based classification. Our classification acknowledges the complexity and heterogeneity of the disease by use of a three-component system (SynNeurGe): presence or absence of pathological α-synuclein (S) in tissues or CSF; evidence of underlying neurodegeneration (N) defined by neuroimaging procedures; and documentation of pathogenic gene variants (G) that cause or strongly predispose to Parkinson's disease. These three components are linked to a clinical component (C), defined either by a single high-specificity clinical feature or by multiple lower-specificity clinical features. The use of a biological classification will enable advances in both basic and clinical research, and move the field closer to the precision medicine required to develop disease-modifying therapies. We emphasise the initial application of these criteria exclusively for research. We acknowledge its ethical implications, its limitations, and the need for prospective validation in future studies.
Article
The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.