Skip to main content

Progranulin and GPNMB: interactions in endo-lysosome function and inflammation in neurodegenerative disease

Abstract

Background

Alterations in progranulin (PGRN) expression are associated with multiple neurodegenerative diseases (NDs), including frontotemporal dementia (FTD), Alzheimer’s disease (AD), Parkinson’s disease (PD), and lysosomal storage disorders (LSDs). Recently, the loss of PGRN was shown to result in endo-lysosomal system dysfunction and an age-dependent increase in the expression of another protein associated with NDs, glycoprotein non-metastatic B (GPNMB).

Main body

It is unclear what role GPNMB plays in the context of PGRN insufficiency and how they interact and contribute to the development or progression of NDs. This review focuses on the interplay between these two critical proteins within the context of endo-lysosomal health, immune function, and inflammation in their contribution to NDs.

Short conclusion

PGRN and GPNMB are interrelated proteins that regulate disease-relevant processes and may have value as therapeutic targets to delay disease progression or extend therapeutic windows.

Background

A significant challenge in the field of human health is neurodegenerative disease (ND). An estimated 4.7–6 million individuals in the United States alone are diagnosed with some form of a ND, presenting a significant burden on both caregivers and patients alike [1,2,3,4,5]. Medication to address symptoms of NDs exists, but disease-modifying therapeutics to reverse or delay disease progression do not yet exist. This makes the identification and manipulation of therapeutic targets a critical pre-requisite for advancing the development of effective treatment strategies. The specific symptoms and progression of NDs can vary widely, but a unifying and newly recognized feature of the most prevalent NDs, including Parkinson’s disease (PD), Alzheimer’s disease (AD), and Frontotemporal dementia (FTD), is the role of the immune system and central–peripheral neuroimmune crosstalk in both the etiology and progression of ND. Two proteins in particular have been highlighted by analysis of ND-associated genes: progranulin (PGRN) and glycoprotein non-metastatic B (GPNMB) [6,7,8,9,10,11,12,13,14,15]. Furthermore, recent evidence supports that a loss of PGRN results in an increase in GPNMB expression [17, 18]. The mechanistic relationship between these two proteins, as well as the potential phenotype associated with increased GPNMB, has not been elucidated. The purpose of this review is to discuss the field’s current understanding of these two critical and interrelated proteins, with a focus on their roles in endo-lysosomal health and immune cell function.

Progranulin and GPNMB biology

Progranulin structural biology

The structure of PGRN has been described as “beads on a string”, where individual granulins (GRNs), the cysteine-enriched repeats that compose full-length PGRN, are connected together with short linker regions [19]. Once translation has been completed, the nascent PGRN is folded in the endoplasmic reticulum (ER), where the cysteine residues form disulfide bonds promoting the beta-folds in the GRNs and glycosylation of PGRN in both the ER and the Golgi at five asparagine loci: 118, 236, 265, 368, and 530. From the Golgi body, PGRN can be trafficked directly into the endo-lysosomal pathway or secreted out of the cell and allowed to circulate in biofluids (Fig. 1).

Fig. 1
figure 1

Diagram of the cell biology of progranulin and GPNMB. Figure created with BioRender.com

To be internalized, PGRN can bind to one of its receptors, sortilin, using a C-terminus motif near granulin 7 (granulin E) [20], or the C/D GRNs can bind to the BC linker region of prosaposin and piggy-back into the cell via prosaposin receptors, M6PR and LRP1 [21]. Once internalized, PGRN localizes to the endo-lysosomal system, where it is processed into individual, 6–8 kDa GRNs by lysosomal proteases, including multiple cathepsin species [22, 23], where they remain stable for a considerable but undetermined length of time [24]. In the extracellular space, PGRN can also be cleaved into GRNs by several matrix metalloproteases, such as MMP12 [25], ADAMST 7 and 12 [26], proteinase 3 [27], and elastase [27]. In contrast, secretory lymphocyte precursor inhibitor (SLPI) protects PGRN from proteolytic cleavage and allows it to persist in the extracellular space [28]. However, inhibition of PGRN uptake also decreases the extracellular GRNs concentration, strongly suggesting that GRNs are primarily generated by PGRN degradation in the lysosome [29].

Mature, secreted PGRN is a homodimer in the extracellular space, and it is this form that circulates in the plasma of both humans and mice [30]. Healthy individuals have an average of 96–125 ng/mL of blood PGRN [31, 32]. Altered levels of plasma PGRN can be used both as a diagnostic biomarker and a predictive variable for disease severity and/or progression [33, 34].

Progranulin expression

PGRN is expressed at varying levels in multiple cell types throughout the body, including neurons, muscle cells, endothelial cells, and adipocytes. Interestingly, myeloid cells, including microglia, macrophages, and monocyte-derived dendritic cells, express higher levels PGRN than most other cells [35, 36]. The amount of PGRN expression varies among myeloid cells residing in different body compartments, suggesting the cellular micro-environment modulates the expression, and several studies indicate that myeloid cells increase PGRN expression in response to immunological challenges [37, 38].

PGRN is necessary for healthy aging, but there are limited data to suggest that PGRN expression changes over the course of an organism’s lifespan, and genetic factors are far more likely to influence PGRN levels. Various frameshift, nonsense, and deletion mutations in the GRN gene exist in the human population, but the most common FTD-associated mutation in GRN is R493X, where an arginine codon is replaced with a termination signal [39]. This results in less PGRN being produced from the affected allele, as both the mRNA and protein product are degraded by nonsense-mediated decay (NMD). A single defunct allele results in less than 50 ng/mL of PGRN in blood, which is less than half of the average circulating level of PGRN in healthy controls, 96–125 ng/mL [31, 32].

Several other FTD-associated mutations result in the same effect, but three other major mechanisms of PGRN deficiency exist: a mutation in the signal recognition peptide (SRP), 5’ untranslated region (UTR) mediated suppression, and DNA methylation. The swap of an alanine to an aspartic acid at position 9 (A9D) in the nascent protein interferes with the signal recognition peptide (SRP) that successfully traffics PGRN to the ER. The resulting PGRN protein lacks the appropriate environment for protein folding and post-translational modifications (PTM) and is not viable [40]. Another method involves the 5’ UTR-mediated regulation of GRN mRNA. A previous study identified a novel mutation in the splice site of the 5’ UTR region of PGRN in FTD patients from the same family [41]. No other FTD-associated genes were abnormal, and a test for NMD was negative, yet the splice site mutation resulted in roughly half as much PGRN [41]. Subsequent work identified that the upstream open reading frame (uORF) elements within the 5’ UTR interfere with both the efficiency and the stability of the GRN mRNA, resulting in less PGRN protein being produced [42], but it remains unclear what factors determine the inclusion or exclusion of the longer 5’UTR. Finally, the degree of DNA methylation can epigenetically determine the expression of the GRN gene. Higher amounts of GRN DNA methylation result in less PGRN mRNA and protein [43].

One final mechanism regulates PGRN expression but does not rely on changes to the GRN gene itself. Micro RNAs (miR) are small pieces of RNA that modulate mRNA expression. The GRN gene has three miRs that bind to the 3’ UTR region and repress its expression [44,45,46]. Understanding the factors that regulate PGRN expression is important given the known association between alterations in PGRN expression and NDs and the potential of targeting these factors to modulate PGRN expression.

GPNMB structural biology

GPNMB is a glycosylated type-1 transmembrane protein that is localized to both the cell membrane and to components of the autophagy pathway. Originally identified in a low-metastatic cell line in 1995 [47], GPNMB was later found to not necessarily correlate with low metastatic activity and instead has been investigated for its ability and shown to promote metastasis in some mice models [48,49,50]. GPNMB is a member of the PMEL/NMB family and has a large extracellular domain (ECD) that includes a cell-attachment site at a.a. 64–66 (RGD), a PKD domain at 240–327, and a disordered region at 320–362 [51]. GPNMB is expressed as two isoforms generated from alternative splicing, which determines whether 12 amino acids are included or excluded at aa 339–340 [52, 53]. No distinct function has been ascribed to either isoform, but the modification of a disordered region may allow or preclude different protein–protein interactions. The short intracellular domain is only 53 amino acids, and it contains a half immunoreceptor tyrosine-activation motif (hemITAM) and a dileucine motif [51]. GPNMB is heavily glycosylated, with 12 asparagine sites for N-linked glycosylation [51]. Nascent GPNMB has a molecular weight of approximately 65 kDa, but the glycosylated forms have a weight of around 90 kDa and 115kDa [53]. The 115 k Da species of GPNMB is localized to the cell membrane and is susceptible to ECD cleavage by ADAM10 [54], but the 90 kDa species is localized to the endo-lysosomal pathway. All glycosylated GPNMB is subject to serine phosphorylation (S542), but only the fully mature 115 kDa species can be cleaved to generate the ~ 100 kDa GPNMB extracellular fragment (ECF) (sometimes called soluble GPNMB (sGPNMB)) [53, 54] (Fig. 1).

GPNMB expression

While low levels of expression exist in numerous cell types, GPNMB is primarily expressed by osteoclasts, osteoblasts, melanocytes, endothelial cells, myeloid cells, and antigen-presenting cells (APCs), such as macrophages and dendritic cells [48], which means GPNMB is expressed in multiple compartments throughout the body. The GPNMB ECF is also present in biofluids, including circulating in blood serum [55, 56] and urine [57]. Myeloid cells and astrocytes in the central nervous system (CNS) can secrete GPNMB ECF into the cerebrospinal fluid (CSF) [58,59,60]. At rest, control participants had an average blood serum GPNMB ECF of 31 ± 4.9 ng/mL [56], but the cleavage of GPNMB ECF is dynamic and can reflect the health of a patient, including immune cell activity [61,62,63], general organ health [64], and altered metabolism and/or endocrine function [56]. The regulation of GPNMB expression in immune cells has not been fully characterized, but inhibition of microphthalmia-associated transcription factor (MITF) activity resulted in decreased GPNMB expression in RAW264.7 cells [65], dendritic cells [66], and osteoclasts [67]. The MITF gene can be alternatively spliced into multiple isoforms of MITF [68], but it is unclear if any isoform(s) control GPNMB expression to a greater degree than the others.

GPNMB was also identified as a gene the expression of which is increased with aging in bone-marrow mesenchymal stem cells (bmMSC) collected from human subjects [69] and skeletal muscle macrophages in mice [70]. However, it is unclear if this increase is beneficial or deleterious. Vaccination against GPNMB significantly increased lifespan in a progeroid (Zmpste24 KO) mouse model of aging [71]. This approach could not distinguish between cell types, but the removal of GPNMB-positive cells was beneficial in this model, suggesting that GPNMB has a role in aging that has not been fully elucidated.

PGRN and GPNMB biological functions

PGRN

As described previously, PGRN localizes to organelles in the endo-lysosomal system and is processed into individual GRNs by lysosomal proteases. Within the lysosome, PGRN deficiency has been linked to an increase in lysosomal pH [72, 73], but there is no change in the amount of vacuolar-type-H + ATPase (V-ATPase) present in cells with and without sufficient PGRN [73]. Regardless of the precise mechanism, the increase in lysosomal pH and, therefore, decrease in available H + ions would decrease the ability of acidic lysosomal enzymes to function, which in turn increases the expression of those lysosomal enzymes [72, 73]. Indeed, insufficient PGRN dysregulates cathepsin D expression [74,75,76,77], beta-glucocerebrosidase (GCase) [78,79,80], and B-hexosaminidase A (HEXA) [81]. Each of these enzymes has an important role in lysosomal metabolism, and like PGRN, a loss of function in these enzymes is associated with lysosomal storage disorders (LSDs).

Another underrepresented facet of lysosome biology that is related to PGRN is lipid metabolism. Multiple groups have identified altered lipid metabolism in the cortex of mouse progranulin (mPGRN)-deficient mice [82, 83] and in FTD patient plasma [84]. The precise mechanism is not fully understood, but a lack of mPGRN has also been associated with decreased amounts of bis(monoacylglycerol)phosphate (BMP) [82]. BMP is a phospholipid that exists exclusively in the lysosome, where the negatively charged phosphate group is believed to act as a raft or a dock for lipid degrading enzymes such as saposins B and C, acid sphingomyelinase (ASM), lysosomal phospholipase A2 (LPLA2), and GCase [85,86,87,88] that promotes their activity and allows lysosomal lipid metabolism to occur. BMP also interacts with apoptosis-linked gene 2 interaction protein X (ALIX), Niemann–Pick disease type C2 protein (NPC2), and heat shock protein 70 (HSP70) [89,90,91]. These BMP-binding partners also play a role in endo-lysosomal membrane and sorting dynamics by altering the curvature and cholesterol trafficking of the endo-lysosome membrane [92, 93]. Further work is needed to determine the extent to which PGRN-deficient lipid metabolism is mediated by BMP and/or through other pathways.

Within the brain, PGRN is largely associated with neurogenesis and neuroprotection. Five days after sustaining a closed-cortical impact, Grn knock-out (KO) mice developed increased perilesional axonal injury relative to wild-type (WT) controls despite having the same size lesion and a similar number of IBA + myeloid cells in the area [38]. qPCR from RNA collected from the ipsilateral side of the injury revealed increased mRNA of inflammatory markers, including Il-1β, Il-6, and Tnf, in the Grn KO mice relative to WT controls but decreased mRNA for the anti-inflammatory Il-10 [38]. In a stroke model, WT mice underwent surgery to permanently block their middle cerebral artery (pMCAO) and, 30 min later, were administered recombinant PGRN directly into the right ventricle (ICV). Mice that received a single dose of PGRN showed increased numbers of proliferating neuronal stem cells (NSCs) in the subgranular zone of the dentate gyrus relative to both mice that did not receive PGRN or had a sham surgery [94]. The same PGRN treatment paradigm also rescued anxiety-like phenotypes and cognitive deficits that were apparent in the pMCAO mice [94]. Finally, while there is no genetic association between GRN and ALS, PGRN immunopositivity is increased in microglia found in the spinal cord of ALS patients relative to controls [95, 96]. A similar increase in Grn expression is noted during the progression of symptoms in mouse models of ALS [97].

Collectively, it seems that PGRN expression is beneficial in the CNS, with a loss of PGRN being deleterious in multiple models of CNS injury or neurodegeneration. Outside of the brain, the loss of PGRN impacts various organs and peripheral compartments differently, but the peripheral immune system has been reported to be significantly impacted by a decrease in PGRN expression. Human GRN-mutation carriers with clinically symptomatic FTD symptoms were reported to have increased plasma levels of sCD163 and CCL18, both of which are peripheral myeloid cell markers, suggesting an increase in peripheral immune activity relative to asymptomatic GRN-mutation carriers and healthy controls [98]. This also highlights the importance of compensatory mechanisms during aging in GRN-mutation carriers, as asymptomatic GRN-mutation carriers that were, on average, 7.6 years younger than the symptomatic GRN-mutation carriers, had immune profiles that were not significantly different from healthy controls [98]. Furthermore, another marker of peripheral immune activity and inflammation, lipopolysaccharide-binding protein (LBP), was increased in GRN-mutation carriers and correlated with white-matter changes in the frontal cortex of both symptomatic and asymptomatic GRN-mutation carriers [98], suggesting there is a spectrum of peripheral immune activation and CNS changes that exist prior to clinical diagnoses.

Macrophages from mouse models of PGRN deficiency also exhibit altered responses to lipopolysaccharide (LPS), including increased transcription of Mcp-1, Il-12p40, and Tnf, as well as decreased transcription of Il-10 relative to WT controls [99]. This is consistent with a greater pro-inflammatory response in Grn KO macrophages than in WT controls. Grn KO mice also demonstrated increased vulnerability to challenge by Listeria monocytogenes despite their increased pro-inflammatory activity [99]. In-depth investigation into the phenotypes of peripheral and central immune cell populations of 20-month-old Grn KO mice revealed sex-specific differences, including a higher frequency of Ly6C high monocytes in the blood of Grn KO females relative to WT females [100]. Specifically, Grn KO males showed a similar increase in MHCII, but it did not reach statistical significance. In the brain, Grn KO females displayed decreased MHCII expression on microglia compared to WT controls, while males showed no difference regardless of genotype. There was also a difference in T-cell populations, with Grn KO males exhibiting an increase in peripheral blood CD8 + T cells relative to WT controls, with no significant difference observed in Grn KO females [100]. In addition, CD44 expression was altered in T-cell populations: CD4 + T cells expressed less CD44 in the blood of Grn KO females, and the loss of PGRN abrogated the sex-dependent difference of CD44 expression on CD8 + T cells in the blood of WT controls [100]. In the brain, Grn KO males had increased counts of CD8 + T cells relative to WT controls, while Grn KO females had no change in CD8 + T cells relative to controls [100]. It is unclear what mechanism(s) are behind the sex differences of these immune cell populations, although modulation of Grn expression in the rodent brain by sex hormones has been reported [101], and in humans, there are known sex differences in the prevalence of neurodegenerative conditions, such as FTD–GRN, AD, and PD [102,103,104]. Altogether, it is reasonable to expect that mutations impacting PGRN levels may have differential sex-dependent immunomodulatory effects.

Although full-length PGRN in the extracellular matrix was reported by one group to interact directly with TNF receptors 1 and 2 (TNFR1/2) in a fashion that competed with native TNF binding for its own receptors [105], several other groups were unable to replicate this effect using multiple assays and experimental approaches [106,107,108]. Recently, new evidence has suggested that another protein, Y-box-binding protein (YB-1), may interact with both progranulin and TNFR, potentially explaining the discordant results [109]. YB-1 has several notable intracellular effects, but the protein can also exist outside of the cell, where its function is less understood [110, 111]. As an example, the addition of a combination of YB-1 and full-length mPGRN blunted the TNF-mediated inflammatory effect on bone-marrow-derived macrophages (BMDMs) by showing decreased phosphorylated nuclear factor kappa-light-chain enhancer of activated B cells (pNFkB), which is a critical transcription factor for inflammatory responses [109]. Further work on mPGRN–YB-1 interactions with TNFR will be needed to determine the kinetics of inflammatory modulation and the extent to which YB-1-enhancing therapy could be a viable approach to PGRN insufficiency.

GPNMB

The precise role of GPNMB in the phagolysosome is unclear, but evidence generated from epithelial cells and macrophages suggests that GPNMB aids in LC3 recruitment to the phagosome, thereby allowing phagosome fusion to the lysosome [112]. However, the majority of LC3 + /GPNMB + vesicles observed were not double-membrane vesicles, which is the signature of autophagosomes [112]. GPNMB also appears to have a role in signaling and contains a conserved hemi-ITAM domain (YxxI) on the C-terminus [51]. Src, a tyrosine kinase, phosphorylates this domain to activate downstream effects [113]. The precise effects are likely cell-dependent, but in an epithelial cell line, the expression of the GPNMB ITAM mutant (GPNMB YF) leads to a significant decrease in cell migration and sphere formation, which is consistent with the epithelial–mesenchymal transition (EMT), a process related to embryogenesis, wound healing, and cancer metastasis [113,114,115]. It remains unclear the extent to which the hemi-ITAM on GPNMB contributes to downstream effects in other cell types.

The role of GPNMB in the CNS is less understood, but it has been implicated in development. An early mouse model harboring a mutant Gpnmb allele develops pigmentary glaucoma between 1 and 3 months of age, but the presence of a functional Gpnmb allele prevented the development of glaucoma entirely [116]. IBA1 staining of the retina and optic nerve of D2 mice revealed increased numbers of IBA1 + cells at 1, 3, and 5 months of age relative to D2G mice [116]. This supports the argument that mutant GPNMB, specifically in IBA1 + myeloid cells, promotes the mechanism(s) of pigmentary glaucoma to occur at a young age, but a single functional allele of Gpnmb prevents the development of glaucoma.

Interestingly, GPNMB expression has also been reported to increase in CNS disorders. Hexb−/− mice, a model of Sandhoff disease, have a significantly increased amount of Gpnmb transcript relative to Hexb+/− mice [117]. In addition, immunohistochemical staining shows an increase in GPNMB-positive signal in both the thalamus and the brainstem relative to Hexb+/− mice [117]. However, both the increase in transcript and immunohistochemical staining can be rescued with the inhibition of glucosylceramide synthase (GCS), the enzyme that produces beta-D-glucosylceramide, the precursor to the sphingolipid species that accumulate in Sandhoff disease [117]. GPNMB expression also increases with neuroinflammation. Relative to non-neurological disease (NND) controls, Neuro-HIV brains showed an increase in GPNMB + IBA1 + cells in the brain [118]. This increase in GPNMB expression also correlated with an increase in disease-associated associated microglia (DAM) signature proteins, including HLA-dr, LGALS3, and CTSB, which is believed to influence the rate of synaptic pruning in Neuro-HIV patients [118]. The role(s) of GPNMB is still under investigation, but it is clear that GPNMB is regulated in response to changes in CNS health.

GPNMB has a well-documented effect on T cells, in both membrane-bound and cleaved forms, via interaction with the syndecan-4 receptor [119]. Syndecan-4 is expressed on both endothelial cells and T cells, but GPNMB exerts a cell-specific on them. Upon binding syndecan-4, membrane-bound GPNMB modulates the ability of naïve T cells to enter the S phase and produce Il-2, effectively inhibiting the primary response; and GPNMB also decreases re-activation of previously activated T cells [120]. Specifically, in oxazolone-sensitized mice, the supplementation of intravenous GPNMB ECD selectively inhibited the infiltration of T cells [121]. Instead of successfully infiltrating the inflamed skin, T cells were bound by endothelial cells that also expressed syndecan-4 [121], suggesting that the GPNMB–Syndecan-4 axis is responsible for physically orchestrating this critical immune response.

GPNMB ECD binds to CD44 and prevents the phosphorylation and translocation of NFkB in macrophages, which decreases the expression of inflammatory genes, such as Tnf, Il-1α, Il-1β, and Ccl2 [122]. A similar effect has been reported to occur in microglia and astrocytes [123]. Blocking the CD44 receptor prevented the effect of GPNMB ECD on modulating inflammation [122]. GPNMB ECD has also been observed to bind the alpha-1 and alpha-3 subunits of the Na + /K + ATP-ase (NKA) in NSC-34 cells, a motor-neuron-like cell type [124]. NSC-34 cells treated with GPNMB ECF showed an increase in cell membrane potential relative to untreated controls and increased the ratio of p-ERK/ERK and p-AKT/AKT [124]. This effect was abrogated by pharmacological inhibition of NKA, suggesting that GPNMB ECF both binds to and modulates the activity of NKA and the downstream signaling cascades.

Progranulin and GPNMB in neurodegeneration

PGRN has been described as the connecting genetic link between multiple neurodegenerative diseases potentially through regulating inflammation and immunity [6]. Here, a meta-analysis of a published data set from brain gene expression was performed to investigate the extent to which PGRN contributes to AD, PD, and amyotrophic lateral sclerosis (ALS) risk. Interestingly, a significant functional association was identified between increased genetic risk in the GRN locus region and decreased PGRN expression in PD, AD, and ALS. The authors concluded that PGRN is involved in the body’s immunological response and could potentially contribute to the etiology of several neurodegenerative diseases [6]. As noted above, while there is no genetic association between GRN and ALS, PGRN immunopositivity is increased in microglia in ALS spinal cord [95, 96] and in microglia in spinal cord of mouse models of ALS [97]. The following sections will explore the known effects and interconnections between PGRN and GPNMB in ND with a focus on neuroinflammation and central–peripheral neuroimmune cross-talk (Fig. 2).

Fig. 2
figure 2

Summary of progranulin and GPNMB in neurodegenerative disease. Figure created with BioRender.com

Frontotemporal dementia (FTD)

It is known that PGRN is required for healthy lysosomal function throughout the lifespan, because individuals who do not produce enough PGRN of their own are prone to disease. A single defunct GRN allele increases the risk of developing FTD, but two mutant alleles result in Neuronal Ceroid Lipofuscinosis (NCL-11), a lysosomal storage disorder (LSD) [125]. Symptoms of FTD can vary widely, even within a single family, but generally include progressive changes in behavior and personality and difficulties with language as well as some motor abnormalities [126]. Accordingly, clinical subtypes categorize FTD patients into behavioral variants (bv) or primary progressive aphasia (PPA), which contain two variants: non-fluent variant (nfvPPA) or semantic variant (svPPA) [126].

Age of onset also varies, but the R493X mutation has approximately a 60% penetrance by the age of 60, which increases to 90% by the age of 70 [127]. Histopathological investigation of FTD–GRN brains reveals an intracellular and intra-lysosomal buildup of lipofuscin, a pigment that is the result of failed lysosomal metabolism, and an accumulation of TDP-43, a protein involved in RNA management and gene expression [128, 129]. The mechanisms behind TDP-43 aggregation are unclear, but the significant burden of lipofuscin is consistent with other diseases with endo-lysosomal dysfunction.

Another feature of FTD is peripheral inflammation. Given the association of PGRN with inflammation, it is unsurprising that FTD–GRN patients exhibit a systemic inflammatory phenotype. One study completed in 2017 demonstrated that PGRN mutation carriers had increased levels of sCD163 and CCL18 in their blood, suggesting increased myeloid cell activation [98]. Furthermore, the amount of LBP correlated with white matter changes in the frontal lobe via MRI [98].

Changes in GPNMB have also been associated with GRN–FTD. Bulk RNA sequencing in the cortices of Grn KO mice showed an increase in Gpnmb mRNA relative to WT control [18]. This was later corroborated with proteomic data also from the cortex of Grn KO mice and analysis of human FTD–GRN patient brains [17]. These demonstrate an increase in both the RNA and protein of GPNMB in response to insufficient amounts of PGRN. The precise mechanism(s) driving this increase is unclear, but an increase in GPNMB expression has been noted in other varieties of lysosomal disorder, such as Gaucher’s disease (GD) and Niemann–Pick type C (NPC) [55, 60, 130]. Importantly, both pharmacological and gene transfer treatment for GCase deficiency can reverse the increase in GPNMB [55]. This finding suggests that the increase in GPNMB is specific to a compensatory pathway for endo-lysosomal dysfunction and not a result of secondary storage in the lysosome.

Lysosomal storage disorders (LSDs)

Lysosomal storage disorders (LSDs) are a heterogeneous group of heritable (inborn) metabolism defects that affect the function of lysosomes, comprising 70 monogenic disorders of lysosomal catabolism [131]. These disorders are caused by mutations in genes encoding lysosomal proteins, including membrane proteins, transporters, proteases, and lysosomal glycosidases, most of which are inherited as autosomal recessive traits. Mutations in these genes result in lysosomal malfunction and the gradual accumulation of substrates inside the lysosome, leading to cell dysfunction and cell death; these monogenic disorders can be subclassified according to the biochemical type of stored material, such as the sphingolipidoses, mucopolysaccharidoses, and glycoproteinoses. Of interest to this review are the neuronal ceroid lipofuscinoses (NCLs), which are neurodegenerative diseases characterized by the storage of abnormal lipopigment and lipofuscin in lysosomes. NCLs are fatal disorders that are clinically and genetically heterogeneous, with 14 genes implicated to date, (CLN1 through 14), including GRN [132].

As previously discussed, the majority of FTD–TDP-causing mutations identified are located in the GRN gene [133, 134], although more recently, it has been shown that other NCL-related genes are implicated in FTD cases, and the degree to which GRN mutations are identified in FTD-patients is region-specific [135]. Interestingly, it was subsequently reported that a complete loss of GRN results in adult-onset NCL [138]. It was reported that two siblings were shown to carry a homozygous deletion of four base pairs in the GRN gene (c.813_816del), which lead to a frameshift and premature termination of translation, and previously shown to cause FTD–TDP when inherited as a heterozygous mutation [137,138,139]. The co-occurrence of FTD and NCL within a family due to GRN mutations has been reported and provides the unique opportunity to investigate the genotype–phenotype correlations regarding GRN dosage effect; the homozygous relative displayed dysarthria, cerebellar ataxia, retinal dystrophy, and severe global cerebellar atrophy [140]. In contrast, heterozygous relatives presented behavioral variant FTD (bvFTD) and some extrapyramidal features compatible with corticobasal syndrome.

The discovery of the pleiotropic effect of homozygous and heterozygous GRN mutations was remarkable, given that the two clinically distinct neurological disorders have very different pathologies. Furthermore, given that the predominant clinical and pathological features of FTD and NCL are distinct, it is controversial whether the disease mechanisms associated with complete and partial PGRN loss are similar or distinct. Given the link between GRN mutations and NCL, it has been suggested that impaired lysosomal function may represent an important pathway leading to GRN-linked neurodegeneration. Interestingly, iPSC-derived cortical neurons from an FTD patient with a heterozygous GRN mutation exhibit neuronal phenotypes similar to characteristic hallmarks of NCL patient pathology, including lipofuscin accumulation and the appearance of fingerprint-like profiles and granular osmiophilic deposits [76]. Similarly, iPSC-derived cortical neurons derived from an NCL patient with homozygous GRN mutations exhibit TDP-43 mislocalization, enlarged autofluorescent lysosomes, and electron-dense vesicles containing storage material with granular, curvilinear, and fingerprints profiles [141]. GRN-mutant neurons from an FTD patient also showed decreased activity of the lysosomal enzyme cathepsin D via a loss of the PGRN cleavage product, cathepsin E. Interestingly, homozygous GRN and CTSD, which encodes for cathepsin D, mutations lead to a similar form of NCL [142,143,144], suggesting this functional relationship may provide a possible mechanism for the overlapping NCL-like pathology observed in GRN-neurodegeneration. Similarly, it has been shown that PGRN haploinsufficiency in humans leads to preclinical retinal lipofuscinosis and increased lipofuscinosis and intracellular NCL-like storage material also in the cortex, postmortem [125], all of which are NCL-like features and were seen, in some instances, prior to the onset of dementia.

It is clear from such human data that there are perhaps similar mechanisms between FTD and NCL despite the clinical distinctions between the two. However, studies from mice have thus far not been consistent with human data. Mice homozygous for a targeted deletion of the mouse Grn gene develop a robust neuropathological phenotype with age, with lipofuscin deposition and accumulation of NCL-like storage material as well as microgliosis and astrogliosis in multiple brain regions [145,146,147,148,149], as well as hyperinflammatory responses in both microglia and peripheral macrophages [25, 99, 150]. However, heterozygous Grn-targeted mice do not develop such phenotypes [145, 147], with phenotypes limited to decreased sociability and altered social dominance [151, 152]. It has been suggested that the lack of phenotypes in these mice may be due to these mice containing disrupted Grn alleles; therefore, they have limited utility in phenocopying progranulin-deficient FTD and NCL caused by nonsense mutations [153]. Recently, the GrnR493X mouse model was generated to more accurately model FTD–GRN by introducing one of the most common human nonsense mutations leading to FTD (R493X) at the analogous mouse Grn codon (R504X) [153], as previously discussed. Homozygous GrnR493X mice phenotypically replicate several neuropathological hallmarks previously demonstrated in Grn null mice and have been shown to have lysosomal dysfunction, neuroinflammation, and thalamic neurodegeneration [154]. Notably, evidence of lysosomal dysfunction has been demonstrated in Grn null mice as young as 2 months of age [155], while lysosomal dysfunction has only been reported in aged 18-month-old GrnR493X mice [155]. Future efforts should seek to understand whether the presence of a semi-functional, truncated Grn-R493X might delay the onset of this early lysosomal phenotype and how these phenotypes from murine models inform researchers of the shared and disparate phenotypes and mechanisms between FTD and NCL.

It has previously been demonstrated that the conditional loss of PGRN in neurons is not sufficient to cause NCL-like neuropathology in mice [156], leading researchers to hypothesize that GRN mutations in NCL may exert their disease effect in a cell-autonomous manner and that microglia may be the culprits underlying disease development and progression. However, selective reduction of microglial PGRN in neuronal PGRN-deficient mice failed to induce lipofuscinosis or gliosis observed in GRN null mice [157]. These data lead the authors to conclude that PGRN from extracellular sources may prevent pathology. However, PGRN expression is not confined to the brain, and it is clear that many LSDs have a peripheral component. Indeed, evidence suggests that the accepted name of NCLs as ‘neuronal’ may be a misnomer, since it is emerging that the effects of the disease are certainly not confined to neurons or even within the brain. Indeed, NCL proteins, including PGRN, are widely expressed in various tissues and cell types [100, 158,159,160,161], and therefore, it is unsurprising that other organ systems in the body are affected by deficiency in these proteins [162]. Indeed, as previously discussed, PGRN loss has recently been described to result in sex-dependent dysregulation of both the peripheral and the central immune system in mice [100], raising the interesting possibility that peripheral inflammation may also play a role in neurodegeneration due to Grn haploinsufficiency in human FTD–GRN mutation carriers.

Although the function of GPNMB in modifying NCL pathology has not been investigated in a clinical or pre-clinical settings, it has been shown that GPNMB is elevated in Niemann–Pick disease type C1 (NPC1) mice and patients, a rare and fatal neurodegenerative–LSD that arises from lysosomal accumulation of unesterified cholesterol and glycosphingolipids [130] and may be a potential biomarker for therapeutic trials [163]. In addition, GPNMB expression levels have been shown to be modulated in both central and peripheral myeloid cells in Grn-deficient mice, suggesting that PGRN and GPNMB may jointly regulate the peripheral and central immune system, and future research would benefit from further exploring the mechanisms behind this relationship and how it may be utilized as a potential biomarker for GRN-associated NDs, including NCL.

Alzheimer’s disease (AD)

AD is a ND characterized by the presence of amyloid beta (Abeta) plaques and tau neurofibrillary tangles, which clinically results in severe cognitive impairment [164]. The link between PGRN and AD are supported by genetics, where several GRN mutations in human studies have been associated with increased risk for AD, including a null mutation IVSO + 5G > C, the GRN nonsense mutation p.Arg535X [7], two missense mutations, p.Cys139Arg and p.Pro451Leu [8] and the exon 1 GRN mutations, p.Gly35Arg and pASP33Asp [9]. Among all, one of the most well-studied GRN variants is the rs5848 variant, which has been described in different AD populations [10,11,12] and has been associated with increased risk for AD by three meta-analyses [14, 165, 166]. However, some discrepancies have been suggested regarding its implications in PGRN expression, where the rs5848 variant translated in the decrease of GRN mRNA levels in the parietal lobe and PBMCs of AD patients [10] but also has been described to no altered GRN expression in serum levels between controls and AD patients [167].

Importantly, the role of PGRN as a biomarker for AD has been proposed, but it is also controversial. Specifically, GRN levels appear to be upregulated at the transcriptomic level in the blood of AD patients in comparison with controls. However, patients that carried the rs5848 variant displayed a decreased GRN expression [168, 169]. In contrast, the extent of PGRN protein expression in the blood showed no relationship with disease severity [169]. In addition to measurements in the blood, PGRN levels have been examined in the CSF of AD samples, with discordant outcomes reported. In one study, CSF PGRN levels were highest in AD samples compared with controls and MCI samples and were correlated with cortical thickness [170]; however, in another study, PGRN CSF levels did not differ between AD and non-AD patients and did not correlate with cortical thickness [171]. Importantly, the role of PGRN as a biomarker for AD has been considered due to its relationship to neuroinflammation [172], where CSF PGRN levels have been described to increase and to be associated with an increase in inflammatory markers in tau-neurodegenerative (TN) + patients [173]. Finally, a recent paper suggested that CSF levels of PGRN may represent a marker of AD disease progression rather than a diagnostic marker for AD [174].

A regional characterization of PGRN and GRNs expression in AD, FTD–TDP human brains, and unaffected controls with and without GRN mutations has been examined [175]. Here, neuronal PGRN immunopositivity was decreased in AD samples with and without GRN mutations, while PGRN was increased in microglia. Furthermore, differences in GRN immunopositivity were found; GRN C was found in microglia-positive cells forming patches in the cortex of control and AD brains, whereas GRN B showed the strongest signal in hippocampal pyramidal neurons [175]. In other studies, PGRN expression has been studied in the context of amyloid plaques. In the middle temporal gyrus, the size and number of PGRN-associated plaques increased in AD samples, and PGRN expression colocalized with microglia (IBA1 + cells) and vasculature (CD31 + cells) but not with astrocytes (GFAP + cells) [176]. In addition, a correlation between increased protein PGRN and Abeta and phosphorylated tau (p-tau) expression was observed [176]; however, there was no colocalization between PGRN and p-tau at cellular levels by immunohistochemistry. This relationship between PGRN and neurofibrillary tangles was further explored in a separate study, where PGRN, and also prosaposin, are decreased in neurons that develop neurofibrillary tangles compared with neurons without them [177].

Considering the findings in human studies, mouse AD models have been used to further understand the consequences of PGRN loss on AD-like phenotypic aspects such as Abeta plaque load, tau phosphorylation, inflammation, or synaptic loss and the protective effects of PGRN replacement in such models. The link between PGRN and AD through Abeta pathology has been explored in mouse models such as Tg2576, APP/PS1, or 5xFAD crossed with mPGRN-deficient mice. In Tg2576 mice, Grn has been shown to be upregulated in the hippocampus, and mPGRN immunoreactivity was observed around dense core plaques, which increased in aged mice. In these mice and at the cellular level, Grn overexpression was described in microglia, neurons, and neurites around dense plaques [178]. In APP transgenic mice, mPGRN levels decreased in the cortex of 3- and 7-month-old APPhigh mice and 11–12-month-old APPlow mice but increased in 13-month-old 5xFAD mice [179]. In addition, the role of the immune system and, more specifically, of microglia was suggested in the context of APP and PGRN deficiency. For example, Grn KO mice crossed with APPlow mice displayed differences in microglia burden, with higher CD68 expression in the hippocampus of APPlow Grn KO relative to APPlow WT controls. In addition, the expression of pro-inflammatory cytokines (such as Tnf and Il1b) were increased, and expression of anti-inflammatory cytokines (such as Il-4 or Cox2) was decreased. Finally, the selective depletion of mPGRN in microglia in APPhigh mice resulted in a 50% increase in hippocampal plaque load, hypothesized to be caused by an impairment of microglia phagocytosis resulting from mPGRN deficiency [179]. A similar finding on microglia was reported in the Grn KO x 5xFAD mice model [180]. In this study, mPGRN was shown to be expressed in microglia near Abeta plaques, and mPGRN depletion decreased Thioflavin S-reactive and Abeta plaques in the cortex of 4–5-month-old male mice. Finally, RNA-seq analyses of the hippocampus of 5xFAD mice with or without mPGRN revealed that lysosomal and inflammatory genes such as Cd68 or Gpnmb were involved in this process and, specifically, Gpnmb was shown to be upregulated in microglia around Abeta plaques in 5xFAD mice lacking mPGRN [180]. Together, these data indicate that PGRN has a potential role in Abeta deposition and clearance through the immune system, mainly microglial cells; hence, several studies have aimed to increase PGRN levels as a way to improve AD-like pathology in mice [179, 181, 182]. To examine the protective effects of PGRN in pre-clinical models of AD, Grn viral delivery has been used in the 5xFAD and the Tg2576 mouse models to increase mPGRN levels. In the 5xFAD mouse model, the increase in mPGRN levels decreased amyloid-beta plaque load, protected against Abeta toxicity, and a decrease in plaque load in the hippocampus and on neurons in the hippocampus was observed [179]. Similarly, Tg2573 mice that received a viral vector delivery of mPGRN (ND-602) displayed reduced amyloid plaque burden in the hippocampus and the entorhinal cortex, a reduction of the inflammatory proteins fluorescein-conjugated isolectin B4 (IB4), IBA1 and GFAP in the hippocampus, and a reduction in synaptophysin in the dentate gyrus and hippocampus. In addition, ND-602 gene therapy in Tg2573 increased neprilysin, an Abeta-degrading enzyme [182]. In addition to viral vector delivery, mPGRN levels have been increased in AD models using an intrahippocampal injection of mPGRN in the 5xFAD mouse model [181]. In this study, the injection of mPGRN to 5xFAD mice decreased amyloid deposition in the hippocampus of 5XFAD mice and decreased BACE1, which is responsible for the APP processing and generation of Abeta peptides. Finally, differences in neuroinflammation were observed after mPGRN intracerebral injection in 5xFAD mice, with IBA1 + cells colocalizing with Abeta plaques, and CD68/IBA1 area were both increased with PGRN injection [181]. Taken together, mPGRN deficiency models and strategies to replace or overexpress mPGRN in mouse models support its role in mitigating neuroinflammation and progression of amyloid-associated pathology.

In addition to Abeta, the relationship between PGRN and tau has also been studied in pre-clinical models [183]. P301L tau mice crossed with Grn KO mice revealed that tau phosphorylation was increased in the Tris-saline soluble fraction of 13-month-old, and the sarkosyl-insoluble fraction of 19-month-old P301L tau mice crossed with mPGRN-deficient mice [183]. Interestingly, an opposite effect on Abeta and tau was reported in APP/PS1 mice deficient in mPGRN, which displayed reduced diffuse amyloid-beta plaque deposition and increased neuronal injury [184]. Additional studies to clarify the role of mPGRN in the setting of amyloid pathology with and without accompanying tau pathology will be needed.

Presently, no known GPNMB mutations are associated with an altered risk for developing AD, but a histopathological study of AD brains showed a significant increase in GPNMB-positive staining in the frontal cortex relative to control [185]. The GPNMB-positive staining primarily colocalized with microglia cell markers, and the GPNMB-positive microglia were clustered around Abeta deposits and phosphorylated tau pathology, suggesting that GPNMB expression is associated with microglial activation [185]. This result is consistent with previous work showing increased GPNMB expression in the brains of sporadic AD patients, which localized around both amyloid plaques and blood vessels [186].

The same study also investigated the role of GPNMB on amyloid-associated pathology in the 5xFAD mouse model. One interesting observation was an age-dependent increase in cerebral Gpnmb mRNA beginning at 7 months of age in the 5XFAD mice relative to WT controls. A similar effect in cerebral Gpnmb mRNA was observed in the APP/PS1KI model relative to PSKI animals but was absent in the APP23 mouse model [186]. Immunofluorescent staining of 12-month-old 5xFAD mice brains showed GPNMB colocalizing with IBA1 but not with GFAP or NeuN, confirming the increase in GPNMB occurred predominantly in myeloid cells [186], which may consist of both microglia and infiltrating peripheral monocytes. It was also demonstrated that GPNMB-positive microglia in 12-month-old 5xFAD mice localize around amyloid plaques, consistent with the findings in human AD patient brains, yet age-matched APP23 mice showed no positive GPNMB staining [186]. These findings suggest that differences in GPNMB expression may be model dependent and potentially associated with different levels of inflammation at different states of pathology.

Recent work has been aimed at investigating the effects and significance of increased GPNMB expression in both AD patient brains and 5xFAD mouse brains. Lentivirus vector overexpression of GPNMB in the brains of APP/PS1 mice rescued Morris Water Maze performance in APP/PS1 mice [187], suggesting that the observed increases in GPNMB in some models of AD-like neurodegeneration may be adaptive or protective and not maladaptive or detrimental. In addition, immunohistochemical analysis of the GPNMB overexpression mouse group revealed decreased amyloid deposition relative to the control mouse group that received the empty vector. This decrease in amyloid deposition was correlated with alterations in LC3-II, p62, and mTOR phosphorylation levels in hippocampal lysate from GPNMB-overexpressing mice [187]. Further mechanistic work in BV2 microglia cells confirmed the changes in LC3-II, p62, and mTOR phosphorylation and further demonstrated that GPNMB overexpression resulted in increased phagocytosis of Abeta relative to empty vector controls, and this effect was abrogated with the addition of an autophagy inhibitor, 3-MA [187]. These findings suggest that GPNMB modulates increases in autophagy, which may be the mechanism responsible for the associated decrease in Abeta deposition.

Given that the extracellular domain of GPNMB can be shed as an extracellular soluble fragment (ECF), it is a potential biomarker candidate for neurological disease, including AD, but different groups disagree as to whether GPNMB is a reliable biomarker of disease progression. A paper published in 2020 compared multiple sets of AD CSF proteomes with their own two-dimensional liquid chromatography fractionation and high-resolution tandem mass spectrometry (abbreviated as TMT–LC/LC–MS/MS) [188]. Here, GPNMB ECF was detected in their own mass spec data, but GPNMB ECF only overlapped with one of the three CSF proteomes that were used to compare expression [188]. Follow-up analysis with an ELISA showed an increase in GPNMB ECF in the CSF of AD patients relative to non-AD controls, and the concentration of GPNMB ECF correlated with the spectrometry data from the same CSF sample [188]. Given their detection and confirmation of GPNMB ECF in the CSF of AD patients, the authors contend that GPNMB is a viable biomarker for AD but caution that less sensitive approaches to mass spectrometry may miss a majority of the proteins detected in the AD CSF proteome. In contrast to that work, a paper published in 2021 found no difference in GPNMB ECF concentration in the CSF of AD patients versus healthy non-demented controls [65]. It should be noted that these papers used different methods to detect GPNMB ECF in addition to having studied different cohorts of AD patients with dissimilar disease stage. At present, there is no definitive conclusion about the reliability of GPNMB ECF as a viable and specific biomarker for AD and future studies will be needed to better understand its role in AD and potential as a biomarker.

Parkinson’s disease (PD)

PD is a neurodegenerative motor disorder characterized by the loss of dopaminergic neurons in the nigrostriatal pathway and the presence of alpha-synuclein (α-syn) inclusions [189]. Although the majority of research regarding the role of PGRN in neurodegeneration has been focused on FTD, LSDs, and AD, there is mounting evidence linking the importance of PGRN function with development of PD. In 2007, Leverenz et al. used a combination of neuropathological and molecular genetic approaches to phenotype two families with the GRN mutation c.709-2A > G which reduces GRN mRNA expression [13]. In this study, patients that carried this specific PGRN mutation exhibited language impairment, behavior disturbances, and parkinsonism as clinical manifestations, probably explained by the loss of neurons in the neocortex, striatum, hippocampus, and substantia nigra, respectively. In addition, gliosis was observed in the striatum and substantia nigra of the affected patients relative to controls implicating PGRN in modulation of immune responses associated with PD pathophysiology. Interestingly, six cases exhibited ubiquitin/TDP43 immunopositive inclusions, all but two had tau pathology, and two of them exhibited α-syn inclusions, suggesting that this specific GRN mutation does not only contribute to the development of FTD but also to parkinsonism and α-syn pathology [13].

More recently, a meta-analysis was performed in 16 case study to investigate the link between the GRN polymorphism at rs5848 with neurodegenerative diseases, such as FTD, AD, PD, and ALS [14], and the authors conclude that the polymorphism at rs5848 is associated with a higher risk of AD and PD. Supporting this, reduced PGRN plasma levels have been associated with PD severity [34], where PGRN measured by ELISA negatively correlates with the PD severity and disease duration measured by the Unified Parkinson’s Disease Rating Scale scores (UPDSR-III) [34]. Collectively, therefore, it seems PGRN expression and function may be implicated in modulation of PD onset and/or progression, although the precise mechanism linking this to disease is currently unknown, but we posit is associated with neuroinflammation and lysosomal dysfunction that contributes to α-syn pathology.

The role of PGRN in modulation of PD onset and progression have been studied in human patients and mouse models of PD-like pathology. In this context, Martens et al. investigated whether the putative anti-inflammatory properties of PGRN [151] contributed to neurodegeneration in a PGRN-deficient setting by exacerbating neuroinflammation. The authors used mPGRN-sufficient mice (Grn + / +) and mPGRN-deficient mice (Grn KO) treated with MPTP, a known neurotoxin that affects the dopaminergic neurons in the substantia nigra pars compacta (SNpc). The authors observed that mPGRN-deficient mice exhibited an increase in MPTP-induced neuronal loss in the SNpc and that this effect was mitigated in MPTP-treated mPGRN-deficient mice after mPGRN replacement via lentivirus [151]. In addition to this and using conditional mutants lacking mPGRN in microglia, the authors confirmed that the loss of dopaminergic neurons in the SNpc is driven by microgliosis and not because of the selective vulnerability of neurons due to an increase of activated microglia in mPGRN-deficient mice treated with MPTP [151]. Together, these important findings suggest that, in response to oxidative stress, mPGRN-deficient microglia and/or other myeloid cells adopt a hyperactive pro-inflammatory state, contributing to neuronal loss. Hence, mPGRN gene delivery seemed like a reasonable neuroprotective strategy in the MPTP model. Using a unilateral intranigral infusion of the lentiviral vector ND-602 encoding full-length mPGRN to efficiently transfect nigral neurons in MPTP-treated B6 mice, the neuroprotective effects of mPGRN gene therapy were demonstrated as measured by reduced locomotor deficits, including decreased akinesia/bradykinesia, and increased locomotion velocity and coordination [190]. ND-602 treatment also ameliorated neuronal death in the SNpc and striatum and exerted anti-inflammatory properties as measured by attenuated expression of IB4-positive microglial cells in comparison with mice treated with MPTP but not ND-602 [190]. Finally, a potential link between PGRN expression in microglia and α-syn burden has also been observed. In this context, mPGRN protein was reported to be decreased in mouse microglia exposed to α-syn ex vivo and in human microglia surrounding α-syn deposits [191], suggesting that ingestion of α-syn may negatively regulate mPGRN protein expression in microglia which could result in dysregulated phagocytosis and inflammatory responses. Taken together, all these studies implicate PGRN in development and progression of PD, most likely through modulation of inflammatory responses; yet the specific mechanisms by which this occurs have yet to be explored and will be needed to inform on the therapeutic potential of PGRN replacement in sporadic PD and other parkinsonisms.

Just like for PGRN, the link between GPNMB and PD has been explored in human samples and in mouse models of PD-like pathology. For instance, a single nucleotide polymorphism (SNP) in GPNMB at rs199347 is associated with PD risk [15, 18], and GPNMB expression has been reported to be increased in the SNpc of PD patient brains post-mortem [124, 192, 193]. In a paper published in 2018 by Moloney et al., the authors investigate two pathogenic mechanisms by which GPNMB contributes to PD; changes associated with lysosomal dysfunctions and regulation of glycolipids (in a CBE (conduritol-ß-epoxide) mouse model) or α-syn (in the Thy1-ASYN mice) [192]. In the CBE mouse model, GPNMB was increased in brain regions, such as the motor cortex, hippocampus, and SNpc upon CBE treatment. By contrast, in the brain of α-syn-overexpressing mice (Thy1–ASYN mice), no differences in GPNMB expression were found in any of the studied brain regions, indicating that GPNMB may contribute to PD pathology indirectly through lipidopathy changes [192].

Another recent study independently investigated the link between the rs199347 SNP and PD using a combination of computational, cell biological studies, and clinical samples [194]. Here, the rs199347 variant was associated with higher GPNMB expression in the brain and blood. In addition, using human pluripotent stem cells-derived cortical neurons expressing different levels of GPNMB, a key role of GPNMB in synaptic defects was suggested, where GPNMB was reported to be required for α-syn pre-formed fibril (PFF) uptake by neurons; importantly, its potential role in immune cells was not explored [194]. Interestingly, interrogation of the role of GPNMB in α-syn biology using mouse models has yielded controversial results. In this context, the work published by Brendza et al. showed that GPNMB loss did not modify changes in a remyelination mouse model or in two different mouse models of PD-like α-syn pathology (synthetic human α-syn PFFs and AAV1/2-CMV/CBA virus vector driving human mutant A53T α-syn) [195]. With regards to the α-syn models, human α-syn PFF injections into the striatum were employed to evaluate the differences in α-syn spreading and aggregation, motor dysfunction, and gene expression between WT and GPNMB-deficient mice. While α-syn PFF aggregation caused α-syn spreading in the midbrain, striatum, and brainstem, neurodegeneration in the midbrain and brainstem, and both astrogliosis and microgliosis, no difference was found in PFF-treated mice that lacked GPNMB compared to PFF-treated WT mice. In addition to these neuropathological measures, behavior studies such as wire hang and open field showed no differences in mice lacking GPNMB. Finally, RNA-sequencing from the striatum, midbrain, and brainstem revealed that Gpnmb was the only differentially expressed gene (DEG) that was identified between WT and Gpnmb KO PFF injected mice, indicating that at the transcriptional level, there were also no differences between WT and GPNMB-deficient mice treated with PFFs [195]. Similar findings were observed in a second α-syn model, where the over-expression of human mutant A53T by AAV in WT and GPNMB-deficient mice showed no differences in TH loss, and Gpnmb was again the only DEG upregulated gene in midbrain and striatum in RNA-seq analyses [195]. Together, these surprising findings indicate that, at least in two mouse models of α-syn pathology, GPNMB is not required to protect against it. An alternative interpretation is that global GPNMB deficiency in mice resulted in genetic compensation that rendered neurons resistant to α-syn-induced pathology. Additional studies on the role of GPNMB using acute knockdown strategies in different cell types will be needed to distinguish between these and other possibilities. Finally, Budge et al. explored the possibility of using Gpnmb overexpression as an anti-inflammatory regimen to prevent neuronal loss in the MPTP mouse model [196], where it was shown that transgenic overexpression of Gpnmb protects against dopaminergic neuron loss. In the context of neuroinflammation, at the gene expression level, the overexpression of Gpnmb in MPTP-treated mice reduced Aif1 and Gfap expression in the striatum and primary microglia treated with recombinant GPNMB and LPS increased the expression of anti-inflammatory genes, such as Arg1, Mrc1, Nrf2, Ym1, and Jmjd3, which are downregulated in LPS-treated WT microglia [196]. Together, these findings suggest that Gpnmb overexpression can exert protective anti-inflammatory effects. Additional studies will be needed to reconcile differences in the protective effects of GPNMB that appear to be model dependent and/or dependent on global versus conditional deletion of GPNMB in relevant cell types.

Summary of animal models

A great deal of the data discussed in this review was generated using Grn KO mouse lines to interrogate the consequences of progranulin-deficiency. However, there are slight differences in how each knockout line was generated. To succinctly present the differences, we produced a table that contains the commonly utilized Grn KO mouse lines, the differences in the manipulation of the Grn gene, as well as brief summaries of notable findings that demonstrate endo-lysosome dysfunction and altered inflammation and immune cell phenotypes (Table 1).

Table 1 Summary of PGRN KO mouse lines and their endo-lysosomal and immune cell phenotypes

Therapeutics in development

Currently, no disease-modifying treatments exist for FTD–GRN mutation carriers. Prior to onset, GRN mutation carriers maintain the same level of circulating PGRN as symptomatic FTD patients [32], but do not experience the behavioral changes and language difficulties associated with FTD. This highlights the role of aging in the development of FTD–GRN and suggests that a compensatory mechanism(s) exists that prevents or delays the progression of disease development. Successful identification of these mechanism(s) will enable targeted intervention to extend therapeutic windows or augment disease-modifying treatments for FTD–GRN mutation carriers.

The most logical approach to treat diseases rooted in PGRN deficiency would be to supplement the amount of PGRN or GRNs in the patient, and therefore, multiple therapeutics are in development to do exactly this. Denali Therapeutics has developed a protein transport vector progranulin (PTV–PGRN or DNL593) that is a full-length human PGRN protein attached to a transferrin ligand. This allows the human PGRN to bind to the transferrin receptor, which promotes the successful uptake of the PGRN protein into the CNS. This approach was shown to be effective in reversing the glial reactivity, BMP deficiency, and lipofuscin storage in a Grn KO mouse model [207]. Importantly, the authors also demonstrated similar rescue in mice dosed every week versus every other week, suggesting that the rescue may be, at least in part, mediated by the more stable lysosomal-resident GRNs thereby persisting over time without requiring more frequent dosing of PGRN. DNL593 is currently under clinical trials in patients carrying Grn mutations (NCT05262023) [212]. The potential for GRN-mediated therapeutic replacement was specifically explored using rAAV-mediated overexpression of human GRN2 or GRN4 in Grn KO mice. In a recent pre-print, the authors reported full-length PGRN was not required to rescue lipofuscinosis, microgliosis, and lysosomal function in Grn KO mice, and these two individual GRNs rescued to similar but not identical extents [213]. These exciting findings raise the possibility that direct therapy with GRNs may be an additional therapeutic approach in PGRN/GRN-deficient states. An alternative PGRN replacement therapeutic consists of gene therapy. Developed independently by Prevail Therapeutics and Passage Bio, PR006 and PBFT02 are adeno-associated virus (AAV)-based therapeutics that leverage a viral vector to selectively infect the CNS with a viral vector encoding PGRN [214]. This allows for PGRN to be generated in the CNS without the need for uptake and transport from the periphery and may be therapeutically efficacious after a single injection into the cisterna magna [214]. This approach has been used in human IPSC models and mouse models of PGRN deficiency and has shown significant rescue of PGRN levels [211, 215]. Finally, both are in clinical trials with outcome measures focused on the successful expression of PGRN, tolerance for the AAV vector, and neurological and neurocognitive improvements from baseline levels (NCT04408625 and NCT04747431) [216, 217]. Finally, ARKD-104, developed by Arkuda Therapeutics, is labeled a “PGRN enhancer” [214]. ARKD-104 is a small molecule drug that increases PGRN and GRN products and is capable of penetrating the brain [214]. However, the precise mechanism is unknown and may not be tolerated in all cases at an effective dose.

AL001, also known as latozinemab, is another approach to altering progranulin levels. Co-developed by Alector and GlaxoSmithKline (GSK), latozinemab is an antibody specific for the sortilin receptor designed to block full-length PGRN uptake [214]. In brief, the interruption of the PGRN–sortilin interaction prevents the internalization of PGRN and its traffic through the endo-lysosomal system, where it is cleaved into GRNs in the lysosome. The net effect of this inhibition is an increase in the amount of extracellular PGRN, while the intracellular PGRN remains fairly stable [20]. It is believed that sufficient amounts of PGRN are able to intracellularly traffic to the lysosome via non-sortilin-binding partners, such as the M6PR or LRP1, preventing lysosomal dysfunction, while the increased extracellular PGRN can be taken up by neurons or other cell types, where expression is relatively low. The PGRN therapeutics and their mechanisms of action have been summarized in the table below (Table 2).

Table 2 Summary of PGRN therapeutics and their mechanisms of action

Regardless of the method, multiple concerns with increasing PGRN levels as a therapeutic approach exist. Toxicity from increased PGRN is the primary concern. While necessary for endo-lysosomal health during aging, PGRN in excess could become toxic and contribute to hyperplasia of certain tissues [218,219,220,221]. Furthermore, the AAV vectors must also be tolerated by patients’ immune systems. Poor tolerance of AAV would make some candidate therapies not viable for every patient group. Similarly, off-target effects from small molecule drugs and immunotherapy are possible and must be carefully considered before widespread use in the clinic.

GPNMB as a therapeutic target has not been explored in the field of neurodegeneration, but in oncology, GPNMB has been used as a therapeutic for osteosarcoma and squamous cell carcinoma [222,223,224], but the efficacy of this therapy is questionable [222, 223]. Previous work has shown that GPNMB increases in response to endo-lysosome dysfunction [55, 117], and decreases with the resolution of the endo-lysosomal challenge [55], which is consistent with GPNMB expression as a compensatory mechanism to aid in endo-lysosome dysfunction. This would argue for enhancing GPNMB expression and/or function in vulnerable cell types, perhaps at greater levels or earlier timepoints than biology would eventually reach unaided, rather than decreasing GPNMB. However, care must be taken to avoid unintended immunomodulatory effects given the role of GPNMB in immune cell communication and coordination.

Conclusions and future directions

The main objective of this review was to present the current biological knowledge on PGRN and GPNMB interactions within the context of neuroinflammation and neurodegenerative disease. A critical biological function of both proteins is believed to be their role in lysosomal health. Therefore, it is reasonable to expect that mutations or exposures which reduce the levels of these proteins may contribute to dysfunction of endo-lysosomal traffic which would have a negative impact on effector function in the immune system. If this prediction is correct, it is important to ascertain the mechanisms by which these two genes and their protein products interact to develop sound therapeutics for the clinic.

The fact that dysfunctions in the lysosome and the immune system are considered the main pathological mechanisms contributing to ND makes PGRN and GPNMB exciting and important candidates to study in the context of ND. Indeed, we have reviewed the implications of both PGRN and GPNMB in NDs, including FTD, LSDs, PD, and AD, where they separately have been described to be biomarkers of disease and/or to directly contribute to crucial neurodegenerative processes, such as protein aggregation, neuroinflammation, and neuronal loss. However, the specific role of PGRN–GPNMB interactions at the lysosome to regulate lysosomal health and inflammatory responses and how these contribute to NDs needs further investigation.

Taking this together opens up the possibility that the modulation of PGRN and/or GPNMB could serve as a powerful and effective therapeutic approach against NDs. In this context, different therapies are in place targeting PGRN, where its increased expression has been suggested to be protective. However, little has been explored considering GPNMB as a potential therapeutic venue linked to PGRN deficits. Based on our current and emerging knowledge, we expect that more in-depth knowledge of PGRN/GPNMB interplay will result in more effective mechanism-based therapeutics, where targeting PGRN and GPNMB will impact both neurons and neuroimmune health overall as preventive mechanisms towards neuronal loss.

Availability of data and materials

Not applicable.

Abbreviations

PGRN, Progranulin; mPGRN, mouse:

Progranulin

NDs:

Neurodegenerative diseases

FTD:

Frontotemporal dementia

AD:

Alzheimer’s disease

PD:

Parkinson’s disease

LSDs:

Lysosomal storage disorders

GPNMB:

Glycoprotein non-metastatic B

GRNs:

Granulins

ER:

Endoplasmic reticulum

SLPI:

Secretory lymphocyte precursor inhibitor

NMD:

Nonsense-mediated decay

SRP:

Signal recognition peptide

UTR:

Untranslated region

A9D:

Aspartic acid at position 9

PTM:

Post-translational modifications

uORF:

Upstream open reading frame

miR:

Micro RNAs

ECD:

Extracellular domain

hemITAM:

Half immunoreceptor tyrosine-activation motif

ECF:

Extracellular fragment

sGPNMB:

Soluble GPNMB

APCs:

Antigen-presenting cells

CNS:

Central nervous system

MITF:

Microphthalmia-associated transcription factor

bmMSC:

Bone-marrow mesenchymal stem cells

V-ATPase:

Vacuolar-type-H + ATPase

GCase:

Beta-glucocerebrosidase

HexA:

B-hexosaminidase A

BMP:

Bis(monoacylglycerol)phosphate

ASM:

Acid sphingomyelinase

LPLA2:

Lysosomal phospholipase A2

ALIX:

Apoptosis linked gene 2 interaction protein X

NPC2:

Niemann–Pick disease type C2 protein

Hsp70:

Heat shock protein 70

KO:

Knock-out

WT:

Wild type

NSCs:

Neuronal stem cells

LBP:

Lipopolysaccharide-binding protein

LPS:

Lipopolysaccharide

TNFR1/2:

TNF receptors 1 and 2

YB-1:

Y-box-binding protein

BMDMs:

Bone-marrow-derived macrophages

pNFkB:

Phosphorylated nuclear factor kappa-light-chain enhancer of activated B cells

EMT:

Epithelial–mesenchymal transition

GCS:

Glucosylceramide synthase

NND:

Non-neurological disease

DAM:

Disease-associated associated microglia

NKA:

Na +/K + ATP-ase

bv:

Behavioral variants

PPA:

Primary progressive aphasia

nfvPPA:

Non-fluent variant

svPPA:

Semantic variant

MRI:

Magnetic resonance imaging

NPC:

Niemann–Pick type C

NCLs:

Neuronal ceroid lipofuscinoses

NPC1:

Niemann–Pick disease type C1

SNpc:

Substantia nigra pars compacta

PFFs:

Pre-formed fibrils

DEGs:

Differential expressed genes

AAV:

Adeno-associated virus

GSK:

GlaxoSmithKline

M6PR:

Mannose-6 phosphate receptor

LRP1:

Lipoprotein receptor-related protein 1

References

  1. Dorsey ER, Sherer T, Okun MS, Bloem BR. The emerging evidence of the Parkinson pandemic. J Parkinson’s Dis. 2018;8(s1):S3-8.

    Article  Google Scholar 

  2. Logroscino G, Piccininni M, Marin B, Nichols E, Abd-Allah F, Abdelalim A, et al. Global, regional, and national burden of motor neuron diseases 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018;17(12):1083–97.

    Article  Google Scholar 

  3. Matthews KA, Xu W, Gaglioti AH, Holt JB, Croft JB, Mack D, et al. Racial and ethnic estimates of Alzheimer’s disease and related dementias in the United States (2015–2060) in adults aged ≥65 years. Alzheimer’s & Dementia. 2019;15(1):17–24.

    Article  Google Scholar 

  4. Nichols E, Szoeke CEI, Vollset SE, Abbasi N, Abd-Allah F, Abdela J, et al. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(1):88–106.

    Article  Google Scholar 

  5. Yang W, Hamilton JL, Kopil C, Beck JC, Tanner CM, Albin RL, et al. Current and projected future economic burden of Parkinson’s disease in the U.S. npj Parkinson’s Dis. 2020;6(1).

  6. Nalls MA, Blauwendraat C, Sargent L, Vitale D, Leonard H, Iwaki H, et al. Evidence for GRN connecting multiple neurodegenerative diseases. Brain Commun. 2021;3(2).

  7. Brouwers N, Nuytemans K, Julie Z, Gijselinck I, Engelborghs S, Theuns J, et al. Alzheimer and Parkinson diagnoses in progranulin null mutation carriers in an extended founder family. Arch Neurol. 2007;64(10):1436–46.

    Article  PubMed  Google Scholar 

  8. Brouwers N, Sleegers K, Engelborghs S, Maurer-Stroh S, Gijselinck I, van der Zee J, et al. Genetic variability in progranulin contributes to risk for clinically diagnosed Alzheimer disease. Neurology. 2008;71(9):656–64.

    Article  CAS  PubMed  Google Scholar 

  9. Cortini F, Fenoglio C, Guidi I, Venturelli E, Pomati S, Marcone A, et al. Novel exon 1 progranulin gene variant in Alzheimer’s disease. Eur J Neurol. 2008;15(10):1111–7.

    Article  CAS  PubMed  Google Scholar 

  10. Fenoglio C, Galimberti D, Cortini F, Kauwe JSK, Cruchaga C, Venturelli E, et al. Rs5848 variant influences GRN mRNA levels in brain and peripheral mononuclear cells in patients with Alzheimer’s disease. J Alzheimer’s Dis. 2009;18(3):603–12.

    Article  CAS  Google Scholar 

  11. Kämäläinen A, Viswanathan J, Natunen T, Helisalmi S, Kauppinen T, Pikkarainen M, et al. GRN variant rs5848 reduces plasma and brain levels of granulin in Alzheimer’s disease patients. J Alzheimer’s Dis. 2012;33(1):23–7.

    Article  Google Scholar 

  12. Lee MJ, Chen TF, Cheng TH, Chiu MJ. rs5848 variant of progranulin gene is a risk of Alzheimer’s disease in the Taiwanese population. Neurodegener Dis. 2011;8(4):216–20.

    Article  CAS  PubMed  Google Scholar 

  13. Leverenz JB, Yu CE, Montine TJ, Steinbart E, Bekris LM, Zabetian C, et al. A novel progranulin mutation associated with variable clinical presentation and tau, TDP43 and alpha-synuclein pathology. Brain. 2007;130(5):1360–74.

    Article  CAS  PubMed  Google Scholar 

  14. Yongdui C, Li S, Su L, Sheng J, Wen L, Chen G, et al. Association of progranulin polymorphism rs5848 with neurodegenerative diseases: a meta-analysis. J Neurol. 2015;262(4):814–22.

    Article  Google Scholar 

  15. Murthy M, Blauwendraat C, Ukbec GS, Ipdgc HJ, et al. Increased brain expression of GPNMB is associated with genome wide significant risk for Parkinson’s disease on chromosome 7p15.3. Neurogenetics. 2017;18(3):121–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kumaran R, Cookson MR. Pathways to Parkinsonism Redux: convergent pathobiological mechanisms in genetics of Parkinson’s disease. Hum Mol Genet. 2015;24(R1):R32-44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Huang M, Modeste E, Dammer E, Merino P, Taylor G, Duong DM, 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).

  18. Lui H, Zhang J, Makinson Stefanie R, Cahill Michelle K, Kelley Kevin W, Huang HY, et al. Progranulin deficiency promotes circuit-specific synaptic pruning by microglia via complement activation. Cell. 2016;165(4):921–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hrabal R, Chen Z, James S, Bennett, Ni F. The hairpin stack fold, a novel protein architecture for a new family of protein growth factors. Nat Struct Mol Biol. 1996;3(9):747–52.

    Article  CAS  Google Scholar 

  20. Lee WC, Almeida S, Prudencio M, Caulfield TR, Zhang YJ, Tay WM, et al. Targeted manipulation of the sortilin–progranulin axis rescues progranulin haploinsufficiency. Hum Mol Genet. 2013;23(6):1467–78.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Zhou X, Sullivan PM, Sun L, Hu F. The interaction between progranulin and prosaposin is mediated by granulins and the linker region between saposin B and C. J Neurochem. 2017;143(2):236–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee CW, Stankowski JN, Chew J, Cook CN, Lam YW, Almeida S, et al. The lysosomal protein cathepsin L is a progranulin protease. Mol Neurodegeneration. 2017;12(1).

  23. Zhou X, Paushter DH, Feng T, Sun L, Reinheckel T, Hu F. Lysosomal processing of progranulin. Mol Neurodegeneration. 2017;12(1).

  24. Holler CJ, Taylor G, Deng Q, Kukar T. Intracellular proteolysis of progranulin generates stable, lysosomal granulins that are haploinsufficient in patients with frontotemporal dementia caused by GRN mutations. eneuro. 2017;4(4):ENEURO.0100-17.2017.

  25. Suh HS, Choi N, Tarassishin L, Lee SC. Regulation of progranulin expression in human microglia and proteolysis of progranulin by matrix metalloproteinase-12 (MMP-12). PLoS ONE. 2012;7(4):e35115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Guo FJ, Lai Y, Tian Q, Lin E, Kong L, Liu C. Granulin-epithelin precursor (GEP) binds directly to ADAMTS-7 and ADAMTS-12 and inhibits their degradation of cartilage oligomeric matrix protein. Arthritis Rheumatism. 2010;62(7):2023.

    Article  CAS  PubMed  Google Scholar 

  27. Kessenbrock K, Fröhlich LF, Sixt M, Lämmermann T, Pfister H, Bateman A, et al. Proteinase 3 and neutrophil elastase enhance inflammation in mice by inactivating antiinflammatory progranulin. J Clin Invest. 2008;118(7).

  28. Zhu J, Nathan C, Jin W, Sim D, Ashcroft GS, Wahl SM, et al. Conversion of proepithelin to epithelins. Cell. 2002;111(6):867–78.

    Article  CAS  PubMed  Google Scholar 

  29. Du H, Zhou X, Feng T, Hu F. Regulation of lysosomal trafficking of progranulin by sortilin and prosaposin. Brain Commun. 2022;4(1).

  30. Nguyen A, Nguyen T, Basar Cenik YuG, Herz J, Walther TC, et al. Secreted progranulin is a homodimer and is not a component of high density lipoproteins (HDL). J Biol Chem. 2013;288(12):8627–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Galimberti D, Fumagalli G, Fenoglio C, Cioffi S, Arighi A, Serpente M, et al. Progranulin plasma levels predict the presence of GRN mutations in asymptomatic subjects and do not correlate with brain atrophy: results from the GENFI study. Neurobiol Aging. 2018;62(245):e9-12.

    Google Scholar 

  32. Sellami L, Rucheton B, Younes IB, Camuzat A, Saracino D, Rinaldi D, et al. Plasma progranulin levels for frontotemporal dementia in clinical practice: a 10-year French experience. Neurobiol Aging. 2020;91:167.

    Article  Google Scholar 

  33. Root J, Merino P, Nuckols A, Johnson M, Kukar T. Lysosome dysfunction as a cause of neurodegenerative diseases: lessons from frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis. 2021;154: 105360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yao YN, Wang MD, Tang XC, Wu B, Sun HM. Reduced plasma progranulin levels are associated with the severity of Parkinson’s disease. Neurosci Lett. 2020;725: 134873.

    Article  CAS  PubMed  Google Scholar 

  35. Zhou X, Sun L, Bracko O, Choi JW, Jia Y, Nana AL, et al. Impaired prosaposin lysosomal trafficking in frontotemporal lobar degeneration due to progranulin mutations. Nat Commun. 2017;8(1).

  36. Ong C, He Z, Kriazhev L, Shan X, Roger BA. Regulation of progranulin expression in myeloid cells. Am J Physiol Regul Integr Comp Physiol. 2006;291(6):R1602–12.

    Article  CAS  PubMed  Google Scholar 

  37. Kawashima K, Ishiuchi Y, Konnai M, Komatsu S, Sato H, Kawaguchi H, et al. Glucose deprivation regulates the progranulin-sortilin axis in PC12 cells. FEBS Open Bio. 2016;7(2):149–59.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Menzel L, Kleber L, Friedrich C, Hummel R, Dangel L, Winter J, et al. Progranulin protects against exaggerated axonal injury and astrogliosis following traumatic brain injury. Glia. 2016;65(2):278–92.

    Article  PubMed  Google Scholar 

  39. Chen-Plotkin A, Martinez-Lage M, Patrick, Hu WT, Greene R, Wood EM, et al. Genetic and clinical features of progranulin-associated frontotemporal lobar degeneration. Arch Neurol. 2011;68(4):488–98.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Shankaran SS, Capell A, Hruscha AT, Fellerer K, Neumann M, Schmid B, et al. Missense mutations in the progranulin gene linked to frontotemporal lobar degeneration with ubiquitin-immunoreactive inclusions reduce progranulin production and secretion. J Biol Chem. 2007;283(3):1744–53.

    Article  PubMed  Google Scholar 

  41. Puoti G, Lerza MC, Ferretti MG, Bugiani O, Tagliavini F, Rossi G. A Mutation in the 5′-UTR of GRN gene associated with frontotemporal lobar degeneration: phenotypic variability and possible pathogenetic mechanisms. J Alzheimer’s Dis. 2014;42(3):939–47.

    Article  CAS  Google Scholar 

  42. Capell A, Fellerer K, Haass C. Progranulin transcripts with short and long 5′ untranslated regions (UTRs) are differentially expressed via posttranscriptional and translational repression. J Biol Chem. 2014;289(37):25879–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Banzhaf-Strathmann J, Claus R, Mücke O, Rentzsch K, van der Zee J, Engelborghs S, et al. Promoter DNA methylation regulates progranulin expression and is altered in FTLD. Acta Neuropathol Commun. 2013;1(1).

  44. Jiao J, Herl L, Farese RV, Gao FB. MicroRNA-29b regulates the expression level of human progranulin, a secreted glycoprotein implicated in frontotemporal dementia. PLoS ONE. 2010;5(5):e10551–61.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Rademakers R, Eriksen JL, Baker M, Robinson T, Ahmed Z, Lincoln S, et al. Common variation in the miR-659 binding-site of GRN is a major risk factor for TDP43-positive frontotemporal dementia. Hum Mol Genet. 2008;17(23):3631–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wang WX, Wilfred BR, Madathil SK, Tang G, Hu Y, Dimayuga J, et al. miR-107 regulates granulin/progranulin with implications for traumatic brain injury and neurodegenerative disease. Am J Pathol. 2010;177(1):334–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Weterman MAJ, Ajubi N, van Dinter IMR, Degen WGJ, van Muijen GNP, Ruiter DJ, et al. nmb, a novel gene, is expressed in low-metastatic human melanoma cell lines and xenografts. Int J Cancer. 1995;60(1):73–81.

    Article  CAS  PubMed  Google Scholar 

  48. Liguori M, Digifico E, Vacchini A, Avigni R, Colombo F, Borroni EM, et al. The soluble glycoprotein NMB (GPNMB) produced by macrophages induces cancer stemness and metastasis via CD44 and IL-33. Cell Mol Immunol. 2020;18(3):711–22.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Rose AAN, Pepin F, Russo C, Khalil JG, Hallett M, Siegel PH. Osteoactivin promotes breast cancer metastasis to bone. Mol Cancer Res. 2007;5(10):1001–14.

    Article  CAS  PubMed  Google Scholar 

  50. Rose AAN, Grosset AA, Dong Z, Russo C, MacDonald PA, Bertos NR, et al. Glycoprotein nonmetastatic B is an independent prognostic indicator of recurrence and a novel therapeutic target in breast cancer. Clin Cancer Res. 2010;16(7):2147–56. Available from: https://aacrjournals.org/clincancerres/article/16/7/2147/76148/Glycoprotein-Nonmetastatic-B-Is-an-Independent.

  51. UniProt. www.uniprot.org. Available from: https://www.uniprot.org/uniprotkb/Q14956/entry.

  52. Kuan CT, Wakiya K, Dowell JM, Herndon JE, Reardon DA, Graner MW, et al. Glycoprotein nonmetastatic melanoma protein B, a potential molecular therapeutic target in patients with glioblastoma multiforme. Clin Cancer Res. 2006;12(7):1970–82.

    Article  CAS  PubMed  Google Scholar 

  53. Hoashi T, Sato S, Yamaguchi Y, Passeron T, Tamaki K, Hearing VJ. Glycoprotein nonmetastatic melanoma protein b, a melanocytic cell marker, is a melanosome-specific and proteolytically released protein. The FASEB J. 2010;24(5):1616–29.

    Article  CAS  PubMed  Google Scholar 

  54. Rose AAN, Annis MG, Dong Z, Pepin F, Hallett M, Park M, et al. ADAM10 releases a soluble form of the GPNMB/osteoactivin extracellular domain with angiogenic properties. PLoS ONE. 2010;5(8):e12093.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Kramer GJ, Wegdam W, Donker-Koopman WE, Ottenhoff R, Gaspar P, Verhoek M, et al. Elevation of glycoprotein nonmetastatic melanoma protein B in type 1 Gaucher disease patients and mouse models. FEBS Open Bio. 2016;6(9):902–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lin J, Zhang P, Huang Y, Wei X, Guo D, Liu J, et al. Elevated circulating Gpnmb levels are associated with hyperthyroidism. Endocr Connect. 2020;9(8):783–92.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Conway BR, Manoharan D, Manoharan D, Jenks S, Dear JW, McLachlan S, et al. Measuring urinary tubular biomarkers in type 2 diabetes does not add prognostic value beyond established risk factors. Kidney Int. 2012;82(7):812–8.

    Article  CAS  PubMed  Google Scholar 

  58. Murugesan V, Liu J, Yang R, Lin H, Lischuk A, Pastores GM, et al. Validating glycoprotein non-metastatic melanoma B (gpNMB, osteoactivin), a new biomarker of Gaucher disease. Blood Cells Mol Dis. 2018;1(68):47–53.

    Article  Google Scholar 

  59. Tanaka H, Shimazawa M, Kimura M, Takata M, Tsuruma K, Yamada M, et al. The potential of GPNMB as novel neuroprotective factor in amyotrophic lateral sclerosis. Sci Rep. 2012;2(1).

  60. Zigdon H, Savidor A, Levin Y, Meshcheriakova A, Schiffmann R, Futerman AH. Identification of a biomarker in cerebrospinal fluid for neuronopathic forms of gaucher disease. PLoS ONE. 2015;10(3):e0120194.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ripoll VM, Irvine KM, Ravasi T, Sweet MJ, Hume DA. Gpnmb is induced in macrophages by IFN-γ and lipopolysaccharide and acts as a feedback regulator of proinflammatory responses. J Immunol. 2007;178(10):6557–66.

    Article  CAS  PubMed  Google Scholar 

  62. Zhou L, Zhuo H, Ouyang H, Liu Y, Yuan F, Sun L, et al. Glycoprotein non-metastatic melanoma protein b (Gpnmb) is highly expressed in macrophages of acute injured kidney and promotes M2 macrophages polarization. Cell Immunol. 2017;316:53–60.

    Article  CAS  PubMed  Google Scholar 

  63. Shi F, Duan S, Cui J, Yan X, Li H, Wang Y, et al. Induction of matrix metalloproteinase-3 (MMP-3) expression in the microglia by lipopolysaccharide (LPS) via upregulation of glycoprotein nonmetastatic melanoma B (GPNMB) expression. J Mol Neurosci. 2014;54(2):234–42.

    Article  CAS  PubMed  Google Scholar 

  64. Aichholzer F, Klafki HW, Ogorek I, Vogelgsang J, Wiltfang J, Scherbaum N, et al. Evaluation of cerebrospinal fluid glycoprotein NMB (GPNMB) as a potential biomarker for Alzheimer’s disease. Alzheimer’s Res Therapy. 2021;13(1).

  65. Gabriel TL, Tol MJ, Ottenhof R, van Roomen C, Aten J, Claessen N, et al. Lysosomal stress in obese adipose tissue macrophages contributes to MITF-dependent gpnmb induction. Diabetes. 2014;63(10):3310–23.

    Article  CAS  PubMed  Google Scholar 

  66. Gutknecht M, Geiger J, Joas S, Dörfel D, Salih HR, Müller M, et al. The transcription factor MITF is a critical regulator of GPNMB expression in dendritic cells. Cell Commun Signaling. 2015;13(1).

  67. Ripoll VM, Meadows NA, Raggatt LJ, Chang MK, Pettit AR, Cassady AI, et al. Microphthalmia transcription factor regulates the expression of the novel osteoclast factor GPNMB. Gene. 2008;413(1–2):32–41.

    Article  CAS  PubMed  Google Scholar 

  68. Hartman ML, Czyz M. MITF in melanoma: mechanisms behind its expression and activity. Cell Mol Life Sci. 2014;72(7):1249–60.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Jiang SS, Chen CH, Tseng KY, Tsai FY, Wang MJ, Chang IS, et al. Gene expression profiling suggests a pathological role of human bone marrow-derived mesenchymal stem cells in aging-related skeletal diseases. Aging. 2011;3(7):672–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Krasniewski LK, Chakraborty P, Cui CY, Mazan-Mamczarz K, Dunn CA, Piao Y, et al. Single-cell analysis of skeletal muscle macrophages reveals age-associated functional subpopulations. eLife. 2022;11.

  71. Suda M, Shimizu I, Katsuumi G, Yoshida Y, Hayashi Y, Ikegami R, et al. Senolytic vaccination improves normal and pathological age-related phenotypes and increases lifespan in progeroid mice. Nat Aging. 2021;1–10.

  72. Hasan S, Fernandopulle MS, Humble SW, Frankenfield AM, Li H, Prestil R, et al. Multi-modal proteomic characterization of lysosomal function and proteostasis in progranulin-deficient neurons. bioRxiv (Cold Spring Harbor Laboratory). 2023.

  73. Tanaka Y, Suzuki G, Matsuwaki T, Hosokawa M, Serrano G, Beach TG, et al. Progranulin regulates lysosomal function and biogenesis through acidification of lysosomes. Hum Mol Genet. 2017;26(5):ddx011.

    Article  Google Scholar 

  74. Beel S, Moisse M, Damme M, De Muynck L, Robberecht W, Van Den Bosch L, et al. Progranulin functions as a cathepsin D chaperone to stimulate axonal outgrowth in vivo. Hum Mol Genet. 2017;26(15):2850–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Butler V, Cortopassi WA, Argouarch A, Ivry SL, Craik CS, Jacobson MP, et al. Progranulin stimulates the in vitro maturation of pro-cathepsin D at Acidic pH. J Mol Biol. 2019;431(5):1038–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Valdez C, Wong YC, Schwake M, Bu G, Wszolek ZK, Krainc D. Progranulin-mediated deficiency of cathepsin D results in FTD and NCL-like phenotypes in neurons derived from FTD patients. Hum Mol Genet. 2017;26(24):4861–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhou X, Paushter DH, Feng T, Pardon CM, Mendoza CS, Hu F. Regulation of cathepsin D activity by the FTLD protein progranulin. Acta Neuropathol. 2017;134(1):151–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Arrant AE, Roth JR, Boyle NR, Kashyap SN, Hoffmann MQ, Murchison CF, et al. Impaired β-glucocerebrosidase activity and processing in frontotemporal dementia due to progranulin mutations. Acta Neuropathol Commun. 2019;7(1).

  79. Valdez C, Ysselstein D, Young T, Zheng J, Krainc D. Progranulin mutations result in impaired processing of prosaposin and reduced glucocerebrosidase activity. Hum Mol Genet. 2019;29(5):716–26.

    Article  PubMed Central  Google Scholar 

  80. Zhou X, Paushter DH, Pagan MD, Kim D, Santos MN, Lieberman RL, et al. Progranulin deficiency leads to reduced glucocerebrosidase activity. PLoS ONE. 2019;14(7):e02123822.

    Article  Google Scholar 

  81. Chen Y, Jian J, Hettinghouse A, Zhao X, Setchell KDR, Sun Y, et al. Progranulin associates with hexosaminidase A and ameliorates GM2 ganglioside accumulation and lysosomal storage in Tay-Sachs disease. J Mol Med. 2018;96(12):1359–73.

    Article  CAS  PubMed  Google Scholar 

  82. Boland S, Swarup S, Ambaw YA, Malia PC, Richards RC, Fischer AW, et al. Deficiency of the frontotemporal dementia gene GRN results in gangliosidosis. Nat Commun. 2022;13(1):5924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Evers BM, Rodriguez-Navas C, Tesla RJ, Prange-Kiel J, Wasser CR, Yoo KS, et al. Lipidomic and transcriptomic basis of lysosomal dysfunction in progranulin deficiency. Cell Rep. 2017;20(11):2565–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Vandevrede L, Rojas JC, Wang P, Heuer HW, Karydas AM, Ljubenkov PA, et al. Lipid metabolism dysfunction in progranulin mutation carriers: Unbiased metabolomics reveals strong relationship to clinical status in FTLD. Alzheimer’s & Dementia. 2020;16(S5).

  85. Abe A, Shayman JA. The role of negatively charged lipids in lysosomal phospholipase A2 function. J Lipid Res. 2009;50(10):2027–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Remmel N, Locatelli-Hoops S, Breiden B, Schwarzmann G, Sandhoff K. Saposin B mobilizes lipids from cholesterol-poor and bis(monoacylglycero)phosphate-rich membranes at acidic pH. FEBS J. 2007;274(13):3405–20.

    Article  CAS  PubMed  Google Scholar 

  87. Shayman JA, Kelly RJ, Kollmeyer J, He Y, Abe A. Group XV phospholipase A2, a lysosomal phospholipase A2. Prog Lipid Res. 2011;50(1):1–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wilkening G, Linke T, Sandhoff K. Lysosomal degradation on vesicular membrane surfaces. J Biol Chem. 1998;273(46):30271–8.

    Article  CAS  PubMed  Google Scholar 

  89. Chevallier J, Chamoun Z, Jiang G, Prestwich G, Sakai N, Matile S, et al. Lysobisphosphatidic acid controls endosomal cholesterol levels. J Biol Chem. 2008;283(41):27871–80.

    Article  CAS  PubMed  Google Scholar 

  90. Kirkegaard T, Roth AG, Petersen NHT, Mahalka AK, Olsen OD, Moilanen I, et al. Hsp70 stabilizes lysosomes and reverts Niemann-Pick disease-associated lysosomal pathology. Nature. 2010;463(7280):549–53.

    Article  CAS  PubMed  Google Scholar 

  91. Matsuo H, Chevallier J, Mayran N, Blanc IL, Ferguson C, Fauré J, et al. Role of LBPA and alix in multivesicular liposome formation and endosome organization. Science. 2004;303(5657):531–4.

    Article  CAS  PubMed  Google Scholar 

  92. Giray E, Mikkolainen H, Gungor B, Ikonen E, Vattulainen I. Concerted regulation of npc2 binding to endosomal/lysosomal membranes by bis(monoacylglycero)phosphate and sphingomyelin. PLoS Compuat Biol. 2017;13(10):e1005831-1.

    Google Scholar 

  93. Hullin-Matsuda F, Luquain-Costaz C, Bouvier J, Delton-Vandenbroucke I. Bis(monoacylglycero)phosphate, a peculiar phospholipid to control the fate of cholesterol: implications in pathology. Prostaglandins Leukotrienes Essential Fatty Acids. 2009;81(5–6):313–24.

    Article  CAS  PubMed  Google Scholar 

  94. Sun S, Zhou J, Li Z, Wu Y, Wang H, Zheng Q, et al. Progranulin promotes hippocampal neurogenesis and alleviates anxiety-like behavior and cognitive impairment in adult mice subjected to cerebral ischemia. CNS Neurosci Ther. 2022;28(5):775–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Beel S, Herdewyn S, Fazal R, De Decker M, Moisse M, Robberecht W, et al. Progranulin reduces insoluble TDP-43 levels, slows down axonal degeneration and prolongs survival in mutant TDP-43 mice. Mol Neurodegeneration. 2018;13(1).

  96. Philips T, De Muynck L, Thu HNT, Weynants B, Vanacker P, Dhondt J, et al. Microglial upregulation of progranulin as a marker of motor neuron degeneration. J Neuropathol Exp Neurol. 2010;69(12):1191–200.

    Article  CAS  PubMed  Google Scholar 

  97. Irwin D, Lippa CF, Rosso A. Progranulin (PGRN) expression in ALS: an immunohistochemical study. J Neurol Sci. 2009;276(1–2):9–13.

    Article  CAS  PubMed  Google Scholar 

  98. Ljubenkov PA, Miller ZA, Mumford P, Zhang J, Allen Isabel Elaine, Mitic LL, et al. Peripheral innate immune activation correlates with disease severity in GRN haploinsufficiency. Front Neurol. 2019;10.

  99. Yin F, Banerjee R, Thomas B, Zhou P, Qian L, Jia T, et al. Exaggerated inflammation, impaired host defense, and neuropathology in progranulin-deficient mice. J Exp Med. 2009;207(1):117–28.

    Article  PubMed  Google Scholar 

  100. Houser MC, Uriarte Huarte O, Wallings R, Keating CE, MacPherson KP, Herrick MK, et al. Progranulin loss results in sex-dependent dysregulation of the peripheral and central immune system. Front Immunol. 2022;22:13.

    Google Scholar 

  101. Suzuki M, Lee HC, Kayasuga Y, Chiba S, Nedachi T, Matsuwaki T, et al. Roles of progranulin in sexual differentiation of the developing brain and adult neurogenesis. J Reprod Dev. 2009;55(4):351–5.

    Article  CAS  PubMed  Google Scholar 

  102. Curtis AF, Masellis M, Hsiung GYR, Moineddin R, Zhang K, Au B, et al. Sex differences in the prevalence of genetic mutations in FTD and ALS: a meta-analysis. Neurology. 2017;89(15):1633–42.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Ferretti MT, Iulita MF, Cavedo E, Chiesa PA, Schumacher Dimech A, Santuccione Chadha A, et al. Sex differences in Alzheimer disease—the gateway to precision medicine. Nat Rev Neurol. 2018;14(8):457–69.

    Article  PubMed  Google Scholar 

  104. Reekes TH, Higginson CI, Ledbetter CR, Sathivadivel N, Zweig RM, Disbrow EA. Sex specific cognitive differences in Parkinson disease. npj Parkinson’s Dis. 2020;6(1).

  105. Tang W, Lu Y, Tian QY, Zhang Y, Guo FJ, Liu GY, et al. The growth factor progranulin binds to TNF receptors and is therapeutic against inflammatory arthritis in mice. Science. 2011;332(6028):478–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Chen X, Chang J, Deng Q, Xu J, Nguyen TA, Martens LH, et al. Progranulin does not bind tumor necrosis factor (TNF) receptors and is not a direct regulator of TNF-dependent signaling or bioactivity in immune or neuronal cells. J Neurosci. 2013;33(21):9202–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Etemadi N, Webb A, Bankovacki A, Silke J, Nachbur U. Progranulin does not inhibit TNF and lymphotoxin-α signalling through TNF receptor 1. Immunol Cell Biol. 2013;91(10):661–4.

    Article  CAS  PubMed  Google Scholar 

  108. Lang I, Füllsack S, Wajant H. Lack of evidence for a direct interaction of progranulin and tumor necrosis factor receptor-1 and tumor necrosis factor receptor-2 from cellular binding studies. Front Immunol. 2018;23:9.

    Google Scholar 

  109. Hessman CL, Hildebrandt J, Shah A, Brandt S, Bock A, Frye BC, et al. YB-1 interferes with TNFα–TNFR binding and modulates progranulin-mediated inhibition of TNFα signaling. Int J Mol Sci. 2020;21(19):7076–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Frye BC, Halfter S, Djudjaj S, Muehlenberg P, Weber S, Raffetseder U, et al. Y-box protein-1 is actively secreted through a non-classical pathway and acts as an extracellular mitogen. EMBO Rep. 2009;10(7):783–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Rauen T, Raffetseder U, Frye B, Djudjaj S, Mühlenberg PJT, Eitner F, et al. YB-1 acts as a ligand for notch-3 receptors and modulates receptor activation. J Biol Chem. 2009;284(39):26928–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Li B, Castano AP, Hudson TE, Nowlin BT, Lin SL, Bonventre JV, et al. The melanoma-associated transmembrane glycoprotein Gpnmb controls trafficking of cellular debris for degradation and is essential for tissue repair. FASEB J. 2010;24(12):4767–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Okita Y, Kimura M, Xie R, Chen C, Shen LTW, Kojima Y, et al. The transcription factor MAFK induces EMT and malignant progression of triple-negative breast cancer cells through its target GPNMB. Sci Signaling. 2017;10(474).

  114. Marconi GD, Fonticoli L, Rajan TS, Pierdomenico SD, Trubiani O, Pizzicannella J, et al. Epithelial-mesenchymal transition (EMT): the Type-2 EMT in wound healing, tissue regeneration and organ fibrosis. Cells. 2021;10(7):1587.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Xie R, Okita Y, Ichikawa Y, Muhammad AF, Tuyen K, PhuongDung ST, et al. Role of the kringle-like domain in glycoprotein NMB for its tumorigenic potential. Cancer Sci. 2019;110(7):2237–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Bosco A, Steele MR, Vetter ML. Early microglia activation in a mouse model of chronic glaucoma. J Comp Neurol. 2011;519(4):599–620.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Marshall J, Nietupski JB, Park H, Cao J, Bangari DS, Silvescu C, et al. Substrate reduction therapy for sandhoff disease through inhibition of glucosylceramide synthase activity. Mol Ther. 2019;27(8):1495–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Di Liberto G, Egervari K, Kreutzfeldt M, Schürch CM, Hewer E, Wagner I, et al. Neurodegenerative phagocytes mediate synaptic stripping in Neuro-HIV. Brain. 2022;145(8):2730.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Ariizumi K, Chung JS, Dougherty I, Cruz P. Function of DC-HIL on T cell activation syndecan-4 mediates the coinhibitory. J Immunol References. 2007;179(9):5778–84.

    Google Scholar 

  120. Chung J, Sato K, Dougherty I, Cruz PD, Ariizumi K. DC-HIL is a negative regulator of T lymphocyte activation. Blood. 2007;109(10):4320–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Ramani V, Chung JS, Ariizumi K, Cruz PD. Soluble DC-HIL/Gpnmb modulates T-lymphocyte extravasation to inflamed skin. J Investig Dermatol. 2022;142(5):1372–80.

    Article  CAS  PubMed  Google Scholar 

  122. Prabata A, Ikeda K, Rahardini EP, Hirata KI, Emoto N. GPNMB plays a protective role against obesity-related metabolic disorders by reducing macrophage inflammatory capacity. J Biol Chem. 2021;297(5): 101232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Neal ML, Boyle AM, Budge KM, Safadi FF, Richardson JR. The glycoprotein GPNMB attenuates astrocyte inflammatory responses through the CD44 receptor. J Neuroinflammation. 2018;15(1).

  124. Ono Y, Kazuhiro Tsuruma, Takata M, Masamitsu Shimazawa, Hara H. Glycoprotein nonmetastatic melanoma protein B extracellular fragment shows neuroprotective effects and activates the PI3K/Akt and MEK/ERK pathways via the Na+/K+-ATPase. Sci Rep. 2016;6(1).

  125. Ward ME, Chen R, Huang HY, Ludwig C, Telpoukhovskaia M, Taubes A, et al. Individuals with progranulin haploinsufficiency exhibit features of neuronal ceroid lipofuscinosis. Sci Transl Med. 2017;9(385).

  126. Mackenzie IRA. The neuropathology and clinical phenotype of FTD with progranulin mutations. Acta Neuropathol. 2007;114(1):49–54.

    Article  PubMed  Google Scholar 

  127. Rademakers R, Baker M, Gass J, Adamson J, Huey ED, Momeni P, et al. Phenotypic variability associated with progranulin haploinsufficiency in patients with the common 1477C→T (Arg493X) mutation: an international initiative. Lancet Neurol. 2007;6(10):857–68.

    Article  CAS  PubMed  Google Scholar 

  128. Chang MC, Srinivasan K, Friedman BA, Suto E, Modrusan Z, Lee WP, et al. Progranulin deficiency causes impairment of autophagy and TDP-43 accumulation. J Exp Med. 2017;214(9):2611–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Ward ME, Taubes A, Chen R, Miller BL, Sephton CF, Gelfand JM, et al. Early retinal neurodegeneration and impaired Ran-mediated nuclear import of TDP-43 in progranulin-deficient FTLD. J Exp Med. 2014;211(10):1937–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Marques ARA, Gabriel TL, Aten J, van Roomen CPAA, Ottenhoff R, Claessen N, et al. Gpnmb is a potential marker for the visceral pathology in niemann-pick type C disease. PLoS ONE. 2016;11(1):e0147208.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Platt FM, d’Azzo A, Davidson BL, Neufeld EF, Tifft CJ. Lysosomal storage diseases. Nat Rev Dis Primers. 2018;4(1).

  132. La Cognata V, Guarnaccia M, Polizzi A, Ruggieri M, Cavallaro S. Highlights on genomics applications for lysosomal storage diseases. Cells. 2020;9(8).

  133. Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature. 2006;442(7105):916–9.

    Article  CAS  PubMed  Google Scholar 

  134. Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, et al. Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature. 2006;442(7105):920–4.

    Article  CAS  PubMed  Google Scholar 

  135. Sassi C, Capozzo R, Hammer M, Zecca C, Federoff M, Cornelis Blauwendraat, et al. Exploring dementia and neuronal ceroid lipofuscinosis genes in 100 FTD-like patients from 6 towns and rural villages on the Adriatic Sea cost of Apulia. Sci Rep. 2021;11(1).

  136. Smith Katherine R, Damiano J, Franceschetti S, Carpenter S, Canafoglia L, Morbin M, et al. Strikingly different clinicopathological phenotypes determined by progranulin-mutation dosage. Am J Human Genet. 2012;90(6):1102–7.

    Article  CAS  Google Scholar 

  137. Benussi L, Binetti G, Sina E, Gigola L, Meitinger T, Ghidoni R. A novel deletion in progranulin gene is associated with FTDP-17 and CBS. Neuobiol Aging. 2008;29(3):427–35.

    Article  CAS  Google Scholar 

  138. Le Ber I, Camuzat A, Hannequin D, Pasquier F, Guedj E, Rovelet-Lecrux A, et al. Phenotype variability in progranulin mutation carriers: a clinical, neuropsychological, imaging and genetic study. Brain J Neurol. 2008;131(Pt 3):732–46.

    Article  Google Scholar 

  139. Yu CE, Bird TD, Bekris LM, Montine TJ, Leverenz JB, Steinbart E, et al. The spectrum of mutations in progranulin. Arch Neurol. 2010;67(2).

  140. Almeida MR, Macário MC, Ramos L, Baldeiras I, Ribeiro MH, Santana I. Portuguese family with the co-occurrence of frontotemporal lobar degeneration and neuronal ceroid lipofuscinosis phenotypes due to progranulin gene mutation. Neurobiol Aging. 2016;41(200):e1-5.

    Google Scholar 

  141. Bossolasco P, Cimini S, Maderna E, Bardelli D, Canafoglia L, Cavallaro T, et al. GRN-/- iPSC-derived cortical neurons recapitulate the pathological findings of both frontotemporal lobar degeneration and neuronal ceroidolipofuscinosis. Neurobiol Dis. 2022;175:105891.

    Article  CAS  PubMed  Google Scholar 

  142. Siintola E. Cathepsin D deficiency underlies congenital human neuronal ceroid-lipofuscinosis. Brain. 2006;129(6):1438–45.

    Article  PubMed  Google Scholar 

  143. Steinfeld R, Reinhardt K, Schreiber K, Hillebrand M, Kraetzner R, Brück W, et al. Cathepsin D deficiency is associated with a human neurodegenerative disorder. Am J Human Genet. 2006;78(6):988–98.

    Article  CAS  Google Scholar 

  144. Kyttälä A, Lahtinen U, Braulke T, Hofmann SL. Functional biology of the neuronal ceroid lipofuscinoses (NCL) proteins. Biochimica et Biophysica Acta (BBA) Mol Basis Dis. 2006;1762(10):920–33.

    Article  Google Scholar 

  145. Ahmed Z, Sheng H, Xu Y, Lin WL, Innes AE, Gass J, et al. Accelerated lipofuscinosis and ubiquitination in granulin knockout mice suggest a role for progranulin in successful aging. Am J Pathol. 2010;177(1):311–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Ghoshal N, Dearborn JT, Wozniak DF, Cairns NJ. Core features of frontotemporal dementia recapitulated in progranulin knockout mice. Neurobiol Dis. 2012;45(1):395–408.

    Article  CAS  PubMed  Google Scholar 

  147. Petkau TL, Hill A, Leavitt BR. Core neuropathological abnormalities in progranulin-deficient mice are penetrant on multiple genetic backgrounds. Neuroscience. 2016;315:175–95.

    Article  CAS  PubMed  Google Scholar 

  148. Yin F, Dumont M, Banerjee R, Ma Y, Li H, Lin MT, et al. Behavioral deficits and progressive neuropathology in progranulin-deficient mice: a mouse model of frontotemporal dementia. FASEB J. 2010;24(12):4639–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Wils H, Kleinberger G, Pereson S, Janssens J, Capell A, Van Dam D, et al. Cellular ageing, increased mortality and FTLD-TDP-associated neuropathology in progranulin knockout mice. J Pathol. 2012;228(1):67–76.

    Article  CAS  PubMed  Google Scholar 

  150. Martens LH, Zhang J, Barmada SJ, Zhou P, Kamiya S, Sun B, et al. Progranulin deficiency promotes neuroinflammation and neuron loss following toxin-induced injury. J Clin Investig. 2012;122(11):3955–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Arrant AE, Filiano AJ, Warmus BA, Hall AM, Roberson ED. Progranulin haploinsufficiency causes biphasic social dominance abnormalities in the tube test. Genes Brain Behav. 2016;15(6):588–603.

    Article  CAS  PubMed  Google Scholar 

  152. Filiano AJ, Martens LH, Young AJ, Warmus BA, Zhou P, Diaz-Ramirez G, et al. Dissociation of frontotemporal dementia-related deficits and neuroinflammation in progranulin haploinsufficient mice. J Neurosci. 2013;33(12):5352–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Nguyen AD, Nguyen TA, Zhang J, Devireddy S, Zhou P, Karydas AM, et al. Murine knockin model for progranulin-deficient frontotemporal dementia with nonsense-mediated mRNA decay. Proc Natl Acad Sci. 2018;115(12).

  154. Frew J, Nygaard HB. Neuropathological and behavioral characterization of aged Grn R493X progranulin-deficient frontotemporal dementia knockin mice. Acta Neuropathol Commun. 2021;9(1).

  155. Klein Z, Takahashi H, Ma M, Stagi M, Zhou M, Lam T, et al. Loss of TMEM106B ameliorates lysosomal and frontotemporal dementia-related phenotypes in progranulin-deficient mice. Neuron. 2017;95(2):281-296.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Petkau TL, Blanco J, Leavitt BR. Conditional loss of progranulin in neurons is not sufficient to cause neuronal ceroid lipofuscinosis-like neuropathology in mice. Neurobiol Dis. 2017;106:14–22.

    Article  CAS  PubMed  Google Scholar 

  157. Arrant AE, Filiano AJ, Patel AR, Hoffmann MQ, Boyle NR, Kashyap S, et al. Reduction of microglial progranulin does not exacerbate pathology or behavioral deficits in neuronal progranulin-insufficient mice. Neurobiol Dis. 2019;1(124):152–62.

    Article  Google Scholar 

  158. Petkau TL, Life B, Lu G, Yang JJ, Fornes O, Wasserman WW, et al. Human progranulin-expressing mice as a novel tool for the development of progranulin-modulating therapeutics. Neurobiol Dis. 2021;1(153):105314–24.

    Article  Google Scholar 

  159. Daniel RL, Daniels E, He Z, Bateman A. Progranulin (acrogranin/PC cell-derived growth factor/granulin-epithelin precursor) is expressed in the placenta, epidermis, microvasculature, and brain during murine development. Dev Dyn. 2003;227(4):593–9.

    Article  CAS  PubMed  Google Scholar 

  160. Daniel R, He Z, Carmichael KP, Halper J, Bateman A. Cellular localization of gene expression for progranulin. J Histochem Cytochem. 2000;48(7):999–1009.

    Article  CAS  PubMed  Google Scholar 

  161. Ahmed Z, Mackenzie IR, Hutton ML, Dickson DW. Progranulin in frontotemporal lobar degeneration and neuroinflammation. J Neuroinflammation. 2007;4(1).

  162. Nelvagal HR, Lange J, Takahashi K, Tarczyluk-Wells MA, Cooper JD. Pathomechanisms in the neuronal ceroid lipofuscinoses. Biochimica et Biophysica Acta (BBA) Mol Basis Dis. 2020;1866(9):165570.

    Article  CAS  Google Scholar 

  163. Rodriguez-Gil JL, Baxter LL, Watkins-Chow DE, Johnson NL, Davidson C, Carlson SR, et al. Transcriptome of HPβCD-treated Niemann-Pick disease type C1 cells highlights GPNMB as a biomarker for therapeutics. Hum Mol Genet. 2021;30(24):2456–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Knopman DS, Amieva H, Petersen RC, Chételat G, Holtzman DM, Hyman BT, et al. Alzheimer disease. Nat Rev Dis Primers. 2021;7(1).

  165. Sheng J, Su L, Xu Z, Chen G. Progranulin polymorphism rs5848 is associated with increased risk of Alzheimer’s disease. Gene. 2014;542(2):141–5.

    Article  CAS  PubMed  Google Scholar 

  166. Xu HM, Tan L, Wan Y, Tan MS, Zhang W, Zheng ZJ, et al. PGRN is associated with late-onset Alzheimer’s disease: a case-control replication study and meta-analysis. Mol Neurobiol. 2016;54(2):1187–95.

    Article  PubMed  Google Scholar 

  167. Mateo I, González-Aramburu I, Pozueta A, Vázquez-Higuera JL, Rodríguez-Rodríguez E, Sánchez-Juan P, et al. Reduced serum progranulin level might be associated with Parkinson’s disease risk. Eur J Neurol. 2013;20(12):1571–3.

    Article  CAS  PubMed  Google Scholar 

  168. Coppola G, Karydas A, Rademakers R, Wang Q, Baker M, Hutton M, et al. Gene expression study on peripheral blood identifies progranulin mutations. Ann Neurol. 2008;64(1):92–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Cooper YA, Nachun D, Dokuru D, Yang Z, Karydas AM, Serrero G, et al. Progranulin levels in blood in Alzheimer’s disease and mild cognitive impairment. Ann Clin Transl Neurol. 2018;5(5):616–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Batzu L, Westman E, Pereira JB. Cerebrospinal fluid progranulin is associated with increased cortical thickness in early stages of Alzheimer’s disease. Neurobiol Aging. 2019;88.

  171. Morenas-Rodríguez E, Cervera-Carles L, Vilaplana E, Alcolea D, Carmona-Iragui M, Dols-Icardo O, et al. Progranulin protein levels in cerebrospinal fluid in primary neurodegenerative dementias. J Alzheimer’s Dis. 2016;50(2):539–46.

    Article  Google Scholar 

  172. McGrowder DA, Miller F, Vaz K, Nwokocha C, Wilson-Clarke C, Anderson-Cross M, et al. Cerebrospinal fluid biomarkers of Alzheimer’s disease: current evidence and future perspectives. Brain Sci. 2021;11(2):215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Xu W, Tan CC, Cao XP, Tan L. Neuroinflammation modulates the association of PGRN with cerebral amyloid-β burden. Neurobiol Aging. 2021;103:52–9.

    Article  CAS  PubMed  Google Scholar 

  174. Suárez‐Calvet M, Capell A, Araque Caballero MÁ, Morenas‐Rodríguez E, Fellerer K, Franzmeier N, et al. CSF progranulin increases in the course of Alzheimer’s disease and is associated with sTREM2, neurodegeneration and cognitive decline. EMBO Mol Med. 2018;10(12).

  175. Mao Q, Zheng X, Gefen T, Rogalski E, Spencer C, Rademakers R, et al. FTLD-TDP with and without GRN mutations cause different patterns of CA1 pathology. J Neuropathol Exp Neurol. 2019;78(9):844–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Mendsaikhan A, Tooyama I, Walker DG. Microglial progranulin: involvement in Alzheimer’s disease and neurodegenerative diseases. Cells. 2019;8(3):230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Mendsaikhan A, Tooyama I, Serrano GE, Beach TG, Walker DG. Loss of lysosomal proteins progranulin and prosaposin associated with increased neurofibrillary tangle development in Alzheimer disease. J Neuropathol Exp Neurol. 2021;80(8):741–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Pereson S, Wils H, Kleinberger G, McGowan E, Vandewoestyne M, Van Broeck B, et al. Progranulin expression correlates with dense-core amyloid plaque burden in Alzheimer disease mouse models. J Pathol. 2009;219(2):173–81.

    Article  CAS  PubMed  Google Scholar 

  179. Minami SS, Min SW, Krabbe G, Wang C, Zhou Y, Asgarov R, et al. Progranulin protects against amyloid β deposition and toxicity in Alzheimer’s disease mouse models. Nat Med. 2014;20(10):1157–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Du H, Ying Wong M, Zhang T, Nunez Santos M, Hsu C, Zhang J, et al. A multifaceted role of progranulin in regulating amyloid-beta dynamics and responses. Life Sci Alliance. 2021;4(7):e202000874–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Guan Z, Chen Z, Fu S, Dai L, Shen Y. Progranulin administration attenuates β-amyloid deposition in the hippocampus of 5xFAD mice through modulating BACE1 expression and microglial phagocytosis. Front Cell Neurosci. 2020;18:14.

    Google Scholar 

  182. Kampen V, Kay DG. Progranulin gene delivery reduces plaque burden and synaptic atrophy in a mouse model of Alzheimer’s disease. PLoS ONE. 2017;12(8):e0182896-e182906.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Hosokawa M, Arai T, Masuda-Suzukake M, Kondo H, Matsuwaki T, Nishihara M, et al. Progranulin reduction is associated with increased tau phosphorylation in P301L tau transgenic mice. J Neuropathol Exp Neurol. 2015;74(2):158–65.

    Article  CAS  PubMed  Google Scholar 

  184. Takahashi H, Klein ZA, Bhagat SM, Kaufman AC, Kostylev MA, Ikezu T, et al. Opposing effects of progranulin deficiency on amyloid and tau pathologies via microglial TYROBP network. Acta Neuropathol. 2017;133(5):785–807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Satoh J, Kino Y, Yanaizu M, Ishida T, Saito Y. Microglia express GPNMB in the brains of Alzheimer’s disease and Nasu-Hakola disease. Intractable Rare Dis Res. 2019;8(2):120–8.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Hüttenrauch M, Ogorek I, Klafki H, Otto M, Stadelmann C, Weggen S, et al. Glycoprotein NMB: a novel Alzheimer’s disease associated marker expressed in a subset of activated microglia. Acta Neuropathol Commun. 2018;6(1).

  187. Zhu Z, Liu Y, Li X, Zhang L, Liu H, Cui Y, et al. GPNMB mitigates Alzheimer’s disease and enhances autophagy via suppressing the mTOR signal. Neurosci Lett. 2022;767: 136300.

    Article  CAS  PubMed  Google Scholar 

  188. Wang H, Dey KK, Chen PC, Li Y, Niu M, Cho JH, et al. Integrated analysis of ultra-deep proteomes in cortex, cerebrospinal fluid and serum reveals a mitochondrial signature in Alzheimer’s disease. Mol Neurodegeneration. 2020;15(1).

  189. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al. Parkinson disease. Nat Rev Dis Primers. 2017;3(3):17013.

    Article  PubMed  Google Scholar 

  190. Van Kampen JM, Baranowski D, Kay DG. Progranulin gene delivery protects dopaminergic neurons in a mouse model of Parkinson’s disease. PLoS ONE. 2014;9(5):e97032.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Sarkar S, Dammer EB, Malovic E, Olsen AL, Ali Raza S, Gao T, et al. Molecular signatures of neuroinflammation induced by αsynuclein aggregates in microglial cells. Front Immunol. 2020;31:11.

    Google Scholar 

  192. Moloney EB, Moskites A, Ferrari EJ, Isacson O, Hallett P. The glycoprotein GPNMB is selectively elevated in the substantia nigra of Parkinson’s disease patients and increases after lysosomal stress. Neurobiol Dis. 2018;1(120):1–11.

    Article  Google Scholar 

  193. Smajić S, Prada-Medina CA, Landoulsi Z, Ghelfi J, Delcambre S, Dietrich C, et al. Single-cell sequencing of human midbrain reveals glial activation and a Parkinson-specific neuronal state. Brain J Neurol. 2022;145(3):964–78.

    Article  Google Scholar 

  194. Diaz-Ortiz ME, Seo Y, Posavi M, Carceles Cordon M, Clark E, Jain N, et al. GPNMB confers risk for Parkinson’s disease through interaction with α-synuclein. Science. 2022;377(6608).

  195. Brendza RP, Lin H, Stark KL, Foreman O, Tao J, Pierce AA, et al. Genetic ablation of Gpnmb does not alter synuclein-related pathology. Neurobiol Dis. 2021;1(159):105494–504.

    Article  Google Scholar 

  196. Budge KM, Neal ML, Richardson JR, Safadi FF. Transgenic overexpression of GPNMB protects against MPTP-induced neurodegeneration. Mol Neurobiol. 2020;57(7):2920–33.

    Article  CAS  PubMed  Google Scholar 

  197. Ma Y, Matsuwaki T, Keitaro Yamanouchi K, Nishihara M. Progranulin protects hippocampal neurogenesis via suppression of neuroinflammatory responses under acute immune stress. Mol Neurobiol. 2016;54(5):3717–28.

    Article  PubMed  Google Scholar 

  198. Takahashi K, Nakamura S, Otsu W, Shimazawa M, Hara H. Progranulin deficiency in Iba-1+ myeloid cells exacerbates choroidal neovascularization by perturbation of lysosomal function and abnormal inflammation. J Neuroinflammation. 2021;18(1).

  199. Götzl JK, Mori K, Damme M, Fellerer K, Tahirovic S, Kleinberger G, et al. Common pathobiochemical hallmarks of progranulin-associated frontotemporal lobar degeneration and neuronal ceroid lipofuscinosis. Acta Neuropathol. 2014.

  200. Hafler BP, Klein ZA, Zhou Z, Strittmatter SM. Progressive retinal degeneration and accumulation of autofluorescent lipopigments in Progranulin deficient mice. Brain Res. 2014;1(1588):168–74.

    Article  Google Scholar 

  201. Tanaka Y, Chambers JK, Matsuwaki T, Yamanouchi K, Nishihara M. Possible involvement of lysosomal dysfunction in pathological changes of the brain in aged progranulin-deficient mice. Acta Neuropathol Commun. 2014;2(1).

  202. Götzl JK, Colombo AV, Fellerer K, Reifschneider A, Werner G, Tahirovic S, et al. Early lysosomal maturation deficits in microglia triggers enhanced lysosomal activity in other brain cells of progranulin knockout mice. Mol Neurodegeneration. 2018;13(1).

  203. Wang C, Zhang L, Ndong JDLC, Hettinghouse A, Sun G, Chen C, et al. Progranulin deficiency exacerbates spinal cord injury by promoting neuroinflammation and cell apoptosis in mice. J Neuroinflammation. 2019;16(1).

  204. Yu Y, Xu X, Liu L, Mao S, Feng T, Lu Y, et al. Progranulin deficiency leads to severe inflammation, lung injury and cell death in a mouse model of endotoxic shock. J Cell Mol Med. 2016;20(3):506–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Zhang T, Feng T, Wu K, Guo J, Nana AL, Yang G, et al. Progranulin deficiency results in sex-dependent alterations in microglia in response to demyelination. Acta Neuropathol. 2023;146(1):97–119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Du H, Cha Y, Nana AL, Seeley WW, Smolka MB, Hu F. Progranulin inhibits phospholipase sPLA2-IIA to control neuroinflammation. bioRxiv (Cold Spring Harbor Laboratory). 2023;

  207. Logan T, Simon MJ, Rana A, Cherf GM, Srivastava A, Davis SS, et al. Rescue of a lysosomal storage disorder caused by Grn loss of function with a brain penetrant progranulin biologic. Cell. 2021;184(18):4651-4668.e25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Smith DM, Aggarwal G, Niehoff ML, Jones SA, Banerjee S, Farr SA, et al. Biochemical, biomarker, and behavioral characterization of theGrnR493Xmouse model of frontotemporal dementia. bioRxiv (Cold Spring Harbor Laboratory). 2023;

  209. Kayasuga Y, Chiba S, Suzuki M, Kikusui T, Matsuwaki T, Yamanouchi K, et al. Alteration of behavioural phenotype in mice by targeted disruption of the progranulin gene. Behav Brain Res. 2007;185(2):110–8.

    Article  CAS  PubMed  Google Scholar 

  210. Zhang J, Velmeshev D, Hashimoto K, Huang YH, Hofmann JW, Shi X, et al. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency. Nature. 2020;588(7838):459–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Arrant AE, Onyilo VC, Unger DE, Roberson ED. Progranulin gene therapy improves lysosomal dysfunction and microglial pathology associated with frontotemporal dementia and neuronal ceroid lipofuscinosis. J Neurosci. 2018;38(9):2341–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. US National Library of Medicine. www.clinicaltrials.gov. [cited 2023 Sep 6]. Available from: https://www.clinicaltrials.gov/study/NCT05262023?intr=DNL593&rank=1.

  213. Root J, Mendsaikhan A, Nandy S, Taylor G, Wang M, Troiano Araujo L, et al. Granulins rescue inflammation, lysosome dysfunction, and neuropathology in a mouse model of progranulin deficiency. bioRxiv (Cold Spring Harbor Laboratory). 2023;

  214. International Conference on Alzheimer’s and Parkinson’s Diseases 2023 | ALZFORUM . www.alzforum.org. [cited 2023 Sep 7]. Available from: https://www.alzforum.org/print-series/2727996.

  215. Heckman LD, Burstein SR, Brandes A, Wong LC, Yang Z, Lin H, et al. Preclinical development of PR006, a gene therapy for the treatment of frontotemporal dementia with progranulin mutations. Alzheimer’s & Dementia. 2020;16(S2).

  216. United States Library of Medicine. www.clinicaltrials.gov. [cited 2023 Sep 6]. Available from: https://www.clinicaltrials.gov/study/NCT04408625?intr=PR006&rank=1.

  217. United States Libray of Medicine. www.clinicaltrials.gov. [cited 2023 Sep 6]. Available from: https://www.clinicaltrials.gov/study/NCT04747431?intr=PBFT02&rank=1.

  218. Arechavaleta-Velasco F, Perez-Juarez CE, Gerton GL, Diaz-Cueto L. Progranulin and its biological effects in cancer. Med Oncol. 2017;34(12).

  219. Ventura E, Ducci G, Benot Dominguez R, Ruggiero V, Belfiore A, Sacco E, et al. Progranulin oncogenic network in solid tumors. Cancers. 2023;15(6):1706–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Purrahman D, Mahmoudian-Sani MR, Saki N, Wojdasiewicz P, Kurkowska-Jastrzębska I, Poniatowski ŁA. Involvement of progranulin (PGRN) in the pathogenesis and prognosis of breast cancer. Cytokine. 2022;151: 155803.

    Article  CAS  PubMed  Google Scholar 

  221. Yabe K, Yamamoto Y, Takemura M, Hara T, Tsurumi H, Serrero G, et al. Progranulin depletion inhibits proliferation via the transforming growth factor beta/SMAD family member 2 signaling axis in Kasumi-1 cells. Heliyon. 2021;7(1):e05849-e5859.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Khan SA, Sun Z, Dahlberg SE, Malhotra J, Keresztes R, Ikpeazu C, et al. Efficacy and safety of glembatumumab vedotin in patients with advanced or metastatic squamous cell carcinoma of the lung (PrECOG 0504). JTO Clin Res Rep. 2021;2(5):100166–76.

    PubMed  PubMed Central  Google Scholar 

  223. Kopp LM, Malempati S, Krailo M, Gao Y, Buxton A, Weigel BJ, et al. Phase II trial of the glycoprotein non-metastatic B-targeted antibody–drug conjugate, glembatumumab vedotin (CDX-011), in recurrent osteosarcoma AOST1521: a report from the Children’s Oncology Group. Eur J Cancer. 2019;121:177–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Jin R, Jin Y, Tang Y, Yang H, Zhou X, Lei Z. GPNMB silencing suppresses the proliferation and metastasis of osteosarcoma cells by blocking the PI3K/Akt/mTOR signaling pathway. Oncol Rep. 2018;

Download references

Acknowledgements

We thank the Tansey lab for their feedback concerning this review.

Funding

The authors are supported by a Grant from the National Institute of Health and the National Institute of Neurological Disorders and Stroke (RF1NS28800). DAG received financial support from an Alzheimer’s disease and related dementias (ADRD) teaching fellowship funded by the National Institute of Health (T32AG061892) at the University of Florida.

Author information

Authors and Affiliations

Authors

Contributions

DAG, OUH, and MGT designed the manuscript. DAG, RLW, OUH, and MGT wrote and edited the manuscript. DAG and OUH created the figures. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Malú Gámez Tansey.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gillett, D.A., Wallings, R.L., Uriarte Huarte, O. et al. Progranulin and GPNMB: interactions in endo-lysosome function and inflammation in neurodegenerative disease. J Neuroinflammation 20, 286 (2023). https://doi.org/10.1186/s12974-023-02965-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-023-02965-w

Keywords