Introduction
Tissue-resident macrophages, including microglia, play a significant role in shaping brain development and connectivity as well as maintaining normal brain function [
8,
24,
49,
67,
80,
86,
100,
111,
119]. Microglia in particular have been implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease (AD) and multiple sclerosis (MS) [
70,
89,
94,
120]. Strikingly, genetic variants in several microglia-specific genes cause ‘microgliopathies’ [
94], a subgroup of leukodystrophies, which are defined as central nervous system (CNS) disorders primarily involving brain white matter. In both neurodegenerative diseases and microgliopathies, microglia increasingly gain interest as potential therapeutic targets, as they can be repopulated using hematopoietic stem cell transplantation (HSCT) or could be pharmacologically depleted by Colony-stimulating factor 1 receptor (CSF1R) inhibitors [
17,
106,
107,
123]. It is, therefore, critical to establish a better understanding of microglial function and the consequences of microglial depletion for the human brain.
CSF1R is a protein primarily located on mononuclear phagocytes and considered a key regulator of macrophage and microglial biology [
50,
110]. Bi-allelic variants in
CSF1R can cause a severe, but variable, developmental disorder termed BANDDOS (brain abnormalities, neurodegeneration, and dysosteosclerosis) [
40,
55,
74,
81,
114]. The few patients described presented with dysosteosclerosis, severe white matter lesions and other congenital brain abnormalities, leading to early death [
40,
74,
81]. Analysis of post-mortem brain tissue of a patient with a homozygous variant causing disruption of a splice acceptor site showed brain calcifications, axonal spheroids and an almost complete absence of microglia [
81]. Similarly, loss of CSF1R in rats, mice and zebrafish causes largely halted macrophage/microglia development, resulting in skeletal and CNS abnormalities [
29,
34,
62,
91]. Heterozygous missense variants located in the tyrosine kinase domains (TKDs) of CSF1R have been found to cause the more prevalent adult-onset leukodystrophy, termed ALSP (adult-onset leukoencephalopathy with axonal spheroids and pigmented glia) [
95]. Hallmarks of ALSP are motor and cognitive functional decline, often leading to death within several years after disease onset, and neuropathologically progressive white matter lesions, axonal spheroids, pigmented glia and cerebral calcifications [
1,
11,
121]. Recent evidence showed that heterozygous
CSF1R variants may also be associated with lower density of microglia in ALSP patients [
83,
85,
113]. HSCT, which may act by supplying bone-marrow-derived cells that repopulate the microglial niche, was suggested for treatment of ALSP. In mice, HSCT is indeed capable of repopulating the microglial niche, but only after depletion of endogenous microglia [
9]. More recently, it was shown that HSCT can halt disease progression in ALSP patients and even reduce pathology visible on MRI [
39,
73]. It is possible that the lack of microglia in ALSP facilitates their re-introduction by HSCT. Nevertheless, it remains not fully established whether microglial depletion is the initial pathological event in ALSP, which is important in understanding the mechanisms underlying the highly beneficial effects of HSCT.
Intriguingly, parents of patients with bi-allelic variants, even those with complete loss of function, have not been noted to develop ALSP or neurological dysfunction [
40,
55,
74,
81,
114]. While parents of patients with bi-allelic variants mostly have frameshift variants leading to nonsense-mediated mRNA decay (NMD), most ALSP-causing CSF1R variants are missense and located in the TKDs. This is consistent with a model where missense variants act dominantly and, at the biochemical level, cause dysfunctional heterodimers of wild-type and mutant CSF1R, which fail to show tyrosine kinase activity and lead to a ~ 75% suppression of CSF1R activity [5
0,92]. This is also consistent with findings in haploinsufficient
Csf1r-mutant mice where microglia were not reduced [
7,
19]. However, support for a dominant effect of ALSP-causing missense variants in vivo is lacking. Gaining insight into how these variants affect microglia, and their subsequent effect on brain health, is crucial to establish and refine treatment strategies for ALSP while also aiding in the development of microglia-focused therapies for other CNS diseases.
Here, we investigated the effects of ALSP-causing
CSF1R variants on microglia and pathology correlating with microglial depletion, which could precede white matter degeneration. We integrated neuropathological and multi-omic analysis on post-mortem brain tissue of ALSP patients with in vivo applications in genetic zebrafish models. Zebrafish have been used extensively to study developmental genetics and function of microglia, as they show highly conserved basic properties including CSF1R function [
45,
46,
62,
67,
81,
82,
88,
104,
105]. We provide evidence for microglial depletion both in ALSP post-mortem tissue and in vivo in zebrafish with pathogenic
CSF1R variants. Furthermore, we identified an unexpected astrocytic phenotype correlating with microglial depletion both in zebrafish models and in ALSP patients. Altogether, our data show that microglial depletion caused by dominant
CSF1R variants is a key hallmark and initial pathological event in ALSP. This further supports why HSCT in ALSP patients would be beneficial, possibly by repopulation of the microglial niche.
Human donor examination
Immunohistochemistry (IHC) human brain tissue
For neuropathological examination of axonal spheroids, myelin, microglia and astrocyte, paraffin-embedded post-mortem brain tissue sections were cut (4 µm) using a microtome. In short, paraffin sections were deparaffinized and rehydrated to distilled water. Antigens were retrieved by heating sections in sodium citrate buffer (10 mM; pH 6.0). For horseradish peroxidase (HRP) staining, endogenous peroxidase was quenched in 0.2% H2O2/0.125 sodiumazide in 1 × phosphate-buffered saline (PBS) for a maximum of 30 min at room temperature (RT). Sections were washed in blocking buffer (PBS, 0.5% protifar (Nutricia), 0.15% glycine (Sigma-Aldrich, St. Louis, USA), 0.4% Tween20). Then, sections were incubated overnight at 4 °C with blocking buffer containing primary anti-bodies: rabbit anti-TMEM119 (Atlas, HPA051870; 1:500), rabbit anti-P2RY12 (Anaspec. AS55042a; 1:500), rabbit anti-ALDH1L1 (Sigma, HPA031332, 1:500), rabbit anti-GFAP (Sigma, G9269; 1:100), mouse anti-S100β (Abcam, AB218513; 1:100), rabbit anti-MBP (Sigma, M3821; 1:100), rabbit anti-GSTM1 (Proteintech, 12412-1-AP; 1:100) and rabbit anti-LAMP1 (Abcam, AB218513; 1:200). After washing with blocking buffer, sections were incubated with secondary anti-bodies (1:200): DyLight alexa 488 anti-rabbit (ThermoFisher, AB_2313584); DyLight alexa 488 anti-mouse (ThermoFisher, AB_2340846); DyLight alexa 647 anti-rabbit (ThermoFisher, AB_2492288), DyLight alexa 647 anti-mouse (ThermoFisher, AB_2340862) for 1 h at RT. Autofluorescence and background staining was blocked by incubating sections in Sudan Black solution for 3 min at RT. Sections were embedded in ProLong™ Gold Antifade Mountant with DAPI (Invitrogen, P36931) to visualize nuclei. For HRP staining, sections were incubated with goat anti-mouse/rabbit-HRP (ImmunoLogic, DPVO55HRP) for 1 h at RT, and subsequently developed with 3,3'-diaminobenzidine (DAB; 0.05 mg/ml, Sigma-Aldrich). After washing in distilled water, slides were counterstained with hematoxylin and eosin (HE) to visualize nuclei, dehydrated and embedded in Entellan (Merck).
RNA sequencing
Total RNA was isolated from fresh–frozen occipital gyrus (mixed white matter and grey matter) of late-ALSP patient 1 and 2 (in triplicate) and 2 age- and sex-matched controls (in triplicate). Briefly, total RNA was isolated using TRIzol™ Reagent (ThermoFisher Scientific), 200 ng of total RNA was purified using poly-T oligo-attached magnetic beads to end up with poly-A containing mRNA. RIN values ranged from 5.5 to 6.5 for control and ALSP patient 1 RNA, and the RIN value was 7.9 for ALSP patient 2 RNA. The poly-A-tailed mRNA was fragmented and cDNA was synthesized using SuperScript II and random primers in the presence of Actinomycin D. cDNA fragments were end repaired, purified with AMPure XP beads, and A-tailed using Klenow exo-enzyme in the presence of dATP. Paired end adapters with dual index (Illumina) were ligated to the A-tailed cDNA fragments and purified using AMPure XP beads. The resulting adaptor-modified cDNA fragments were enriched by PCR using Phusion polymerase as followed: 30 s at 98 °C, 15 cycles of (10 s at 98 °C, 30 s at 60 °C, 30 s at 72 °C), 5 min at 72 °C. PCR products were purified using AMPure XP beads and eluted in 30 ml of resuspension buffer. One microliter was loaded on an Agilent Technologies 2100 Bioanalyzer using a DNA 1000 assay to determine the library concentration and for quality check.
Bridge amplification, sequencing by synthesis and data analysis Cluster generation was performed according to the Illumina TruSeq SR Rapid Cluster kit v2 (cBot) Reagents Preparation Guide (
www.illumina.com). Briefly, 12 RNA-seq libraries were pooled together to get a stock of 10 nM. One microliter of the 10 nM stock was denaturated with NaOH, diluted to 6 pM and hybridized onto the flowcell. The hybridized products were sequentially amplified, linearized, and end-blocked according to the Illumina Single Read Multiplex Sequencing user guide. After hybridization of the sequencing primer, sequencing-by-synthesis was performed using the HiSeq 2500 with a single read 50-cycle protocol followed by dual index sequencing. Reads were aligned against the GRCh38 genome using HiSat2 (version 2.0.4) [
53]. Counts were generated for each gene from the Ensembl (version 85) transcriptome analysis of GRCh38, using htseq-count (version 0.6.1) [
5].
Differential gene expression analysis was performed using the Bioconductor package edgeR [
98] and biomaRt [
31,
32]. Differentially expressed genes were selected based on the following thresholds: log fold change (logFC) <1.5, FDR false-discovery rate (FDR) < 0.05 (Suppl. Table 3, Online Resources). Pathway analysis was performed using the Bioconductor package topGO (Suppl. Table 4, Online Resources). For further analysis, the DEG were compared to published gene sets to find overlapping genes, including human microglia [
38], extracellular matrix-related genes [
77], mouse and human oligodendrocyte-related genes [
64,
66], MS microglia [
122] and AD microglia [
108] (Suppl. Table 6, Online Resources).
Mass spectrometry
Protein lysates were obtained from fresh–frozen occipital gyrus (mixed white matter and grey matter) from late-ALSP patient 1 and 2 (1 in triplicate, 1 in duplicate) and 2 age- and sex-matched controls (1 in triplicate, 1 in duplicate). The brain tissue was cut and lysed in 1 ml 50 mM Tris/HCl pH 8.2, 0.5% sodium deoxycholate (SDC) and MS-SAFE™ protease and phosphatase inhibitor using a Bioruptor ultasonicator (Diagenode). Protein concentrations were measured using the BCA assay (Thermo Scientific). Lysates were reduced with 5 mM DTT and cysteine residues were alkylated with 10 mM iodoacetamide. Protein was extracted by acetone precipitation at − 20 °C overnight. Samples were centrifuged at 8000 g for 10 min at 4 °C. The acetone was removed and the pellet allowed to dry. The protein pellet (~ 4 mg protein) was dissolved in 1 ml 50 mM Tris/HCl pH 8.2, 0.5% SDC and proteins were digested with LysC (1:200 enzyme:protein ratio) for 4 h at 37 °C. Next, trypsin was added (1:100 enzyme:protein ratio) and the digestion proceeded overnight at 30 °C. Digests were acidified with 50 μl 10% formic acid (FA) and centrifuged at 8000 g for 10 min at 4 °C to remove the precipitated SDC. The supernatant was transferred to a new centrifuge tube. The digests were purified with C18 solid phase extraction (Sep-Pak, Waters), lyophilized and stored at − 20 °C.
Isobaric labeling of the enriched peptides was performed using the 10-plex tandem mass tag (TMT) reagents (Thermo Fisher Scientific) with some modifications to the method of Böhm et al. [
13]. Peptides were loaded onto 20 mg C18 cartridges prepared in-house. The C18 cartridges were washed once with 1 ml 0.1% TFA and two times with 1 ml of 50 mM KH2PO4 (pH 4.5). TMT reagents (0.8 mg) were dissolved in 10 μl of dry ACN and diluted with 200 μl 50 mM KH2PO4. This TMT solution was immediately loaded onto the column and labeling on column proceeded for 1 h at RT. Each of the 9 samples was labeled with a different TMT tag. After labeling columns were washed twice with 1 ml 2% ACN/0.2% FA and the labeled peptides eluted with 1 ml 50% ACN. TMT-labeled samples were pooled and lyophilized.
TMT-labeled peptides were subjected to offline orthogonal high-pH reverse phase fractionation. TMT-labeled peptides were solubilized in 0.1% TFA and loaded onto a 20 mg PLRP-S cartridge made in-house. Cartridges were washed once with 1 ml 0.1% TFA and three times with 1 ml milliQ water. Peptides were eluted step-wise from column with 5%, 10%, 15% and 50% ACN/10 mM ammonium formate (pH 10). The 4 fractions were dried by vacuum centrifugation and each fraction was reconstituted with 2% ACN/0.2% FA for nLC-MS/MS analysis.
Mass spectra were acquired on an Orbitrap Lumos (Thermo) coupled to an EASY-nLC 1200 system (Thermo). Peptides were separated on an in-house packed 75 μm inner diameter column containing 50 cm Waters CSH130 resin (3.5 μm, 130 Å, Waters) with a gradient consisting of 2–20% (ACN, 0.1% FA) over 200 min at 300 nl/min. The column was kept at 50 °C in a NanoLC oven—MPI design (MS Wil GmbH). For all experiments, the instrument was operated in the data-dependent acquisition (DDA) mode. MS1 spectra were collected at a resolution of 120,000, with an automated gain control (AGC) target of 2E5 and a max injection time of 50 ms. The most intense ions were selected for MS/MS, top speed method 3-s cycle time. Precursors were filtered according to charge state (2–7), and monoisotopic peak assignment. Previously interrogated precursors were dynamically excluded for 70 s. Peptide precursors were isolated with a quadrupole mass filter set to a width of 1.2 Th. When applying the MS3 method, ion trap MS2 spectra were collected at an AGC of 5E4, max injection time of 50 ms and CID collision energy of 35%. For Orbitrap MS3 spectra, the operation resolution was 60,000, with an AGC setting of 1E5 and a max injection time of 120 ms. The HCD collision energy was set to 65% to ensure maximal TMT reporter ion yield. Synchronous precursor selection (SPS) was enabled at all times to include up to 5 MS2 fragment ions in the MS3 scan.
Raw mass spectrometry data were analyzed with the MaxQuant software suite ([
22]; version 1.6.4.0) as described previously [
102]. A false-discovery rate of 0.01 for proteins and peptides and a minimum peptide length of 7 amino acids were set. TMT tags on peptide
N-termini and lysine residues (+ 229.162932 Da) and carbamidomethylation of cysteine residues (+ 57.02146 Da) were set as static modifications, whereas methionine oxidation (+ 15.99492 Da) was set as variable modification. The Andromeda search engine was used to search the MS/MS spectra against the Uniprot database (taxonomy:
Homo sapiens, release January 2019) concatenated with the reversed versions of all sequences. The enzyme specificity was set to trypsin and a maximum of two missed cleavages was allowed. The FDR for both peptides and proteins was set to 0.01. The peptide tolerance was set to 10 ppm, the fragment ion tolerance was set to 0.6 Da for CID spectra and 20 ppm for MS3 reporter ion spectra. MaxQuant automatically quantified peptides based on the ‘reporter ion MS3 setting’. Before further statistical analysis, known contaminants and reverse hits were removed. Reporter ion intensities were adjusted to correct for the isotopic impurities of the different TMT reagents (according to the manufacturer’s specifications). For further analysis, we used the R packages vsn and limma [
48,
97].
Differentially expressed proteins were selected based on the following thresholds: logFC <1, FDR < 0.05 (Suppl. Table 5). For further analysis, the ALSP proteomics data set was compared to published gene sets to find overlapping proteins, including human microglia [
38] and astrocytic genes [
126], reactive astrocyte-associated genes [
35,
126] and extracellular matrix-related genes [
77] (Suppl. Table 6). For the comparison to published gene sets, the thresholds for LogFC was set to < − 0.3 and > 0.3, with FDR < 0.05 to not exclude less expressed proteins and pick up subtle changes.
Image acquisition
Confocal imaging was performed using a Leica SP5 intravital imaging setup with a 40x/1.3 NA oil objective for stained sections imaging, with 405, 488 and 633 nm lasers. Z‐stack images (z step size 0.5–1 µm) were acquired for all experiments. Brightfield images of the HRP stained sections were obtained using an Olympus DP72 light microscope or a Leica DM600B microscope (Leica microsystems). Sections were placed under the microscope and the experimenter determined whether the region visible was in grey or white matter. If no artifacts and/or tissue damage were present, the image was acquired on that location (5–10 images per region). We recorded representative images of the microglia population, and hence, microglial clusters were included in the quantification (1–2 images), since these clusters are typical for ALSP neuropathology.
Quantification and statistical analysis
Images were processed and quantified using the Fiji image processing package [
101]. The number of microglia and astrocytes were counted blindly and manually with an even ROI counting surface between ALSP and controls. Morphology of astrocytes was analyzed using the Sholl plugin in ImageJ [
36]. Statistical significance of the Sholl analysis was determined by one- or two-way ANOVA based on the mean area under the curve (AUC). Intracellular LAMP1 staining quantification was done as follows: based on the astrocyte marker channel, an ROI was generated using a standardized threshold to analyze LAMP1 + puncta within single cells. This was overlaid in the corresponding LAMP1 channel, and only the astrocyte ROI was kept using the Clear Outside tool. Intracellular particles were then analyzed on a binary image (thresholded) with the Analyze Particles tool. Statistical analysis was performed using GraphPad Prism 8, including (nested) one-way ANOVA with Bonferroni multiple testing correction. Data are presented as mean ± SD, as indicated. A p value < 0.05 was considered significant.
Discussion
Here, we establish by independent approaches that microglial depletion due to dominant-acting CSF1R missense variants is an early hallmark of ALSP. Furthermore, by multi-omic analyses in post-mortem ALSP brain tissue and in vivo experimentation in zebrafish mutants, we explored putative consequences of microglial depletion, identifying an altered astrocytic phenotype characterized by increased endocytosis. We also demonstrate that in zebrafish heterozygous pathogenic CSF1R missense variants, but not a heterozygous null allele, result in microglial depletion already in embryonic brain development. This provides in vivo mechanistic evidence that such variants act dominantly, resulting in microglial depletion that may far precede onset of symptoms.
Previously, we found that there is a lower density of IBA1 + microglia in ALSP cortical tissue [8
3]. Our current analyses of brain tissue of late- and intermediate-stage ALSP extend these findings and show overall reduced homeostatic P2RY12 + and TMEM119 + microglia, particularly in white matter. Consistently, transcriptomic analyses of relatively spared occipital gyrus revealed downregulation of microglia-specific genes. Furthermore, our findings from two ALSP-causing CSF1R missense variants in genome-edited zebrafish models showed microglial depletion in early development. These findings provide in vivo evidence that microglial depletion may far precede onset of symptoms. Only in intermediate-stage patients, numbers of TMEM119 + microglia did not differ from controls in grey matter, in contrast to P2RY12 + microglia. A possible explanation for this is that TMEM119 is also expressed on amoeboid immunoreactive microglia, whereas P2RY12 is considered more exclusive to homeostatic ramified microglia [
99,
127]. Therefore, subsets of microglia could be affected differently or possibly P2RY12 would be more sensitive to changes in microglial homeostasis. Previously, we and others observed a clustered distribution of microglia in multiple brain regions in ALSP, where some areas were completely devoid of IBA1 + microglia [
83,113]. Kempthorne et al. (2020) also identified reduced expression of microglial genes in ALSP, but speculated that this was due to a loss of homeostatic gene expression rather than a loss of microglia [
52]. However, this does not explain the local density differences we observe, including local clustering. A shortage of microglia in one area could perhaps lead to altered distribution by attracting microglia away from other areas, driven by an imbalance in the need and availability of microglia. Clustering of microglia, e.g., in response to cell death or plaques, is typical for various neuropathologies. However, in ALSP, these clusters of immunoreactive microglia, which could develop later in disease as a response to pathology, may further drive depletion in areas with an already low density of microglia by pulling away microglia towards areas of pathology. Microglial depletion caused by recruitment to areas in need could be further reinforced by the defective microglial proliferation. In
csf1r-deficient zebrafish, we also observed this phenomenon, where microglial recruitment after neuronal ablation involved migration of resident microglia to the site of injury, in the absence of microglial proliferation [
83]. Our data indicate that ALSP involves both a loss of homeostatic microglia as well as a general microglial depletion, predominantly in the white matter. Of note, we observed clusters of CD163 + macrophages/microglia, predominantly in white matter, and an upregulation of
CD163 in ALSP tissue. CD163 is associated with an anti-inflammatory microglial signature driving remyelination and found in active MS lesions [72,125]. Since we also noticed brain areas with few CD163 + cells, the CD163 + clusters may again be a consequence of an overall depletion of microglia. Notably, we cannot exclude the possibility that a proportion of these CD163 + cells are infiltrating peripheral macrophages. Altogether, our findings further support the concept of HSCT as a treatment for ALSP, which may act by repopulation of a depleted microglial niche, comparable to the effect of HSCT in adrenoleukodystrophy where pre-lesion areas are also characterized by reduced numbers of microglia [
12].
We demonstrate that 2 independent pathogenic CSF1R missense variants in the zebrafish
csf1ra locus act dominantly in decreasing density of microglia, whereas a null allele, leading to protein production from only one allele, had no effect. This dominant effect in vivo is further supported by recent observations in mice carrying a heterozygous ALSP-causing CSF1R missense variant (
Csf1rE631K/+), showing reduced numbers of microglia [
109]. Also, in vitro biochemical experiments show that pathogenic CSF1R missense variants act dominantly on CSF1R signaling, and parents of bi-allelic patients with heterozygous haploinsufficient
CSF1R variants have not been noted to develop brain disease [40, 50, 55, 74, 81, 92, 114]. Furthermore, 86% of pathogenic variants in
CSF1R are missense variants, predominantly located in the TKDs, whereas only 4% of pathogenic variants in
CSF1R are frameshift variants (Suppl. Table 2, Online Resources). As most of these frameshift variants are located in the C-terminal end, they could in fact also lead to a dominantly acting truncating protein. It is not clear whether any ALSP case can be explained by haploinsufficiency. Indeed, haploinsufficient
Csf1r+/- mice show no decrease in microglia, whereas complete knockout mice have no microglia [
7,
19,
34]. Therefore, although such models can be very useful, they should be treated with caution when used to study ALSP. What is intriguing, however, is our observation of the differential effects on the number of microglia between CSF1R missense variants. As patients with the same
CSF1R variant show major differences in age of onset and disease severity, it is likely that environmental factors and/or genetic factors play a disease-modifying role. This obscures possible genotype/phenotype relationships, and therefore, it has remained unclear whether pathogenic
CSF1R variants correlate with ALSP age of onset and severity. Our results suggest that there likely are variant-specific effects contributing to the level of microglial depletion, and by extension possibly also disease severity. Together with possible environmental or genetic modifying factors, this could explain the variance in age of onset and disease course of ALSP patients.
Through unbiased analyses, we identified an altered astrocytic phenotype in ALSP brain tissue and in zebrafish already during embryonic development. In ALSP tissue of severely affected frontal gyrus, but also in relatively spared white matter of the occipital gyrus, astrocytes were more hypertrophic, which is associated with elevated reactivity [
35]. Thus, astrocytic abnormalities may be an early consequence of microglial loss—although we cannot exclude that they originate from another cause—and possibly also play a role in early ALSP pathogenesis. Of note, since zebrafish astrocytes morphologically differ from those in mammals, it is not yet evident to what extent the phenotype we observed in radial astrocytes is analogous to that of astrocytes in ALSP, but it nevertheless gives an indication of early embryonic changes correlating with microglial depletion in the vertebrate brain. Notably, radial astrocytic lysosomal inclusions in heterozygous missense zebrafish mutants were transiently present, which could imply that the astrocytic compensatory response is needed temporarily when the phagocytic demand is high, since there is an increased number of apoptotic cells in the zebrafish brain around 3 dpf [
45]. The astrocytic phenotype in our zebrafish mutants is reminiscent of the compensatory endocytic response by astrocytes in microglia-depleted mice [
30,
57]. Indeed,
csf1raV614M/+ larvae showed increased endocytosis of myriad substances, including NR dye, apoptotic particles and even myelin. We also observed phagocytized myelin debris inside astrocytes in ALSP brain tissue. This is reminiscent of observations in demyelinating lesions in MS and metachromatic leukodystrophy [
90], although the amount of myelin debris within astrocytes appeared remarkably high in ALSP patient tissue. Astrocytes needing to process and degrade phagocytic waste could lead to neglected homeostatic functions, including energy supply to neurons and oligodendrocytes, blood–brain-barrier integrity, and uptake of neurotransmitters [
16,
42,
54,
68,
84,
118]. Several leukodystrophies, e.g., Alexander’s disease, Megalencephalic leukoencephalopathy with subcortical cysts and Vanishing White Matter disease, are caused by genetic variants in astrocyte-associated genes or involve astrocytic dysfunction as a pivotal pathogenic mechanism [
65,
121]. Therefore, abnormal astrocytic phenotypes appear sufficient to cause leukodystrophy. On the other hand, astrocytic endocytosis may partially rescue the shortage of microglial debris clearance, which could be important for prolonging the maintenance of a healthy CNS. Hence, compensatory endocytosis by astrocytes could have both beneficial and detrimental effects on brain health, but future research is needed to further establish this.
The importance of understanding the effects of microglial depletion in the human brain is increasingly relevant as multiple clinical trials are focusing on CSF1R inhibitors to deplete microglia in brain disease. We focused our omics analysis on occipital gyrus tissue, where white matter is relatively spared and pathology is relatively mild, allowing us to detect moderate changes that correlate well with an early stage of disease. Tissue analyzed included both grey and white matter, to allow detection of possible changes in both grey and/or in white matter. Of note, since ALSP is a rare disease and high-quality post-mortem brain tissue is scarce, our multi-omics analyses were performed on brain tissue of two ALSP patients who underwent autopsy with a very short post-mortem delay. It is unclear whether our analyses on two patients fully represent the general ALSP population. Nevertheless, we could replicate findings from these datasets by IHC in multiple patients and in zebrafish models. We found relatively few changes by transcriptome and proteome analysis in ALSP brain tissue, including downregulation of microglial genes, an elevated stress/heat shock response and increased abundance of astrocytic proteins. In particular, glutathione S-transferases (GSTs), including GSTM1, were upregulated at the transcript and protein level. In mice, GSTM1 is secreted by astrocytes to stimulate a pro-inflammatory microglial response [
51,
103]. As we find abundant GSTM1 expression in astrocyte-like cells in AD and FTD post-mortem brain tissue, astrocytic GSTM1 upregulation may be present across neurodegenerative diseases. We also found differential expression of ECM proteins in ALSP brain. Recent studies reported microglial ECM remodeling, including pruning of aggrecan and brevican, of which the latter is slightly increased in ALSP brain tissue [
7,
23,
80,
112]. Notably, we cannot exclude whether these changes might in part reflect region-dependent effects rather than disease stage differences, and in fact others have observed regional microglial changes in ALSP [
52]. In our proteome and transcriptome analysis, we cannot distinguish cortical or white matter-specific changes. Nevertheless, based on immunohistochemical staining on the same, relatively spared tissue, we did not detect differences that were exclusive to either cortex or white matter, except for increased expression of LAMP1 + in S100β + astrocytes only in cortical tissue, a more hypertrophic morphology of S100β + astrocytes in spared white matter and astrocytic engulfment of myelin in demyelinating lesions. This could indicate that astrocytes in white versus grey matter respond differently to the lack of microglia. Microglial depletion could influence white matter health by multiple possible independent effects, including possibly (indirectly) affecting astrocytic functioning through their compensatory endocytic response to elevated phagocytic demand. Our results here provide initial steps to further understand consequences of microglial depletion in the human brain.
In addition to having fewer microglia and altered radial astrocytes, heterozygous
csf1raV614M/+ zebrafish also had fewer sox10:RFP + oligodendrocytes in the hindbrain, but normal myelination and normal locomotor activity, whereas homozygous mutants showed a smaller myelinated area and reduced locomotor behavior.
Csf1r-/- mice also present with reduced oligodendrocyte cell numbers, although the overall myelin patterns in both
Csf1rE631K/+ mice and
Csf1r-deficient mice and rats appear relatively normal [
34,
91,
109]. Cunha et al. (2020) reported normal myelin content in the zebrafish
csf1r-mutant spinal cord, as we observed in larvae, but an impaired remyelination capacity [
26]. Thus, as mentioned above, there could be regional- and density-dependent effects of microglial depletion on myelin health in zebrafish. Overall, it is currently unknown to what extent the clinical manifestation of ALSP is preceded by cellular phenotypes as we observe in zebrafish.
Zebrafish as a neurobiology model allows exploring cells in embryonic development, in their natural environment by non-invasive in vivo examination. This has led to new fundamental insight in glial biology [
11,
71,
78]. In addition, options for genetic targeting have improved, including editing of specific genetic variants identified in human disease as we show here. Possible limitations, when used as a model for human brain disease, include dissimilarities at a more macro-scale such as the lack of a layered cortex and a low ratio of myelinated versus non-myelinated brain tissue, which may obscure pathology in adult animals. Furthermore, similar to rodent models, phenotypes in zebrafish models for leukodystrophies are relatively mild. For example, unlike in human disease, fully CSF1R-deficient mutant zebrafish and rats are viable, and have a mild phenotype [
62,
91]. Therefore, comparisons between multiple models are needed to make conceivable conclusions on disease mechanisms. Nevertheless, zebrafish in particular are suitable to detect phenotypes that go unnoticed in other models, as we show here, to subsequently explore further in other model systems and post-mortem brain tissue.
Based on phenotypes described for complete loss of function of CSF1R in humans, rats, mice and zebrafish, most peripheral macrophages are also depleted, leading to various developmental abnormalities outside of the CNS [
11,
29,
34,
50,
62,
81,
91]. As yet, it is still unclear what the effects of pathogenic
CSF1R variants are on peripheral macrophages and CNS-myeloid cells other than microglia in ALSP patients, although two studies reported on effects on peripheral monocytes/macrophages, including impaired phagocytosis and reduced presence of SLAN + macrophages [
41,
47]. Skeletal abnormalities are obvious in patients with bi-allelic CSF1R variants, but no skeletal abnormalities have yet been noted in ALSP patients [58]. Nevertheless, it is intriguing to investigate extra-cerebral tissues and macrophages in ALSP, including possible involvement of skull bone marrow-derived myeloid cells [
25].
Concluding, we provide proof that depletion of (homeostatic) microglia in ALSP patients occurs as a result of pathogenic missense variants in CSF1R and additional evidence of the importance of microglia in the development and maintenance of a healthy brain. Furthermore, our findings indirectly support the beneficial effect of HSCT in ALSP—which may act by repopulation of an under-occupied microglial niche. It remains an open question how loss of microglia can result in severe white matter defects. We speculate that microglial depletion and/or dysfunction results in a cascade of effects on other brain cells, including astrocytes, due to lack of total phagocytic capacity and the disruption of microglial modulation of the CNS environment, that by an as yet unknown mechanism eventually leads to white matter abnormalities [
11]. A better understanding of this cascade would improve our knowledge of ALSP and potentially other disorders where loss of microglial function is implicated. Taken together, further investigating ALSP and the effect of CSF1R missense variants in vivo may provide opportunities for improving microglia-focused treatment strategies and insight into the consequences of depletion of microglia, and possibly other macrophages, for the human brain.