ArticlePDF Available

Galectin‐3 is upregulated in frontotemporal dementia patients with subtype specificity

Wiley
Alzheimer's & Dementia
Authors:

Abstract and Figures

INTRODUCTION Neuroinflammation is a major contributor to the progression of frontotemporal dementia (FTD). Galectin‐3 (Gal‐3), a microglial activation regulator, holds promise as a therapeutic target and potential biomarker. Our study aimed to investigate Gal‐3 levels in patients with FTD and assess its diagnostic potential. METHODS We examined Gal‐3 levels in brain, serum, and cerebrospinal fluid (CSF) samples of patients with FTD and controls. Multiple linear regressions between Gal‐3 levels and other FTD markers were explored. RESULTS Gal‐3 levels were increased significantly in patients with FTD, mainly across brain tissue and CSF, compared to controls. Remarkably, Gal‐3 levels were higher in cases with tau pathology than TAR‐DNA Binding Protein 43 (TDP‐43) pathology. Only MAPT mutation carriers displayed increased Gal‐3 levels in CSF samples, which correlated with total tau and 14‐3‐3. DISCUSSION Our findings underscore the potential of Gal‐3 as a diagnostic marker for FTD, particularly in MAPT cases, and highlights the relation of Gal‐3 with neuronal injury markers.
This content is subject to copyright. Terms and conditions apply.
Received: 26 June 2023 Revised: 4 October 2023 Accepted: 11 October 2023
DOI: 10.1002/alz.13536
RESEARCH ARTICLE
Galectin-3 is upregulated in frontotemporal dementia patients
with subtype specificity
Sergi Borrego–Écija1Agnès Pérez-Millan1,2Anna Antonell1Laura Fort-Aznar1
Elif Kaya-Tilki3Alberto León-Halcón4,3Albert Lladó1,2Laura Molina-Porcel1
Mircea Balasa1Jordi Juncà-Parella1Javier Vitorica4,3,5Jose Luis Venero4,3
Tomas Deierborg6Antonio Boza-Serrano1,3,4Raquel Sánchez-Valle1,2
1Alzheimer’s disease and other cognitive disorders Unit. Service of Neurology,Fundació Recerca Clínic Barcelona-IDIBAPS, Hospital Clínic de Barcelona, Barcelona,
Spain
2Institut of Neurosciences. Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
3Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla,Sevilla, Spain, Sevilla, Spain
4Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla,Spain
5Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED),M adrid, Spain
6Department of Experimental Medical Sciences, Experimental Neuroinflammatory Lab, Lund University, Lund, Sweden
Correspondence
Antonio Boza-Serrano and Raquel
Sánchez-Valle, Alzheimer’s disease and other
cognitive disorders Unit. Service of Neurology,
Fundació Recerca Clínic Barcelona-IDIBAPS,
Hospital Clínic de Barcelona, Barcelona, Spain.
Email: aboza@us.es and rsanchez@clinic.cat
Funding information
Instituto de Salud Carlos III, Grant/Award
Numbers: PI20/0448, PI18/01556,
PI21/00914; Una manera de hacer Europa,
Grant/AwardNumber: PI19/00449;
Generalitat de Catalunya, Grant/Award
Number: SGR 2021-01126; Vetenskapsrådet,
Grant/AwardNumber: 2019-0633; Kungliga
Fysiografiska Sällskapet i Lund, Grant/Award
Numbers: 20191114ABS, 20211129ABS;
Greta och Johan Kocks stiftelser, Grant/Award
Number: 20201201ABS; Juan de la Cierva
Incorporación, Grant/AwardNumber:
IJC2019-040731-I; Spanish Ministerio de
Ciencia e Innovación /FEDER/UE,
Grant/AwardNumber:
PID2021-124096OB-I00; Swedish
Demensfonden; Swedish Brain Foundation;
Crafoord Foundation; Swedish Dementia
Abstract
INTRODUCTION: Neuroinflammation is a major contributor to the progression of
frontotemporal dementia (FTD). Galectin-3 (Gal-3), a microglial activation regulator,
holds promise as a therapeutic target and potential biomarker. Our study aimed to
investigate Gal-3 levels in patients with FTD and assess its diagnostic potential.
METHODS: We examined Gal-3 levels in brain, serum, and cerebrospinal fluid (CSF)
samples of patients with FTD and controls. Multiple linear regressions between Gal-3
levels and other FTD markers were explored.
RESULTS: Gal-3 levels were increased significantly in patients with FTD, mainly across
brain tissue and CSF, compared to controls. Remarkably, Gal-3 levels were higher
in cases with tau pathology than TAR-DNA Binding Protein 43 (TDP-43) pathology.
Only MAPT mutation carriers displayed increased Gal-3 levels in CSF samples, which
correlated with total tau and 14-3-3.
DISCUSSION: Our findings underscore the potential of Gal-3 as a diagnostic marker
for FTD, particularly in MAPT cases, and highlights the relation of Gal-3 with neuronal
injury markers.
KEYWORDS
C9orf72, CSF, frontotemporal dementia, galectin-3, GRN, MAPT, microglia, neuroinflammation
Sergi Borrego–Écija, Agnès Pérez-Millan, Antonio Boza-Serrano, and Raquel Sánchez-Valle contributed equally to this work.
This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any
medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
© 2023 The Authors. Alzheimer’s & Dementia published by Wiley Periodicals LLC on behalf of Alzheimer’s Association.
Alzheimer’s Dement. 2024;20:1515–1526. wileyonlinelibrary.com/journal/alz 1515
1516 BORREGO–ÉCIJA ET AL.
Association; G&J Kock Foundation; Olle
Engkvist Foundation; Gamla Tjänarinnor
Foundation; Swedish Medical Research
Council; Swedish Parkinson Foundation;
Swedish Parkinson Research Foundation; A.E.
Berger Foundation
1BACKGROUND
Frontotemporal dementia (FTD) encloses a group of neurodegener-
ative disorders that have in common the neurodegeneration of the
frontal and temporal lobes.1Due to its early onset, with most peo-
ple presenting symptoms around the sixth decade of life, FTD is
the second most common form of early-onset neurodegenerative
dementia, after Alzheimer’s disease (AD).2,3 Clinically, FTD includes
three different syndromes: the behavioral variant of FTD (bvFTD),
the semantic variant of primary progressive aphasia (svPPA), and
the non-fluent/agrammatical variant of primary progressive apha-
sia (nfvPPA).4,5 FTD is a highly heritable disorder with mutations in
Chromosome 9 open reading frame 72 (C9orf72), Granulin (GRN), and
Microtubule-associated protein Tau (MAPT), explaining most genetic
cases.6,7 The neuropathological substrate of FTD is frontotemporal
lobar degeneration (FTLD). FTD can be classified according to the
abnormally deposited protein, which can be the tau protein (FTD-tau),
the transactive response (TAR) DNA-binding protein 43 (FTD-TDP-43),
or the FET family of proteins (FTD-FET).1,8,9
Along with neuronal dysfunction/death and protein accumulation,
central immune system activation is a major factor in the progres-
sion of the pathology in FTD.10 Abnormal protein conformation and
accumulation activate the immune system, leading to neuroinflam-
mation. This response involves glial activation and increased levels
of pro-inflammatory factors.10 Microglial cells are the main pool of
innate immune cells in the brain, and their phenotype is crucial to
understand the neuroinflammatory response. Recently, Malpetti et al.,
demonstrated that microglial activation measured by [11C]PK11195
in the frontal cortex could predict cognitive decline in patients with
FTD.11 Positron emission tomography (PET) studies using transloca-
tor protein (TSPO) have detected abnormal microglial activity and
protein aggregation in familial cases of FTD.12 PET analysis also
revealed that microglial activation seems more prominent in fron-
totemporal regions.12–14 The microglial activation pattern detected
by PET analysis has also been observed in histological postmortem
studies.15,16 Neuropathological studies reveal microglial activation in
cortical areas and more pronounced involvement in white matter than
gray matter.15,16
A key molecule involved in microglial activation in neurodegen-
erative diseases is galectin-3 (Gal-3), a beta-galactosidase binding
protein expressed mainly by microglial cells and associated with
neurodegeneration.17–19 Gal-3 is released into the extracellular
space and acts in an autocrine or paracrine manner by binding to
different membrane receptors, such as Toll Like Receptor 4 (TLR4)
and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2).20,21
We have demonstrated previously that Gal-3 is an important reg-
ulator microglial activity in AD21 and in the aggregation process of
α-synuclein and Lewy body formation in Parkinson’s disease.22,23 Of
interest, Gal-3 was found primarily in microglia clustering around
amyloid beta (Aβ) plaques and using 5xFAD model of AD lacking Gal-3
reduced the Aβburden and improved their cognitive performance.21
Our study also confirmed that Gal-3 acts as an endogenous TREM2
ligand, a central player in the regulation of microglial activation under
disease conditions.21 It is important to note that in AD we found
Gal-3 in cerebrospinal fluid (CSF) to be strongly associated with
neuroinflammation markers, synapse loss, and cognitive decline.24
A recent study analyzing over 2000 human AD brain tissue samples
identified a microglia module as a highly affected process in AD. The
study found that Gal-3 ranked fifth among the top 30 microglial tran-
scripts associated with AD, suggesting its significance in AD pathology
and microglia dysfunction.25 In support of this perspective and with
regard to FTD, transgenic mice carrying FTD-related genes exhibit
strong and even aberrant microgliosis and inflammatory response,
especially evident in mice lacking GRN26,27 and P301S MAPT mice.28
Moreover, a recent study has shown that mutations in GRN mice led
to significant microglial activation, which seems to be influenced by
GPNMB (glycoprotein non-metastatic melanoma protein B) and Gal-3
in mice.29
In the context of traumatic injury models, it has been observed that
TDP-43 induction contributes to the activation of microglial cells, lead-
ing to the upregulation of Gal-3.30 Like in AD, the role of neuroinflam-
mation and immune-mediated mechanisms in the development of FTD
is well established.10 Indeed, a causal role of neuroinflammation has
been proposed, as evidenced by increased microglial activation in the
frontal and temporal cortices, astrogliosis, and abnormal expression of
pro- and anti-inflammatory factors in CSF and blood.10,31,32
Consequently, the present study explores Gal-3 levels and their link
to FTD through brain, CSF, and serum biomarker analysis for FTD and
its subtypes.
2METHODS
2.1 Clinical cohort
A total of 133 participants were recruited at the Alzheimer’s Dis-
ease and Other Cognitive Disorders Unit of the Hospital Clínic de
Barcelona, including 115 patients fulfilling criteria for bvFTD (62
patients), svPPA (28 patients), or nfvPPA (25 patients), and 18 healthy
controls (HCs) (Table 1). The clinical CSF FTD cohort has 6 MAPT
BORREGO–ÉCIJA ET AL.1517
symptomatic mutation carriers, 5 presymptomatic MAPT mutation car-
riers, 13 GRN mutation carriers, and 13 C9orf72 expansion carriers. All
the participants underwent a complete clinical and neuropsychologi-
cal examination. The participants included in the study had Gal-3 levels
in CSF (N=133 participants) and/or serum (N=120 participants). All
subjects were Caucasian white and did not present other neurological
diseases. The study was approved by the ethics committee of the Hos-
pital Clínic de Barcelona (HCB 2019/0105). Written informed consent
was obtained from all participants.
2.2 Human brain tissue
Frozen frontal cortices from 10 cognitively healthy controls and 37
FTD cases were obtained from the Neurological Tissue Bank, Biobanc-
Hospital Clínic-IDIBAPS, Barcelona, Spain. All subjects were White.
The control cases were not diagnosed with any neurological condi-
tion (other than migraine or essential tremor) during life, and the
postmortem examination did not disclose findings supporting any neu-
ropathological diagnosis, although minor vascular changes or a low
grade of incidental pathologies were not exclusionary.33 FTD cases
included 18 mutation carriers (4 MAPT,5GRN,and8C9orf72;Table2)
and 19 sporadic FTD (10 tau and 9 TDP-43; Table 2). Written informed
consent for using brain tissue and clinical data for research purpose
was obtained from all patients or their next of kin following the Inter-
national Declaration of Helsinki and Europe’s Code of Conduct for
Brain Banking. The medical ethics committee of the institutional review
board of the Hospital Clínic (Barcelona) has approved the procedures
for brain tissue collection.
2.3 ELISA
Enzyme-linked immunosorbent assay (ELISA) plates from Abcam
(ab269555) were used to measure the levels of Gal-3 (detection range
58.8 to 2000 pg/mL) in tissue homogenates, CSF, and serum samples.
The protocol was carried out according to the manufacturer’s instruc-
tions. A Biotek Synergy 2 was used to read the ELISA Gal-3 assay.
All samples were run in duplicate once. Mean inter-assay Coeficients
of Variances (CV) was 6.23. Samples were distributed in the plates
according to the clinical group in similar proportions to avoid a bias
caused by the plate. The ELISA and the analysis of the raw data were
performed by different persons. CSF samples were not diluted. Tis-
sue homogenates were diluted 1:100. Serum samples were diluted
1:20 or 1:50; the correction factor on diluted samples was applied
when needed for the comparison. Our kit has been used previously to
measure Gal-3 levels in brain, CSF, and serum samples24,34
2.4 Protein extraction
Radioinmuno precipitation Assay Buffer (RIPA) solution was pre-
pared with a protease inhibitor (cOmplete Protease Inhibitor Cocktail,
RESEARCH IN CONTEXT
1. Systematic review: Inflammation is a critical component
of frontotemporal dementia (FTD). We explore the liter-
ature using sources such as PubMed. We found only five
publications under the term’s “microglia”, “cerebrospinal
fluid”, and “frontotemporal dementia”. Therefore, the clin-
ical practice suffers from a notable absence of reliable
microglial proinflammatory markers associated with FTD.
2. Interpretation: We have uncovered evidence of a sub-
stantial upregulation of the microglial marker galectin-3
(Gal-3) in patients with FTD, thereby emphasizing the piv-
otal role of neuroinflammation in FTD pathogenesis and
the utility of microglial markers as biomarkers. Notably,
disparities in FTD subtypes, particularly the elevated lev-
els of Gal-3 observed in Microtubule-associated protein
Tau (MAPT) mutation carriers, suggest the possibility of
FTD subtype-specific neuroinflammatory patterns.
3. Future directions: Future research must prioritize cere-
brospinal fluid (CSF) longitudinal and independent cohort
studies to determine the Gal-3-dependent neuroinflam-
matory response in the course of FTD. In addition, neu-
ropathological investigations are needed to identify brain
regions wherein microglial activation manifests most
prominently.
Roche) and a phosphatase inhibitor (PhosphoStop, Roche). Frozen
human tissue samples of the hippocampus and cerebral cortex were
homogenized in RIPA buffer (1 mL/100 mg of tissue, Sigma-Aldrich,
Germany) and sonicated briefly in ice. The pellet was ultracentrifuged
subsequently at 25000 relative centrifugal force (rcf) for 25 min. The
supernatant was isolated and used for analysis. Protein concentration
was determined using a BCA Kit (Bio-Rad) according to the manu-
facturer’s protocols. All the samples were normalized to the same
concentration prior to the analysis.
2.5 CSF biomarker analysis
The samples were processed within 2 h from needle-to-freezer (mean
time 45 min). Both CSF and blood samples were centrifuged at 2000 ×
gfor 10 min at 4C. Then they were stored in polypropilene tubes and
kept at 80C until use: Storage tubes for CSF (eppendorf 0.5 mL Ref.
72.730.007 (Sarstedt)) and for serum (cryotubes 2 mL Ref. 363401PK
(Nunc)).
Core AD biomarker concentrations were measured with INNOTEST
ELISAs following the manufacturer’s instructions (Fujirebio, Ghent,
Belgium). CSF neurofilament light chain (NfL) concentration was mea-
sured using the ELISA kit of Uman Diagnostics distributed by IBL
International (Hamburg, Germany). For γ14-3-3 protein, we used the
1518 BORREGO–ÉCIJA ET AL.
TAB L E 1 Group summaries are given as each measure’s mean and standard deviation in brackets.
Healthy Controls All FTD bvFTD svPPA nfvPPA p-value*
N18 115 62 28 25
NCSF 18 99 36 26 16
NSerum 13 107 50 24 20
Sex (male/female) 6/12 57/58 32/30 15/13 10/15 0.20
Age, years, mean (SD) 54.3 (4.8) 63.5 (9.1) 63.0 (9.2) 64.0 (8.4) 64.2 (9.9) <0.001
Duration, years, mean (SD) NA 3.4 (2.3) 3.8 (2.7) 2.9 (1.3) 3.0 (2.1)
Mean MMSE score (SD) 29.1 (1.1) 23.6 (5.8) 23.4 (6.3) 23.5 (5.9) 24.3 (4.7) <0.001
Genetic (%) 0 (0%) 35 (30%) 25 (40%) 2 (7%) 8 (32%) <0.001
Serum Gal-3, median [Q1, Q3] 1428.7 [1296.6, 1644.4] 1463.3 [1231.0, 1758.4] 1491.6 [1292.1, 1834.4] 1450.6 [1211.0, 1565.9] 1502.8 [1163.2, 1727.3] 0.80
CSF Gal-3, median [Q1, Q3] 459.0 [433.9, 501.6] 545.6 [437.2, 667.2] 567.4 [434.0, 701.1] 532.8 [505.0, 635.4] 484.0 [428.0, 652.6] <0.05
CSF Aβ42, median [Q1, Q3] 901.8 [664.7, 1088.5] 761.7 [589.9, 944.5] 788.5 [550.4, 979.5] 730.5 [627.2, 896.8] 682.0 [589.9, 876.0] 0.13
CSF p-tau, median [Q1, Q3] 44.7 [37.4, 57.1] 40.5 [32.1, 55.2] 39.0 [32.0, 54.0] 37.3 [32.8, 49.0] 46.0 [35.3, 58.3] 0.30
CSF t-tau, median [Q1, Q3] 192.4 [165.5, 266.9] 276.4 [210.1, 409.5] 290.0 [214.0, 427.2] 260.5 [214.9, 327.5] 269.2 [209.4, 473.5] <0.01
CSF YKL-40, median [Q1, Q3] 214.5 [170.5, 245.3] 289.0 [207.6, 371.5] 321.6 [193.1, 391.3] 275.8 [252.0, 317.4] 296.0 [214.5, 381.0] <0.05
CSF NfL, median [Q1, Q3] 401.8 [358.2, 501.5] 2232.6 [1286.2, 3962.0] 2390.3 [1244.7, 4023.6] 1912.9 [1391.0, 2579.6] 2537.1 [1191.0, 4102.7] <0.001
CSF 14-3-3, median [Q1, Q3] 2571.9 [2231.2, 3001.4] 4075.0 [3042.0, 5301.3] 4133.4 [3042.0, 5509.7] 3539.9 [3155, 4997.2] 3835.9[2968.1, 4782.7] <0.001
The p-value is from the comparison between controls and all the FTD patients.
Abbreviations: bvFTD, behavioralvariant frontotemporal dementia; nfvPPA, non-fluent variant primary progressive aphasia; svPPA, semantic variant primary progressive aphasia. *Between all FTD and controls.
BORREGO–ÉCIJA ET AL.1519
TAB L E 2 Group summaries of cortical samples, both genetic and sporadic, used in the study..
Healthy
Controls MAPT GRN C9orf72 sFTD-tau sFTD-TPD-43 All FTD p-value*
N10 4 5 8 10 9 37
Sex (male/female) 5/5 3/1 2/3 4/5 4/6 6/3 20/18
Age, years, mean (SD) 81.3 (12.3) 59.5 (3.1) 66.4(4.8) 70.0 (11.9) 72.1 (5.5) 72.0 (7.9) 67.5 (2.1)
Disease duration, mean (SD) 6.3 (2.1) 6.6 (2.4) 5.3(4.08) 8 (3.5) 11 (4.24) 8.05 (3.9)
Brain Gal-3, median [Q1 Q3] 472 [449 524] 1509 [1156 1837] 1900 [1860 1947] 714 [454 767] 1137 [1046 –1263] 589 [430 735] 974 [669 1286] <0.001
Abbreviations: MAPT, microtubule-associated Protein Tau; GRN, Granulin; C9orf72, chromosome 9 open reading frame 72; sFTD, sporadic Frontotemporal dementia; TDP-43, Tar DNA binding protein 43.
* Between all FTD and controls.
ELISA kit CircuLex 14-3-3 gamma (MBL International Corporation,
Woburn, MA, USA) with a CSF sample dilution of 1:5. CSF YKL-40 con-
centration was measured with an ELISA from QUIDEL (San Diego, CA,
USA) using a CSF sample dilution of 1:2.5. The antibodies for the detec-
tion of these four biomarkers have been used by us and other authors
on previous studies with CSF samples in neurodegenerative demen-
tias. These biomarkers have been also studied individually using other
technologies or antibodies.35–38
All analyses were performed by duplicate and experienced labora-
tory personnel blinded to clinical diagnosis. We are participants of the
Alzheimer’s Association QC program,5and Aβ42, total tau (t-tau), and
phosphorylated tau 181 (p-tau181) levels obtained in our laboratory
have been consistently within mean ±2SD.
2.6 Statistical analysis
All FTD-related variables from the cortical tissue were analyzed with
the Mann–Whitney test to compare independent groups. For multi-
ple comparisons, the Kruskal–Wallis test followed by Dunn test was
used as a post hoc correction to identify the pair-wise group differ-
ences. Receiver-operating characteristic (ROC) curves analysis was
performed to assess the diagnostic accuracy of the Gal-3. For the sta-
tistical analysis of the CSF and serum levels of Gal-3, permutation tests
with age and sex added as covariables were used. The p-values of these
results were correct for multiple comparisons with the Benjamini &
Hochberg correction. We compared the HCs, genetic FTD, and spo-
radic FTD patients with the same procedure. Finally, we studied in
detail group differences for the different FTD groups (clinical pheno-
type and genetic form) and HCs with the same methodology. Multiple
linear regression corrected by age and sex were applied to evaluate
the association between the CSF Gal-3 and the other CSF FTD-related
markers levels (Aβ42, t-tau, p-tau181, 14-3-3, YKL-40, and NfL levels)
for abovementioned groups. For all the analyses, statistical significance
was set at p-value <0.05. Statistical analyses were carried out using
GraphPad Prism version 9 (GraphPad Software, San Diego, CA, USA),
SPSS v. 26 (IBM Corp., Armonk, NY, USA) software, and the language R
in R-studio version 4.2.1 (https://www.r-project.org).
3RESULTS
3.1 Demographic and clinical characteristics of
participants
Demographic information of the study population and group statis-
tics are shown in Table 1. Controls were younger than FTD patients
(p-value <0.001). Because Gal-3 levels in CSF showed a moderate cor-
relation with age in the whole cohort (r =0.42, p-value <0.001), further
statistical analyses were corrected for age. There were statistically sig-
nificant differences in Gal-3 levels grouping by sex in the whole cohort;
thus the analyses were also corrected for sex. We did not find a signifi-
cant link between Gal-3 and disease duration. However, it is important
1520 BORREGO–ÉCIJA ET AL.
FIGURE 1 Galectin-3 (Gal-3)protein levels in brain cortex. (A) Control vs FTD cases. Gal-3 levels were significantly increased in FTD compared
with controls. (B) FTD-tau vs FTD-TDP-43. FTD cases with tau pathology had higher brain levels of Gal-3 than those with TDP-43 pathology. (C)
Brain Gal-3 levels in genetic carriers. MAPT and GRN carriers showed increased Gal-3 levels in brain. Cortical brain tissue was analyzed with the
Mann–Whitney Uand the Kruskal–Wallis test (multiple comparison) followed by Dunn’s test used as a post hoc correction to identify the pair-wise
group differences. (See Section 2.6 for further statistical analysis description.
to mention that most samples in our study were collected at the time
of diagnosis, with only a few collected during the later stages of the
disease.
3.2 Galectin-3 levels are upregulated in cortical
tissue from FTD
First, we evaluated Gal-3 levels in FTD brain samples. Gal-3 level was
upregulated in patients with FTD as a whole compared to control sam-
ples (Figure 1A). When compared according to their neuropathological
substrate, FTD-tau showed higher Gal-3 levels compared to FTD-TDP-
43 cases (here we included genetic and sporadic cases with tau or
TDP-43 deposition in the comparison) (Figure 1B). Finally, we evalu-
ated Gal-3 levels in genetic cases and found increased Gal-3 levels in
MAPT and GRN carriers compared to controls (Figure 1C). Both MAPT
and GRN genetic cases also displayed higher Gal-3 values compared
to C9orf72 (Figure 1C). In contrast, Gal-3 levels in C9orf72 expansion
carriers did not differ from controls.
3.3 Elevated Gal-3 levels in CSF in FTD samples
Following the brain sample analyses, we measured Gal-3 levels in
CSF samples from genetic and sporadic FTD patients (Figure 2).
CSF Gal-3 levels were elevated in FTD comparison to HC samples
(Figure 2A). For the next analysis, we separate sporadic by clinical
phenotype (bvFTD, svPPA, and nfvFTD) and genetic samples by type
of mutation (MAPT,GRN,andC9orf72)(Figure2B, C). The analysis of
the sporadic variants of FTD resulted in higher Gal-3 levels in bvFTD
compared to svPPA (Figure 2B), nfvFTD (Figure 2B), and HC samples
(Figure 2B).
We observed a significant elevation of Gal-3 levels in MAPT carrier
samples compared to GRN carriers (Figure 2C), C9orf72 (Figure 2C),
and HC samples (Figure 2C). No statistically significant differences
were found between GRN and C9orf72 groups. CSF Gal-3 levels were
significantly higher for symptomatic carriers than presymptomatic
MAPT mutation carriers (Figure 2C). In our cohort, Gal-3 CSF could be
used to differentiate FTD from controls: ROC curve (area under the
curve [AUC] 0.67).
3.4 Levels in serum
Serum Gal-3 levels were also increased in FTD patients compared to
controls (Figure 2D). The ROC curve, however, showed a poor perfor-
mance of serum Gal-3 differentiating FTD from controls (AUC: 0.55).
No significant differences were found between any of the sporadic syn-
dromes of FTD and HCs (Figure 2E). No difference in serum Gal-3 levels
were found between the different causal mutations of genetic FTD and
HCs (Figure 2F).
3.5 Association of CSF Gal-3 with other
biomarkers
When studying the multiple linear regressions between different CSF
biomarkers (Aβ42, t-tau, p-tau181, 14-3-3, YKL-40, and NfL) and Gal-
3, we observed a significant association for FTD patients but not
for the HCs (Table 3). The t-tau levels for FTD presented a moder-
ate association with CSF Gal-3 (R =0.43, p-value adjusted <0.001)
and with 14-3-3 levels (R =0.45, p-value adjusted <0.001, respec-
tively). Gal-3 also presented a weak relationship with Aβ42 for
FTD patients (R =0.39, p-value adjusted <0.001). No statistically
BORREGO–ÉCIJA ET AL.1521
FIGURE 2 Galectin-3 levels in CSF (A, B, C) and serum (D, E, F). The p-values of the plots were adjusted for multiple comparisons and corrected
by age and sex. (A) CSF Gal-3 levels in FTD cases are increased compared with controls. (B) Comparing FTD clinical syndromes, the bvFTD group
showed higher levels of CSF Gal-3 than controls, svPPA, and nfvPPA. (C) CSFGal-3 levels in mutation carriers revealed higher levels in MAPT
carriers. (D) Serum levels of Gal-3 were elevated in FTD patients. (E) Serum Gal-3 levels between clinical syndromes showed higher levels in
nfvPPA than in svPPA, with no differences in other comparisons. (F) No differences in serum Gal-3 levels were found between mutation carriers.
(See Section 2.6 for further statistical analysis description.)
significant relationship was found between Gal-3 and p-tau181, NfL, or
YKL-40. Table 3shows the coefficients details of all the multiple linear
regressions.
4DISCUSSION
In this study, we examined Gal-3, a microglial marker, across the
FTD spectrum in neuropathological and clinical cohorts (CSF and
serum levels) of both sporadic and genetic FTD patients. Our findings
revealed elevated Gal-3 levels in FTD subjects’ brains, CSF, and serum,
thereby highlighting neuroinflammation’s significance and the role of
Gal-3-expressing microglia in FTD’s neurodegenerative mechanism.
Galectins play a crucial role in the brain’s neuroinflammatory response
by identifying glycan structures and sensing their modifications both
intracellularly and extracellularly. Despite the importance if galectins,
the regulation of their expression remains elusive.39 Recent investiga-
tions found substantial upregulation of Gal-3 in GRN knockout Induced
Pluripotent stem cells (iPSC)-derived microglia.40 Gal-3 was detected
in human studies and FTD mouse models with GRN gene knockout,
emerging as the primary upregulated protein alongside GPNMB.29
Other galectins such as galectin-1 (Gal-1) and galectin-9 (Gal-9) have
been involved in the regulation of neuroinflammatory processes.41,42
However, Gal-1 has been shown to deactivate microglial activation,41
thereby reducing the associated inflammatory response. On the other
hand, Gal-9 is produced mainly by astrocytes but not microglial cells
and it has been shown to indirectly promote microglial activity.42 Gal-9
CSF levels have been shown to be increased in secondary progressive
multiple sclerosis43 and have been also linked with central nervous
system (CNS) immune activation and poor cognitive performance in
human immunodeficiency virus (HIV) infected individuals.44
Due to FTD’s substantial heterogeneity, significant differences
emerged among clinical, genetic, and neuropathological subtypes. A
relevant maker of microglial activation in disease conditions is TREM2,
which is implicated in the neuroinflammatory response in AD, and
has shown a strong association with FTD-tau.45–48 We demonstrated
1522 BORREGO–ÉCIJA ET AL.
TAB L E 3 Multiple linear regression coefficients for assessing the different CSF biomarkers trajectories by CSF Gal-3 level according to age and sex.
Aβ42 p-Tau181 Total tau NfL YKL-40 14-3-3
βCI 95% p-value βCI 95% p-value βCI 95% p-value βCI 95% p-value βCI 95% p-value βCI 95% p-value
R20.1365 0.2143 0.05659 0.01183 0.1927 0.2590
Gal3 0.6 0.3– 0.9 <0.001 0.0 0.0– 0.10.022 0.4 0.2– 0.6 <0.001 1.0 -1.9– 3.8 0.50 0.1 -0.1– 0.3 0.40 4.7 2.5– 7.0 <0.001
Age -6.5 -12.0– 1.4 0.013 -0.1 -0.7– 0.50.80 1.6 -1.5– 4.8 0.30 12.0 -38.0–62.0 0.60 5.8 2.4– 9.3 0.0010 41.0 4.1– 78.0 0.030
Sex
(Female
vs. male)
-11.0 -107.0 –85.0 0.80 -6.9 -17.0–3.50.20 -59.0 -117.0–0.1 0.050 16.1 -779.0–1102.0 0.70 16.0 -41.0–73.0 0.60 -527.0 -1186.0– 132.0 0.12
previously that Gal-3 expressed by microglial cells can act as a TREM2
ligand.21 In addition, we have shown a clear association of Gal-3 with
tau and p-tau181 in CSF in AD and have demonstrated a clear co-
localization of microglial cells expressing Gal-3 with tau protein in asso-
ciation of Aβplaques.24 Therefore, Gal-3s unique expression, pivotal
role in microglial activation, and relevance in FTD progression mark it
as a key molecule for future exploration, differentiating it from other
galectins in understanding neuroinflammation in neurodegenerative
disorders.
Microglial activation correlates strongly with FTD progression and
cognitive decline.11,12 Therefore, the reactive microgliosis observed
in FTD might contribute to an upregulation of Gal-3 levels. In our
study, FTD-tau exhibited higher Gal-3 levels in the brain than FTD-
TDP-43. Moreover,the patients we analyzed carrying MAPT mutations,
which cause tau pathology, showed increased Gal-3 levels in the brain.
Previous neuropathological works have shown that FTD-MAPT cases
present strong microglial cell activation, even more than other FTD
cases.49,50 Indeed, reactive gliosis is also prevalent in tauopathies and
FTD mouse models.27–29,51,52 Likewise, the elevation of Gal-3 in MAPT
carriers was evident in CSF but not serum samples, indicating Gal-
3 CSF’s superior performance in these cases. Gal-3 serum levels did
not distinguish FTD clinical forms or genetic samples, likely due to
its peripheral origin (e.g., monocytes), rather than CNS microglia,53,54
due to posttranslational modification, like phosphorylation, hampering
bloodstream release55–57 and its upregulation in other comorbidities
outside the brain, such as heart disease.58
Regarding Gal3 levels in patients with genetic FTD, symptomatic
MAPT carriers had higher Gal-3 levels than presymptomatic individu-
als, implying that Gal-3 could be a biomarker for MAPT carrier clinical
onset or progression, pending larger longitudinal validation. In vivo
evidence for presymptomatic neuroinflammation in a MAPT mutation
carrier59 has been found in recently.The study indicated that microglial
activation is a better marker for discriminating MAPT mutation carriers
from controls than tau protein aggregation at this pre-symptomatic
disease stage of FTD.59 This result might indicate that microglial acti-
vation in MAPT mutation carriers might be an early event rather than
a consequence of protein dysregulation,59 which might open up new
possibilities for early anti neuroinflammatory treatments. In mouse
models, Van Olst and colleagues investigated neuroinflammation in
P301S MAPT mice and found that microglia changes started after neu-
ronal p-tau deposition in the early stages of tau processing.52 In this
model, microglia adopted a p-tau-associated phenotype, morpholog-
ical and functionally distinct from wild-type microglia, after neuronal
p-tau accumulation was initiated. Other studies have revealed the
pivotal role of microglial cells and apolipoprotein E gene (APOE)in
driving neurodegeneration in a mouse model of tauopathy,60 under-
scoring the potential critical significance of microglia in the context
of FTD.
Our data demonstrated a positive association between CSF Gal-3
levels and two markers of neuronal dysfunction, 14-3-3, and t-tau.61–63
Indeed, neuroinflammatory response has been linked to synaptic
dysfunction.64 We also demonstrated a positive relationship between
CSF Gal-3 levels and t-tau in AD patients along with GAP-43 and
BORREGO–ÉCIJA ET AL.1523
neurogranin, markers of synaptic dysfunction.24 However, in this study
no association was found between Gal-3 and p-tau 181, contrary to
our previous study in AD patients where we observed a significant
association.24 This suggests that the mechanism of neuroinflammatory
response is similar but not identical in AD compared to FTD. The timing
of neuroinflammation in FTD, whether preceding neuronal dysfunction
or ensuing it, is presently unclear.
Regarding GRN mutation, brain Gal-3 levels were also elevated in
FTD subjects carrying GRN mutations but not in C9orf72 expansion
carriers. Other markers of inflammation, such as Glial Fibrillary Acid
Protein (GFAP), have been shown to be differentially elevated in GRN
carriers.65 The GRN gene encodes the progranulin protein, which is
involved in many biological processes, including inflammation, particu-
larly in deactivating glial cells.30 Mutations in GRN result in progranulin
haploinsufficiency, suggesting that deficiency of progranulin in GRN
mutation carriers may lead to pro-inflammatory glial activation and
increased levels of Gal-3.30 Indeed, recent research has highlighted
the crucial role of activated microglia in GRN knockout mice, which
drive disease progression by inducing neurodegeneration and TDP-43
protein aggregation during aging.66 Of interest, proteomic analysis of
GRN KO identified GPNMB and Gal-3 as two of the most enriched pro-
teins in the GRN KO brain proteome, particularly in aged animals29 and
substantial upregulation of Gal-3 was found in GRN KO iPSC-derived
microglia.40 Notwithstanding, the finding of increased brain Gal-3 lev-
els in the cortex of GRN cases was not reflected in CSF or serum in our
study. This may be related to different magnitudes or dynamics of Gal-
3 levels in these tissues, or due to Gal-3 upregulation occurring only in
the latest stages of the disease in GRN carriers. More research would
be needed to elucidate the role of Gal-3 in GRN mutation carriers.
Recent work from Woollacott and colleagues evaluated three glia-
derived biomarkers in CSF: TREM2, YKL-40, and chitotriosidase in
183 participants from the Genetic FTD Initiative (GENFI), includ-
ing C9orf72,GRN,andMAPT mutation carriers and controls. Only
chitotriosidase showed increased levels in symptomatic GRN muta-
tion carriers; the other group comparisons failed to show statistically
significant differences.65,67
The differences mentioned above between neuropathological and
genetic subgroups of FTD points to Gal-3 as a promising biomarker
to distinguish between molecular subtypes of FTD. Although our clin-
ical cohort did not include cases with confirmed neuropathology, our
results indicated that clinical phenotypes usually associated with FTD-
tau (i.e., nfvPPA) showed increased levels of CSF Gal-3 compared with
clinical phenotypes usually associated with FTD-TDP-43 (i.e., svPPA).
When determining the diagnostic significance of Gal-3 in distinguish-
ing between FTD patients and controls, our analysis of ROC curves
revealed that both serum and CSF Gal-3 levels displayed less accu-
racy compared to more established biomarkers such as NfL for this
comparison.68–71
We note several limitations in our work. First, despite the consider-
able sample size of our cohort of patients with FTD, the heterogeneity
of this disease makes smaller clinical or genetic subgroups leading
to a lack of statistical power needed to explore subtle differences
between subgroups. Second, even though we included a neuropatho-
logical cohort where we found differences in brain Gal-3 levels,
the participants included in the clinical cohort lack neuropatholog-
ical confirmation. Acknowledging the need for validation through
a replication cohort, we are aware of limitations in obtaining CSF
genetic samples from FTD individuals. In addition, we recognize
the constrained sensitivity of the applied Gal-3 ELISA, as more
advanced/validated platforms like single molecule array (Simoa,
Quanterix) or Mesoscale Discovery platform (Mesoscale Diagnos-
tics) lack specific Gal-3 assays. In addition, there was a significant
age difference between FTD patients and controls, which may have
been a confounding factor, although the analysis was adjusted for
age difference. Although we did not find any significant correlation
between Gal-3 and disease duration, it is important to mention that
most samples in our study were collected at the time of the clinical
diagnosis, with few samples collected in the later stages of the disease.
A longitudinal approach would be needed to determine the association
between Gal-3 level and the progression of the pathology. Finally, the
presence of other co-pathologies might also induce Gal-3 elevation.
However, the individuals in our cohort underwent measurements of
AD-related biomarkers, and their values indicated the absence of AD
pathology.
To sum up, our study robustly establishes heightened Gal-3 levels
in patients with FTD, underscoring its pivotal role in neuroinflamma-
tion and potentially driving the disease pathogenesis. This deepens our
comprehension of FTD’s mechanisms, highlighting microglial markers
as valuable biomarkers. Notably, FTD subtype variations, particularly
a unique Gal-3 increase in MAPT mutation carriers, signifying subtype-
specific neuroinflammation. Our findings align with preclinical models,
accentuating neuroinflammation’s acceleration of FTD progression.
This accentuates potential immunomodulatory therapies and suggests
evaluating microglial activation for refined clinical trial participant
selection.
ACKNOWLEDGMENTS
The authors thank patients, their relatives, and healthy controls for
participating in the research. This work was supported by Instituto
de Salud Carlos III, Spain (grant no. PI20/0448 to Dr R. Sanchez-
Valle, Instituto de Salud Carlos III, Spain, co-funded by the EU (FEDER)
“Una manera de hacer Europa” and PI19/00449 to Dr Lladó) and
Generalitat de Catalunya (SGR 2021-01126). Dr S. Borrego-Écija is a
recipient of the Joan Rodés Josep Baselga grant from FBBVA. Anto-
nio Boza-Serrano, PhD is recipient of the Vetenskapsrådet grant,
2019-0633, Kungliga Fysiografiska Sällskapet i Lund, 20191114ABS
and 20211129ABS, Greta och Johan Kocks stiftelser, 20201201ABS,
and Juan de la Cierva Incorporación—IJC2019-040731-I. Professor
Jose Luis Venero is recipient of Spanish Ministerio de Ciencia e
Innovación /FEDER/UE (PID2021-124096OB-I00). Professor Javier
Vitorica is recipient of Instituto de Salud Carlos III, Union PI18/01556,
PI21/00914. Professor Tomas Deierborg is recipient of: Swedish
Demensfonden, The Strategic Research Area MultiPark (Multidisci-
plinary Research in neurodegenerative diseases) at Lund University,
the Swedish Brain Foundation, Crafoord Foundation, Swedish Demen-
tia Association, G&J Kock Foundation, Olle Engkvist Foundation,
1524 BORREGO–ÉCIJA ET AL.
Gamla Tjänarinnor Foundation, the Swedish Medical Research Council,
the Swedish Parkinson Foundation, the Swedish Parkinson Research
Foundation, the A.E. Berger Foundation.
CONFLICT OF INTEREST STATEMENT
R.S.V. has served in advisory boards meetings for Wave Life Sciences,
Ionis, UCB, Prevail, Pfizer, and Novo Nordisk and has received personal
fees for participating in educational activities from Roche Diagnostics
and Neuroxpharma. The other authors declare no conflicts of interest;
they declare that the research was conducted without any commercial
or financial relationships that could be construed as a potential conflict
of interest. Conflicts of Interest Author disclosures are available in the
Supporting Information.
CONSENT STATEMENT
All the subjects provided informed consent and the study was
approved by the Hospital Clínic de Barcelona Ethics Committee (HCB
2019/0105).
REFERENCES
1. Rademakers R, Neumann M, Mackenzie IR. Advances in understand-
ing the molecular basis of frontotemporal dementia. Nat Rev Neurol.
2012;8(8):423-434. doi:10.1038/nrneurol.2012.117
2. Ratnavalli E, Brayne C, Dawson K, Hodges JR. The prevalence of
frontotemporal dementia. Neurology. 2002;58(11):1615-1621. doi:10.
1212/wnl.58.11.1615
3. Garre-Olmo J, Genis Batlle D, del Mar Fernandez M, et al. Incidence
and subtypes of early-onset dementia in a geographically defined
general population. Neurology. 2010;75(14):1249-1255. doi:10.1212/
WNL.0b013e3181f5d4c4
4. Rascovsky K, Hodges JR, Knopman D, et al. Sensitivity of revised
diagnostic criteria for the behavioural variant of frontotempo-
ral dementia. Brain. 2011;134(Pt 9):2456-2477. doi:10.1093/brain/
awr179
5. Gorno-Tempini ML, Hillis AE, Weintraub S, et al. Classifica-
tion of primary progressive aphasia and its variants. Neurology.
2011;76(11):1006-1014. doi:10.1212/WNL.0b013e31821103e6
6. Greaves CV, Rohrer JD. An update on genetic frontotemporal
dementia. JNeurol. 2019;266(8):2075-2086. doi:10.1007/s00415-
019-09363-4
7. Moore KM, Nicholas J, Grossman M, et al. Age at symptom
onset and death and disease duration in genetic frontotemporal
dementia: an international retrospective cohort study. Lancet Neurol.
2020;19(2):145-156. doi:10.1016/S1474-4422(19)30394-1
8. Neumann M, Mackenzie IRA. Review: neuropathology of non-tau
frontotemporal lobar degeneration. Neuropathol Appl Neurobiol.
2019;45(1):19-40. doi:10.1111/nan.12526
9. Kovacs GG. Invited review: neuropathology of tauopathies: princi-
ples and practice. Neuropathol Appl Neurobiol. 2015;41(1):3-23. doi:10.
1111/nan.12208
10. Bright F, Werry EL, Dobson-Stone C, et al. Neuroinflammation in
frontotemporal dementia. Nat Rev Neurol. 2019;15(9):540-555. doi:10.
1038/s41582-019-0231-z
11. Malpetti M, Cope TE, Street D, et al. Microglial activation in the frontal
cortex predicts cognitive decline in frontotemporal dementia. Brain.
2023;146(8):3221-3231. doi:10.1093/brain/awad078
12. Malpetti M, Rittman T, Jones PS, et al. In vivo PET imaging of
neuroinflammation in familial frontotemporal dementia. JNeurol
Neurosurg Psychiatry. 2021;92(3):319-322. doi:10.1136/jnnp-2020-
323698
13. Cagnin A, Rossor M, Sampson EL, Mackinnon T, Banati RB. In vivo
detection of microglial activation in frontotemporal dementia. Ann
Neurol. 2004;56(6):894-897. doi:10.1002/ana.20332
14. Kim MJ, McGwier M, Jenko KJ, et al. Neuroinflammation in frontotem-
poral lobar degeneration revealed by (11) C-PBR28 PET. Ann Clin
Tra ns l Neu ro l. 2019;6(7):1327-1331. doi:10.1002/acn3.50802
15. Woollacott IOC, Toomey CE, Strand C, et al. Microglial burden,
activation and dystrophy patterns in frontotemporal lobar degenera-
tion. J Neuroinflammation. 2020;17(1):234. doi:10.1186/s12974-020-
01907-0
16. Taipa R, Brochado P, Robinson A, et al. Patterns of microglial cell
activation in Alzheimer disease and frontotemporal lobar degen-
eration. Neurodegener Dis. 2017;17(4-5):145-154. doi:10.1159/
000457127
17. Keren-Shaul H, Spinrad A, Weiner A, et al. A unique microglia type
associated with restricting development of Alzheimer’s disease. Cell.
2017;169(7):1276-1290 e17. doi:10.1016/j.cell.2017.05.018
18. Krasemann S, Madore C, Cialic R, et al. The TREM2-APOE pathway
drives the transcriptional phenotype of dysfunctional microglia in neu-
rodegenerative diseases. Immunity. 2017;47(3):566-581 e9. doi:10.
1016/j.immuni.2017.08.008
19. Garcia-Revilla J, Boza-Serrano A, Espinosa-Oliva AM, et al. Galectin-
3, a rising star in modulating microglia activation under conditions
of neurodegeneration. Cell Death Dis. 2022;13(7):628. doi:10.1038/
s41419-022-05058-3
20. Burguillos MA, Svensson M, Schulte T, et al. Microglia-secreted
galectin-3 acts as a toll-like receptor 4 ligand and contributes to
microglial activation. Cell Rep. 2015;10(9):1626-1638. doi:10.1016/j.
celrep.2015.02.012
21. Boza-Serrano A, Ruiz R, Sanchez-Varo R, et al. Galectin-3, a novel
endogenous TREM2 ligand, detrimentally regulates inflammatory
response in Alzheimer’s disease. Acta Neuropathol. 2019;138(2):251-
273. doi:10.1007/s00401-019-02013-z
22. Garcia-Revilla J, Boza-Serrano A, Jin Y, et al. Galectin-3 shapes
toxic alpha-synuclein strains in Parkinson’s disease. Acta Neuropathol.
2023;146(1):51-75. doi:10.1007/s00401-023-02585-x
23. Boza-Serrano A, Reyes JF, Rey NL, et al. The role of Galectin-3 in alpha-
synuclein-induced microglial activation. Acta Neuropathol Commun.
2014;2:156. doi:10.1186/s40478-014-0156-0
24. Boza-Serrano A, Vrillon A, Minta K, et al. Galectin-3 is elevated in CSF
and is associated with Abeta deposits and tau aggregates in brain tis-
sue in Alzheimer’s disease. Acta Neuropathol. 2022;144(5):843-859.
doi:10.1007/s00401-022-02469-6
25. Johnson ECB, Dammer EB, Duong DM, et al. Large-scale proteomic
analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals
early changes in energy metabolism associated with microglia and
astrocyte activation. Nat Med. 2020;26(5):769-780. doi:10.1038/
s41591-020-0815-6
26. Yin F, Dumont M, Banerjee R, et al. Behavioral deficits and progressive
neuropathology in progranulin-deficient mice: a mouse model of fron-
totemporal dementia. FASEB J. 2010;24(12):4639-4647. doi:10.1096/
fj.10-161471
27. Yin F, Banerjee R, Thomas B, et al. Exaggerated inflammation, impaired
host defense, and neuropathology in progranulin-deficient mice. JExp
Med. 2010;207(1):117-128. doi:10.1084/jem.20091568
28. Yoshiyama Y, Higuchi M, Zhang B, et al. Synapse loss and microglial
activation precede tangles in a P301S tauopathy mouse model. Neuron.
2007;53(3):337-351. doi:10.1016/j.neuron.2007.01.010
29. Huang M, Modeste E, Dammer E, et al. Network analysis of
the progranulin-deficient mouse brain proteome reveals pathogenic
mechanisms shared in human frontotemporal dementia caused by
GRN mutations. Acta Neuropathol Commun. 2020;8(1):163. doi:10.
1186/s40478-020-01037-x
30. Zambusi A, Novoselc KT, Hutten S, et al. TDP-43 condensates and lipid
droplets regulate the reactivity of microglia and regeneration after
BORREGO–ÉCIJA ET AL.1525
traumatic brain injury. Nat Neurosci. 2022;25(12):1608-1625. doi:10.
1038/s41593-022-01199-y
31. Kersaitis C, Halliday GM, Kril JJ. Regional and cellular pathology in
frontotemporal dementia: relationship to stage of disease in cases
with and without Pick bodies. Acta Neuropathol. 2004;108(6):515-523.
doi:10.1007/s00401-004-0917-0
32. Arnold SE, Han LY, Clark CM, Grossman M, Trojanowski JQ. Quan-
titative neurohistological features of frontotemporal degeneration.
Neurobiol Aging. 2000;21(6):913-919. doi:10.1016/s0197-4580(00)
00173-1
33. Maya G, Sarto J, Compta Y, et al. Assessment of cognitive symptoms
in brain bank-registered control subjects: feasibility and utility of a
telephone-based screening. J Alzheimers Dis. 2022;85(3):1107-1113.
doi:10.3233/JAD-215444
34. Yazar T, Olgun Yazar H, Cihan M. Evaluation of serum galectin-3 levels
at Alzheimer patients by stages: a preliminary report. Acta Neurol Belg.
2021;121(4):949-954. doi:10.1007/s13760-020-01477-1
35. Schmitz M, Ebert E, Stoeck K, et al. Validation of 14-3-3 protein as a
marker in sporadic creutzfeldt-jakob disease diagnostic. Mol Neurobiol.
2016;53(4):2189-2199. doi:10.1007/s12035-015-9167-5
36. Gaetani L, Hoglund K, Parnetti L, et al. A new enzyme-linked
immunosorbent assay for neurofilament light in cerebrospinal fluid:
analytical validation and clinical evaluation. Alzheimers Res Ther.
2018;10(1):8. doi:10.1186/s13195-018-0339-1
37. Willemse EAJ, De Vos A, Herries EM, et al. Neurogranin as cere-
brospinal fluid biomarker for Alzheimer disease: an assay comparison
study. Clin Chem. 2018;64(6):927-937. doi:10.1373/clinchem.2017.
283028
38. Nordengen K, Kirsebom BE, Henjum K, et al. Glial activation and
inflammation along the Alzheimer’s disease continuum. J Neuroinflam-
mation. 2019;16(1):46. doi:10.1186/s12974-019- 1399-2
39. Liu FT, Rabinovich GA. Galectins: regulators of acute and chronic
inflammation. Ann N Y Acad Sci. 2010;1183:158-182. doi:10.1111/j.
1749-6632.2009.05131.x
40. Pesamaa I, Muller SA, Robinson S, et al. A microglial activity state
biomarker panel differentiates ftd-granulin and Alzheimer’s disease
patients from controls. bioRxiv. 2023;06(15):545187. doi:10.1101/
2023.06.15.545187. bioRxiv.
41. Starossom SC, Mascanfroni ID, Imitola J, et al. Galectin-1 deacti-
vates classically activated microglia and protects from inflammation-
induced neurodegeneration. Immunity. 2012;37(2):249-263. doi:10.
1016/j.immuni.2012.05.023
42. Steelman AJ, Li J. Astrocyte galectin-9 potentiates microglial TNF
secretion. J Neuroinflammation. 2014;11:144. doi:10.1186/s12974-
014-0144-0
43. Burman J, Svenningsson A. Cerebrospinal fluid concentration of
Galectin-9 is increased in secondary progressive multiple sclerosis. J
Neuroimmunol. 2016;292:40-44. doi:10.1016/j.jneuroim.2016.01.008
44. Premeaux TA, D’Antoni ML, Abdel-Mohsen M, et al. Elevated cere-
brospinal fluid Galectin-9 is associated with central nervous system
immune activation and poor cognitive performance in older HIV-
infected individuals. J Neurovirol. 2019;25(2):150-161. doi:10.1007/
s13365-018-0696-3
45. Guerreiro RJ, Lohmann E, Bras JM, et al. Using exome sequenc-
ing to reveal mutations in TREM2 presenting as a frontotemporal
dementia-like syndrome without bone involvement. JAMA Neurol.
2013;70(1):78-84. doi:10.1001/jamaneurol.2013.579
46. Rayaprolu S, Mullen B, Baker M, et al. TREM2 in neurodegeneration:
evidence for association of the p.R47H variant with frontotempo-
ral dementia and Parkinson’s disease. Mol Neurodegener. 2013;8:19.
doi:10.1186/1750-1326-8-19
47. Lill CM, Rengmark A, Pihlstrom L, et al. The role of TREM2 R47H as
a risk factor for Alzheimer’s disease, frontotemporal lobar degenera-
tion, amyotrophic lateral sclerosis, and Parkinson’s disease. Alzheimers
Dement. 2015;11(12):1407-1416. doi:10.1016/j.jalz.2014.12.009
48. Johnson AM, Lukens JR. The innate immune response in tauopathies.
Eur J Immunol. 2023;53(6):e2250266. doi:10.1002/eji.202250266
49. Lant SB, Robinson AC, Thompson JC, et al. Patterns of microglial
cell activation in frontotemporal lobar degeneration. Neuropathol Appl
Neurobiol. 2014;40(6):686-696. doi:10.1111/nan.12092
50. Borrego-Ecija S, Morgado J, Palencia-Madrid L, et al. Frontotemporal
dementia caused by the P301L mutation in the MAPT gene: clinico-
pathological features of 13 cases from the same geographical origin in
Barcelona, Spain. Dement Geriatr Cogn Disord. 2017;44(3-4):213-221.
doi:10.1159/000480077
51. Hartnell IJ, Blum D, Nicoll JAR, Dorothee G, Boche D. Glial cells and
adaptive immunity in frontotemporal dementia with tau pathology.
Brain. 2021;144(3):724-745. doi:10.1093/brain/awaa457
52. van Olst L, Verhaege D, Franssen M, et al. Microglial activation arises
after aggregation of phosphorylated-tau in a neuron-specific P301S
tauopathy mouse model. Neurobiol Aging. 2020;89:89-98. doi:10.
1016/j.neurobiolaging.2020.01.003
53. Karlsson A, Christenson K, Matlak M, et al. Galectin-3 functions as
an opsonin and enhances the macrophage clearance of apoptotic neu-
trophils. Glycobiology. 2009;19(1):16-20. doi:10.1093/glycob/cwn104
54. Karlsson M, Zhang C, Mear L, et al. A single-cell type transcriptomics
map of human tissues. Sci Adv. 2021;7(31):eabh2169. doi:10.1126/
sciadv.abh2169
55. Lo TH, Chen HL, Yao CI, et al. Galectin-3 promotes noncanonical
inflammasome activation through intracellular binding to lipopolysac-
charide glycans. Proc Natl Acad Sci U S A. 2021;118(30):e2026246118.
doi:10.1073/pnas.2026246118
56. Gao X, Liu J, Liu X, Li L, Zheng J. Cleavage and phosphorylation: impor-
tant post-translational modifications of galectin-3. Cancer Metastasis
Rev. 2017;36(2):367-374. doi:10.1007/s10555-017- 9666-0
57. Nabi IR, Shankar J, Dennis JW. The galectin lattice at a glance. JCellSci.
2015;128(13):2213-2219. doi:10.1242/jcs.151159
58. Hara A, Niwa M, Kanayama T, et al. Galectin-3: a potential prog-
nostic and diagnostic marker for heart disease and detection of
early stage pathology. Biomolecules. 2020;10(9):1277. doi:10.3390/
biom10091277
59. Bevan-Jones WR, Cope TE, Jones PS, et al. In vivo evidence for pre-
symptomatic neuroinflammation in a MAPT mutation carrier. Ann Clin
Tra ns l Neu ro l. 2019;6(2):373-378. doi:10.1002/acn3.683
60. Shi Y, Manis M, Long J, et al. Microglia drive APOE-dependent
neurodegeneration in a tauopathy mouse model. J Exp Med.
2019;216(11):2546-2561. doi:10.1084/jem.20190980
61. Antonell A, Tort-Merino A, Rios J, et al. Synaptic, axonal damage
and inflammatory cerebrospinal fluid biomarkers in neurodegenera-
tive dementias. Alzheimers Dement. 2020;16(2):262-272. doi:10.1016/
j.jalz.2019.09.001
62. Nilsson J, Cousins KAQ, Gobom J, et al. Cerebrospinal fluid biomarker
panel of synaptic dysfunction in Alzheimer’s disease and other neu-
rodegenerative disorders. Alzheimers Dement. 2023;19(5):1775-1784.
doi:10.1002/alz.12809
63. Das S, Goossens J, Jacobs D, et al. Synaptic biomarkers in the cere-
brospinal fluid associate differentially with classical neuronal biomark-
ers in patients with Alzheimer’s disease and frontotemporal dementia.
Alzheimers Res Ther. 2023;15(1):62. doi:10.1186/s13195-023- 01212-
x
64. Hong S, Beja-Glasser VF, Nfonoyim BM, et al. Complement and
microglia mediate early synapse loss in Alzheimer mouse models.
Science. 2016;352(6286):712-716. doi:10.1126/science.aad8373
65. Heller C, Foiani MS, Moore K, et al. Plasma glial fibrillary acidic pro-
tein is raised in progranulin-associated frontotemporal dementia. J
Neurol Neurosurg Psychiatry. 2020;91(3):263-270. doi:10.1136/jnnp-
2019-321954
66. Zhang J, Velmeshev D, Hashimoto K, et al. Neurotoxic microglia
promote TDP-43 proteinopathy in progranulin deficiency. Nature.
2020;588(7838):459-465. doi:10.1038/s41586-020-2709-7
1526 BORREGO–ÉCIJA ET AL.
67. Woollacott IOC, Swift IJ, Sogorb-Esteve A, et al. CSF glial markers are
elevated in a subset of patients with genetic frontotemporal demen-
tia. Ann Clin Transl Neurol. 2022;9(11):1764-1777. doi:10.1002/acn3.
51672
68. Sarto J, Ruiz-Garcia R, Guillen N, et al. Diagnostic performance and
clinical applicability of blood-based biomarkers in a prospective mem-
ory clinic cohort. Neurology. 2023;100(8):e860-e873. doi:10.1212/
WNL.0000000000201597
69. Illan-Gala I, Lleo A, Karydas A, et al. Plasma tau and neurofilament light
in frontotemporal lobar degeneration and Alzheimer disease. Neurol-
ogy. 2021;96(5):e671-e683. doi:10.1212/WNL.0000000000011226
70. van der Ende EL, Meeter LH, Poos JM, et al. Serum neurofilament light
chain in genetic frontotemporal dementia: a longitudinal, multicen-
tre cohort study. Lancet Neurol. 2019;18(12):1103-1111. doi:10.1016/
S1474-4422(19)30354-0
71. Meeter LHH, Steketee RME, Salkovic D, et al. Clinical value of cere-
brospinal fluid neurofilament light chain in semantic dementia. JNeurol
Neurosurg Psychiatry. 2019;90(9):997-1004. doi:10.1136/jnnp-2018-
319784
SUPPORTING INFORMATION
Additional supporting information can be found online in the Support-
ing Information section at the end of this article.
How to cite this article: Borrego–Écija S, Pérez-Millan A,
Antonell A, et al. Galectin-3 is upregulated in frontotemporal
dementia patients with subtype specificity. Alzheimer’s Dement.
2024;20:1515–1526. https://doi.org/10.1002/alz.13536
... In total, 15 biomarkers were measured in at least four studies, 14 in three studies, 55 in two studies, and 201 in only one study each. When comparing FTD patients with healthy controls (Table 1), the most substantial evidence for increased biomarker levels was found for glial fibrillary acidic protein (GFAP) in both blood [16][17][18][19][20][21][22][23][24][25] and CSF [21,[26][27][28][29]. Elevated CSF levels of YKL-40 (CHI3L1) [21,[29][30][31][32][33][34][35][36] and chitotriosidase-1 (CHIT1) [21,29,30,37] followed. Progranulin (PGRN) in blood showed the most evidence of decrease [38][39][40][41][42]. Additionally, four biomarkers-CCL19, CXCL1, CXCL6, and Somatostatin (SST)-were reported to have decreased levels in two studies each [43][44][45][46]. ...
... Immune markers such as GFAP, YKL-40, and CHIT1, which were consistently elevated in FTD compared to healthy controls, showed no significant differences across the major FTD phenotypes, suggesting their upregulation is a common feature of FTD. The only significantly altered biomarker was CSF Galectin-3, which was higher in bvFTD compared to svPPA and nfvPPA [32]. Galectin-3, predominantly expressed by microglia, plays a complex role in neuroinflammation, likely via the NF-KB pathway [150]. ...
... Galectin-3, predominantly expressed by microglia, plays a complex role in neuroinflammation, likely via the NF-KB pathway [150]. Interestingly, MAPT-FTD was the only genotype with significantly higher CSF Galectin-3 levels compared to C9ORF72and GRN-mediated FTD [32], and it also displayed the lowest CSF CHIT1 levels [30]. This suggests that in MAPT-FTD, microglial activity is more committed to Galectin-3-mediated phagocytosis and debris clearance, possibly driven by tau accumulation, over CHIT1-mediated inflammation and matrix remodeling. ...
Article
Full-text available
Dysregulated immune activation plays a key role in the pathogenesis of neurodegenerative diseases, including frontotemporal dementia (FTD). This study reviews immunological biomarkers associated with FTD and its subtypes. A systematic search of PubMed and Web of Science was conducted for studies published before 1 January 2025, focusing on immunological biomarkers in CSF or blood from FTD patients with comparisons to healthy or neurological controls. A total of 124 studies were included, involving 6686 FTD patients and 202 immune biomarkers. Key findings include elevated levels of GFAP and MCP1/CCL2 in both CSF and blood and consistently increased CHIT1 and YKL-40 in CSF. Complement proteins from the classical activation pathway emerged as promising targets. Distinct immune markers were found to differentiate FTD from Alzheimer’s disease (AD) and amyotrophic lateral sclerosis (ALS), with GFAP, SPARC, and SPP1 varying between FTD and AD and IL-15, HERV-K, NOD2, and CHIT1 differing between FTD and ALS. A few markers, such as Galectin-3 and PGRN, distinguished FTD subtypes. Enrichment analysis highlighted IL-10 signaling and immune cell chemotaxis as potential pathways for further exploration. This study provides an overview of immunological biomarkers in FTD, emphasizing those most relevant for future research on immune dysregulation in FTD pathogenesis.
... This study suggests that inhibition of the activity of this protein leads to a significant reduction in the inflammatory response [32]. In another study investigating the role of Gal-3, a microglial marker, in the neurodegenerative mechanism of frontotemporal dementia (FTD), high levels of Gal-3 were found in the cerebrospinal fluid and serum of both sporadic and genetic FTD patients [106]. Gal-3 has also been shown to be upregulated in the brains of Alzheimer's patients and 5xFAD (familial Alzheimer's disease) mice and is expressed explicitly in microglia associated with amyloid beta (Aβ) plaques [104,107]. ...
Article
Full-text available
Excitotoxicity caused by excessive concentration of the excitatory neurotransmitter glutamate causes neuronal cell death and promotes neurodegenerative disorders. The neuroexcitant neurotoxin kainic acid (KA) induces excitotoxicity, leading to neuronal death via oxidative stress and inflammation, and its experimental use is widespread. This study was designed to determine the protective effect of Troxerutin (TXR) and its relationship with Galectin-3 (Gal-3) in experimental excitotoxicity with neuroinflammation and oxidative stress. Fifty male Wistar rats were divided into five groups (n = 10): Control group rats received intraperitoneal (ip) normal saline for 6 days. Sham group rats received a single dose of intracerebroventricular (icv) normal saline on the first day. KA group rats were treated with a single dose of KA; icv-0.5 μg/μl). TXR group rats treated with TXR for 6 days: ip-100 mg/kg) and KA + TXR group rats treated with KA (single dose) and TXR (6 days). It was observed that malondialdehyde (MDA) and interleukin-1β (IL-1β) levels increased and reduced glutathione (GSH) levels decreased in the cerebral cortex of rats with KA neurotoxicity. TXR treatment caused a significant improvement in MDA and GSH levels and a significant decrease in IL-1β levels in rats with the excitotoxicity model. Gal-3 expressions in the hippocampus and cerebellum increased in KA-treated rats, whereas TXR treatment decreased Gal-3 expressions. In addition, histopathological changes caused by KA administration showed improvement in TXR-treated groups. In conclusion, the findings showed that TXR treatment attenuated KA-induced neurotoxicity by reducing oxidative tissue damage, inflammatory response and Gal-3 expression.
... A study conducted on patients with frontotemporal dementia found that increased Gal-3 levels were associated with cognitive decline in the disease. This finding suggests that Gal-3 may play a role in neuroinflammatory processes in patients with frontotemporal dementia and could be considered a biomarker and therapeutic target for this disease [37]. Another animal study investigated the role of Gal-3 in modulating anxiety levels. ...
Article
Full-text available
Objectives: This study aimed to assess serum Galectin-3 (Gal-3) and IL-6 levels, along with other inflammatory markers, in type-1 bipolar disorder (BD) patients and explore their relationship with clinical features, metabolic parameters, and symptom severity. Background: The study included 38 manic, 35 euthymic BD patients, and 40 healthy controls. Sociodemographic data, such as age, gender, alcohol and smoking habits, and body mass index (BMI), were recorded. Methods: The Young Mania Rating Scale (YMRS) and Hamilton Depression Rating Scale (HAM-D) were administered to patients. Biochemical measurements included Gal-3, IL-6, CRP, neutrophil, lymphocyte, platelet counts, and inflammatory indices like NLR, PLR, SII, and SIRI. Results: Gal-3 levels significantly differed among the groups (F = 52.251, p < 0.001), with the highest levels in euthymic patients. IL-6 levels were elevated in both manic and euthymic patients compared to controls (F = 7.379, p = 0.001). Manic patients had significantly higher levels of neutrophils, monocytes, CRP, NLR, PLR, SII, and SIRI. A positive correlation was found between Gal-3 levels, the total number of episodes, and YMRS scores in manic patients. In euthymic patients, Gal-3 levels correlated positively with disease duration and episode count. Conclusions: Elevated Gal-3 levels, particularly in the euthymic phase, may serve as a biomarker for BD and indicate ongoing inflammation. These findings suggest Gal-3 could help identify BD and differentiate the euthymic phase.
... Neuronal dysfunction/death, abnormal protein accumulation, and activation of the central immune system are the main factors in the pathological progression of FTD. Abnormal protein conformation and accumulation will activate the immune system, leading to neuroinflammation [49] . White matter hyperintensity usually reflects the abnormality of brain blood vessels and nerve tissues. ...
Preprint
Full-text available
Frontotemporal dementia (FTD) is a very common type of neurodegenerative dementia and is the main cause of dementia in people under 65 years old. It has strong heritability. To explore whether there are urinary proteins and biological pathways that are different between those who have not been identified with FTD under existing diagnostic methods and healthy people, this study conducts urinary proteomics analysis on people without disease with a family history of FTD and healthy people. The results show that there are 387 significantly different proteins (P<0.01; FC≥2.0 or ≤0.5) between the two groups. Among them, many proteins have been reported to play a role in FTD or the nervous system. In particular, Progranulin (PGRN) is considered an effective biomarker for FTD. Among the 139 biological pathways enriched by differential proteins (P<0.01), there are many pathways directly related to neurons and the nervous system, including glial cells and microglia. The differences in urinary proteomes between people without disease with a family history of FTD and healthy people make urinary proteomics have the potential to provide clues in exploring potential FTD patient populations and exploring the mechanisms related to the early occurrence and development of FTD, opening up a new perspective for FTD prevention and early diagnosis.
... FTD and ALS exhibit common molecular pathological features, including the mislocalization and aggregation of TAR DNA-binding protein 43 (TDP-43), a ribonucleotide protein that regulates mRNA metabolism, the accumulation of FTD/ALS-associated mutated proteins in inclusions, and the failure of the PQC system [173,176,177]. FTD/ ALS are also associated with alterations to the autophagy-lysosomal pathway, detectable in postmortem tissue of FTD/ALS patients [87,178] and evidenced by increased levels of galectin-3 in the spinal cord and cerebrospinal fluid, suggesting changes in lysosome dynamics [178,179]. ...
Article
Full-text available
Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs. Graphical Abstract
... Em amostras corticais humanas com DA e em camundongos, um estudo observou regulação positiva e expressão significativa da gal-3 em células microgliais posicionadas próximas a placas de agregados extracelulares de peptídeo beta amilóide (βA), resultados não observados no grupo controle17 . Num estudo com amostras de tecido cortical de pacientes com demência frontotemporal, foi observado regulação positiva da gal-3, aumento significativo de níveis séricos e no LCR da gal-3 e correlação com a proteína tau18 . A ativação microglial promove mecanismos pró-inflamatórios no SNC, como a produção de citocinas e estresse oxidativo, que contribuem com processos neurodegenerativos. ...
Article
Full-text available
Distúrbios neurológicos (DNs) têm crescido junto ao envelhecimento e aumento populacional global, sendo um desafio ao sistema de saúde pública. DNs envolvem disfunções sinápticas e moleculares que comprometem a cognição, memória e aprendizado. As galectinas (gal) são potenciais moduladoras da neuroinflamação, com expressão neurológica. Assim, tem-se como objetivo entender a associação entre as galectinas, vias moleculares e celulares na fisiopatologia de DNs. Realizou-se uma revisão narrativa nas bases de dados Pubmed, LILACS e Scielo, com as palavras-chave em inglês e português “galectins” and “cognition”. 23 estudos in vivo, in vitro e post-mortem foram incluídos nesta revisão. Na Doença de Alzheimer (DA), isquemia/reperfusão renal (IR), HIV, malária cerebral e esquizofrenia, diversos estudos mostraram relação entre expressão significativa aumentada ou reduzida da gal-3, gal-9, gal-8 e gal-1 e marcadores pró-inflamatórios e anti-inflamatórios, neurodegenerativos e sinápticos. Na DA, apneia obstrutiva do sono e IR, o silenciamento ou inibição farmacológica da gal-3 atenuou a neuroinflamação e apoptose neuronal, gerou proteção contra neurotoxicidade e melhorou o comprometimento cognitivo. Na isquemia a administração de gal-3 gerou remielinização e atenuação de déficits cognitivos. Concluiu-se que as galectinas –1, –3, -4, -8 e –9 modulam processos neuroinflamatórios e neurodegenerativos presentes em disfunções cognitivas de DNs, sendo potenciais alvos terapêuticos. Palavras-chave: galectinas; cognição; neurofisiologia; inflamação; mielina.
Article
Full-text available
Activation of the innate immune system following pattern recognition receptor binding has emerged as one of the major pathogenic mechanisms in neurodegenerative disease. Experimental, epidemiological, pathological, and genetic evidence underscores the meaning of innate immune activation during the prodromal as well as clinical phases of several neurodegenerative disorders including Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and frontotemporal dementia. Importantly, innate immune activation and the subsequent release of inflammatory mediators contribute mechanistically to other hallmarks of neurodegenerative diseases such as aberrant proteostatis, pathological protein aggregation, cytoskeleton abnormalities, altered energy homeostasis, RNA and DNA defects, and synaptic and network disbalance and ultimately to the induction of neuronal cell death. In this review, we discuss common mechanisms of innate immune activation in neurodegeneration, with particular emphasis on the pattern recognition receptors (PRRs) and other receptors involved in the detection of damage-associated molecular patterns (DAMPs).
Article
Full-text available
Background With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. Methods Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), genetically modified to yield the most opposite homeostatic ( TREM2- knockout) and disease-associated ( GRN -knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify proteomic changes in microglia and cerebrospinal fluid (CSF) of Grn - and Trem2 -knockout mice. Additionally, we analyzed the proteome of GRN - and TREM2 -knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort ( GRN mutation carriers versus non-carriers), as well as the proteomic data set available from the EMIF-AD MBD study. Results We identified proteomic changes between the opposite activation states in mouse microglia and CSF, as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of Alzheimer’s (AD) patients. Remarkably, each of these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. Conclusions The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the Dominantly Inherited Alzheimer’s Disease Network (DIAN) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders.
Preprint
Full-text available
Background With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. Methods Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), which were genetically modified to yield the most opposite homeostatic ( TREM2- knockout) and disease-associated ( GRN -knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify changes in microglial and cerebrospinal (CSF) proteome of Grn - and Trem2 -knockout mice. Additionally, we analyzed the proteome of GRN - and TREM2 -knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort with 11 GRN mutation carriers and 12 non-carriers, as well as the proteomic data set available from the European Medical Information Framework Alzheimer’s Disease Multimodal Biomarker Discovery (EMIF-AD MBD). Findings We identified proteomic changes between the opposite activation states in mouse microglia and cerebrospinal fluid (CSF), as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of AD patients. In AD, these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. Interpretation The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these marker proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the DIAN (Dominantly Inherited Alzheimer’s Disease Network) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders. Funding This work was supported by 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 CH, SFL and DP) and a Koselleck Project HA1737/16-1 (to CH).
Article
Full-text available
Parkinson’s Disease (PD) is a neurodegenerative and progressive disorder characterised by intracytoplasmic inclusions called Lewy bodies (LB) and degeneration of dopaminergic neurons in the substantia nigra (SN). Aggregated α-synuclein (αSYN) is known to be the main component of the LB. It has also been reported to interact with several proteins and organelles. Galectin-3 (GAL3) is known to have a detrimental function in neurodegenerative diseases. It is a galactose-binding protein without known catalytic activity and is expressed mainly by activated microglial cells in the central nervous system (CNS). GAL3 has been previously found in the outer layer of the LB in post-mortem brains. However, the role of GAL3 in PD is yet to be elucidated. In post-mortem samples, we identified an association between GAL3 and LB in all the PD subjects studied. GAL3 was linked to less αSYN in the LB outer layer and other αSYN deposits, including pale bodies. GAL3 was also associated with disrupted lysosomes. In vitro studies demonstrate that exogenous recombinant Gal3 is internalised by neuronal cell lines and primary neurons where it interacts with endogenous αSyn fibrils. In addition, aggregation experiments show that Gal3 affects spatial propagation and the stability of pre-formed αSyn fibrils resulting in short, amorphous toxic strains. To further investigate these observations in vivo, we take advantage of WT and Gal3KO mice subjected to intranigral injection of adenovirus overexpressing human αSyn as a PD model. In line with our in vitro studies, under these conditions, genetic deletion of GAL3 leads to increased intracellular αSyn accumulation within dopaminergic neurons and remarkably preserved dopaminergic integrity and motor function. Overall, our data suggest a prominent role for GAL3 in the aggregation process of αSYN and LB formation, leading to the production of short species to the detriment of larger strains which triggers neuronal degeneration in a mouse model of PD.
Article
Full-text available
Background Loss of synaptic functionality has been recently identified as an early-stage indicator of neurological diseases. Consequently, monitoring changes in synaptic protein levels may be relevant for observing disease evolution or treatment responses in patients. Here, we have studied the relationship between fluid biomarkers of neurodegeneration and synaptic dysfunction in patients with Alzheimer’s disease (AD), frontotemporal dementia (FTD), and subjective cognitive decline (SCD). Methods The exploratory cohort consisted of cerebrospinal fluid (CSF) samples (n = 60) from patients diagnosed with AD (n = 20), FTD (n = 20), and SCD (n = 20) from the Amsterdam Dementia Cohort. We developed two novel immunoassays for the synaptic proteins synaptosomal-associated protein-25 (SNAP25) and vesicle-associated membrane protein-2 (VAMP2). We measured the levels of these biomarkers in CSF, in addition to neuronal pentraxin-2 (NPTX2), glutamate ionotropic receptor-4 (GluR4), and neurogranin (Ng) for this cohort. All in-house immunoassays were validated and analytically qualified prior to clinical application. CSF neurogranin (Ng) was measured using a commercially available ELISA. Results This pilot study indicated that SNAP25, VAMP2, and Ng may not be specific biomarkers for AD as their levels were significantly elevated in patients with both AD and FTD compared to SCD. Moreover, the strength of the correlations between synaptic proteins was lower in the AD and FTD clinical groups compared to SCD. SNAP25, VAMP2, and Ng correlated strongly with each other as well as with total Tau (Tau) and phosphorylated Tau (PTau) in all three clinical groups. However, this correlation was weakened or absent with NPTX2 and GluR4. None of the synaptic proteins correlated to neurofilament light (NfL) in any clinical group. Conclusion The correlation of the synaptic biomarkers with CSF Tau and PTau but the lack thereof with NfL implies that distinct pathological pathways may be involved in synaptic versus axonal degeneration. Our results reflect the diversity of synaptic pathology in neurodegenerative dementias.
Article
Full-text available
Tauopathies, which include frontotemporal dementia, Alzheimer's disease, and chronic traumatic encephalopathy, are a class of neurological disorders resulting from pathogenic tau aggregates. These aggregates disrupt neuronal health and function leading to the cognitive and physical decline of tauopathy patients. Genome‐wide association studies and clinical evidence have brought to light the large role of the immune system in inducing and driving tau‐mediated pathology. More specifically, innate immune genes are found to harbor tauopathy risk alleles, and innate immune pathways are upregulated throughout the course of disease. Experimental evidence has expanded on these findings by describing pivotal roles for the innate immune system in the regulation of tau kinases and tau aggregates. In this review, we summarize the literature implicating innate immune pathways as drivers of tauopathy.
Article
Full-text available
Frontotemporal dementia is clinically and neuropathologically heterogeneous, but neuroinflammation, atrophy and cognitive impairment occur in all of its principal syndromes. Across the clinical spectrum of frontotemporal dementia, we assess the predictive value of in vivo neuroimaging measures of microglial activation and grey-matter volume on the rate of future cognitive decline. We hypothesized that inflammation is detrimental to cognitive performance, in addition to the effect of atrophy. Thirty patients with a clinical diagnosis of frontotemporal dementia underwent a baseline multimodal imaging assessment, including [11C]PK11195 PET to index microglial activation and structural MRI to quantify grey-matter volume. Ten people had behavioural variant frontotemporal dementia, 10 had the semantic variant of primary progressive aphasia and 10 had the non-fluent agrammatic variant of primary progressive aphasia. Cognition was assessed at baseline and longitudinally with the revised Addenbrooke's Cognitive Examination, at an average of 7-month intervals (for an average of ∼2 years, up to ∼5 years). Regional [11C]PK11195 binding potential and grey-matter volume were determined, and these were averaged within four hypothesis-driven regions of interest: bilateral frontal and temporal lobes. Linear mixed-effect models were applied to the longitudinal cognitive test scores, with [11C]PK11195 binding potentials and grey-matter volumes as predictors of cognitive performance, with age, education and baseline cognitive performance as covariates. Faster cognitive decline was associated with reduced baseline grey-matter volume and increased microglial activation in frontal regions, bilaterally. In frontal regions, microglial activation and grey-matter volume were negatively correlated, but provided independent information, with inflammation the stronger predictor of the rate of cognitive decline. When clinical diagnosis was included as a factor in the models, a significant predictive effect was found for [11C]PK11195 BPND in the left frontal lobe (−0.70, P = 0.01), but not for grey-matter volumes (P > 0.05), suggesting that inflammation severity in this region relates to cognitive decline regardless of clinical variant. The main results were validated by two-step prediction frequentist and Bayesian estimation of correlations, showing significant associations between the estimated rate of cognitive change (slope) and baseline microglial activation in the frontal lobe. These findings support preclinical models in which neuroinflammation (by microglial activation) accelerates the neurodegenerative disease trajectory. We highlight the potential for immunomodulatory treatment strategies in frontotemporal dementia, in which measures of microglial activation may also improve stratification for clinical trials.
Article
Full-text available
Decreasing the activation of pathology-activated microglia is crucial to prevent chronic inflammation and tissue scarring. In this study, we used a stab wound injury model in zebrafish and identified an injury-induced microglial state characterized by the accumulation of lipid droplets and TAR DNA-binding protein of 43 kDa (TDP-43)⁺ condensates. Granulin-mediated clearance of both lipid droplets and TDP-43⁺ condensates was necessary and sufficient to promote the return of microglia back to the basal state and achieve scarless regeneration. Moreover, in postmortem cortical brain tissues from patients with traumatic brain injury, the extent of microglial activation correlated with the accumulation of lipid droplets and TDP-43⁺ condensates. Together, our results reveal a mechanism required for restoring microglia to a nonactivated state after injury, which has potential for new therapeutic applications in humans.
Article
Full-text available
Introduction: Synaptic degeneration is a key part of the pathophysiology of neurodegenerative diseases, and biomarkers reflecting the pathological alterations are greatly needed. Method: Seventeen synaptic proteins were quantified in a pathology-confirmed cerebrospinal fluid cohort of patients with Alzheimer's disease (AD; n = 63), frontotemporal lobar degeneration (FTLD; n = 53), and Lewy body spectrum of disorders (LBD; n = 21), as well as healthy controls (HC; n = 48). Results: Comparisons revealed four distinct patterns: markers decreased across all neurodegenerative conditions compared to HC (the neuronal pentraxins), markers increased across all neurodegenerative conditions (14-3-3 zeta/delta), markers selectively increased in AD compared to other neurodegenerative conditions (neurogranin and beta-synuclein), and markers selectively decreased in LBD and FTLD compared to HC and AD (AP2B1 and syntaxin-1B). Discussion: Several of the synaptic proteins may serve as biomarkers for synaptic dysfunction in AD, LBD, and FTLD. Additionally, differential patterns of synaptic protein alterations seem to be present across neurodegenerative diseases. Highlights: A panel of synaptic proteins were quantified in the cerebrospinal fluid using mass spectrometry. We compared Alzheimer's disease, frontotemporal degeneration, and Lewy body spectrum of disorders. Pathology was confirmed by autopsy or familial mutations. We discovered synaptic biomarkers for synaptic degeneration and cognitive decline. We found differential patterns of synaptic proteins across neurodegenerative diseases.
Article
Full-text available
Background: Neuroinflammation has been shown to be an important pathophysiological disease mechanism in frontotemporal dementia (FTD). This includes activation of microglia, a process that can be measured in life through assaying different glia-derived biomarkers in cerebrospinal fluid. However, only a few studies so far have taken place in FTD, and even fewer focusing on the genetic forms of FTD. Methods: We investigated the cerebrospinal fluid concentrations of TREM2, YKL-40 and chitotriosidase using immunoassays in 183 participants from the Genetic FTD Initiative (GENFI) study: 49 C9orf72 (36 presymptomatic, 13 symptomatic), 49 GRN (37 presymptomatic, 12 symptomatic) and 23 MAPT (16 presymptomatic, 7 symptomatic) mutation carriers and 62 mutation-negative controls. Concentrations were compared between groups using a linear regression model adjusting for age and sex, with 95% bias-corrected bootstrapped confidence intervals. Concentrations in each group were correlated with the Mini-Mental State Examination (MMSE) score using non-parametric partial correlations adjusting for age. Age-adjusted z-scores were also created for the concentration of markers in each participant, investigating how many had a value above the 95th percentile of controls. Results: Only chitotriosidase in symptomatic GRN mutation carriers had a concentration significantly higher than controls. No group had higher TREM2 or YKL-40 concentrations than controls after adjusting for age and sex. There was a significant negative correlation of chitotriosidase concentration with MMSE in presymptomatic GRN mutation carriers. In the symptomatic groups, for TREM2 31% of C9orf72, 25% of GRN, and 14% of MAPT mutation carriers had a concentration above the 95th percentile of controls. For YKL-40 this was 8% C9orf72, 8% GRN and 0% MAPT mutation carriers, whilst for chitotriosidase it was 23% C9orf72, 50% GRN, and 29% MAPT mutation carriers. Conclusions: Although chitotriosidase concentrations in GRN mutation carriers were the only significantly raised glia-derived biomarker as a group, a subset of mutation carriers in all three groups, particularly for chitotriosidase and TREM2, had elevated concentrations. Further work is required to understand the variability in concentrations and the extent of neuroinflammation across the genetic forms of FTD. However, the current findings suggest limited utility of these measures in forthcoming trials.
Article
Background and Objectives Blood-based biomarkers have emerged as minimally-invasive options for evaluating cognitive impairment. Most studies to date have assessed them in research cohorts, limiting their generalization to everyday clinical practice. We evaluated their diagnostic performance and clinical applicability in a prospective, real-world, memory clinic cohort. Methods All patients referred with suspected cognitive impairment between July 2019 and June 2021, were prospectively invited to participate. Five plasma biomarkers (p-tau181, GFAP, NfL, t-tau, UCH-L1) were determined with SiMoA. Performance was assessed in comparison to clinical diagnosis (blinded to plasma results) and amyloid status (CSF/PET). A group of cognitively unimpaired (CU) controls was also included. Results Three hundred forty-nine participants (mean age 68, SD 8.3 years) and 36 CU controls (mean age 61.7, SD 8.2 years) were included. In the sub-cohort with available AD biomarkers (n=268), plasma p-tau181 and GFAP had a high diagnostic accuracy to differentiate AD from non-neurodegenerative causes (AUC 0.94 and 0.92, respectively), with p-tau181 systematically outperforming GFAP. Plasma p-tau181 levels predicted amyloid status (85% sensitivity and specificity) with accurate individual prediction in approximately 60% of the subjects. Plasma NfL differentiated frontotemporal dementia syndromes (FTD) from CU (0.90) and non-neurodegenerative causes (0.93), while the discriminative capacity with AD and between all neurodegenerative and non-neurodegenerative causes was less accurate. A combination of p-tau181 and NfL identified FTD with 82% sensitivity and 85% specificity and had a negative predictive value for neurodegenerative diagnosis of 86%, ruling out half of the non-neurodegenerative diagnoses. In the sub-cohort without AD biomarkers similar results were obtained. T-tau and UCH-L1 did not offer added diagnostic value. Discussion Plasma p-tau181 predicted amyloid status with high accuracy and could have potentially avoided CSF/amyloid PET testing in approximately 60% of subjects in a memory-clinic setting. NfL was useful for identifying FTD from non-neurodegenerative causes but behaved worse than p-tau181 in all other comparisons. Combining p-tau181 and NfL improved diagnostic performance for FTD and non-neurodegenerative diagnoses. However, the 14% false-negative results suggest that further improvement is needed before implementation outside memory clinics. Classification of Evidence This study provides Class I evidence that plasma p-tau181 correlates with the presence or absence of AD and a combination of plasma p-tau181 and NfL correlate moderately well with a diagnosis of FTD.