Volume 158, Issue 2 p. 119-137
REVIEW ARTICLE
Free Access

Progranulin in neurodegenerative dementia

Xiao-Ming Wang

Xiao-Ming Wang

Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China

Search for more papers by this author
Peng Zeng

Peng Zeng

Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China

Search for more papers by this author
Ying-Yan Fang

Ying-Yan Fang

Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, China

Search for more papers by this author
Teng Zhang

Teng Zhang

Department of Neurology, Shanxian Central Hospital, The Affiliated Huxi Hospital of Jining Medical College, Heze, China

Search for more papers by this author
Qing Tian

Corresponding Author

Qing Tian

Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China

Correspondence

Qing Tian, Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.

Email: [email protected]

Search for more papers by this author
First published: 30 April 2021
Citations: 12

Xiao-Ming Wang and Peng Zeng contributed equally to this article.

Abstract

Long-term or severe lack of protective factors is important in the pathogenesis of neurodegenerative dementia. Progranulin (PGRN), a neurotrophic factor expressed mainly in neurons and microglia, has various neuroprotective effects such as anti-inflammatory effects, promoting neuron survival and neurite growth, and participating in normal lysosomal function. Mutations in the PGRN gene (GRN) have been found in several neurodegenerative dementias, including frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). Herein, PGRN deficiency and PGRN hydrolytic products (GRNs) in the pathological changes related to dementia, including aggregation of tau and TAR DNA-binding protein 43 (TDP-43), amyloid-β (Aβ) overproduction, neuroinflammation, lysosomal dysfunction, neuronal death, and synaptic deficit have been summarized. Furthermore, as some therapeutic strategies targeting PGRN have been developed in various models, we highlighted PGRN as a potential anti-neurodegeneration target in dementia.

Abbreviations

  • 5-HT1A
  • serotonin 1A receptor
  • AD
  • Alzheimer's disease
  • ADAMTS7
  • a disintegrin and metalloproteinase with thrombospondin 7
  • ALS
  • amyotrophic lateral sclerosis
  • APP
  • amyloid precursor protein
  • BiP
  • binding immunoglobulin protein
  • CHI3L1
  • chitinase-3-like Protein 1
  • CLEAR
  • coordinated lysosomal expression and regulation
  • CLU
  • clusterin
  • CTSB
  • cathepsin B
  • CTSD
  • cathepsin D
  • CTSL
  • cathepsin L
  • DAM
  • disease-associated microglia
  • DLB
  • dementia with Lewy bodies
  • EphA2
  • ephrin type-A receptor 2
  • ER
  • endoplasmic reticulum
  • FMRP
  • Fragile X mental retardation protein
  • FTLD
  • frontotemporal lobar degeneration
  • GBA
  • glucocerebrosidase
  • GD
  • Gaucher disease
  • GRP94
  • glucose-regulated protein 94
  • HD
  • Huntington's disease
  • HDAC
  • histone deacetylase
  • HSP70
  • heat shock protein 70
  • IL-6
  • interleukin-6
  • iPSCs
  • induced pluripotent stem cells
  • LIMP-2
  • lysosomal integral membrane protein-2
  • LRP1
  • low-density lipoprotein receptor-related protein 1
  • LSD
  • lysosomal storage disease
  • LTP
  • long-term potentiation
  • MCI
  • mild cognitive impairment
  • miRNA
  • microRNAs
  • MMP9
  • matrix metalloproteinase 9
  • MPTP
  • 1-methyl-4-phenyl-1,2,3,6 -tetrahydropyridine
  • mTOR
  • mammalian target of rapamycin
  • NCL
  • neuronal ceroid lipofuscinosis
  • NE
  • neutrophil elastase
  • NFTs
  • neurofibrillary tangles
  • NF-κB
  • nuclear factor κ-B
  • NMDAR
  • endogenous N-methyl-D-aspartate receptor
  • OCD
  • obsessive-compulsive disorder
  • PCDGF
  • PC cell-derived growth factor
  • PD
  • Parkinson’s disease
  • PDI
  • protein disulfide isomerase
  • PGRN
  • Progranulin
  • PHFs
  • paired helical filaments
  • PSAP
  • prosaposin
  • QA
  • quinolinic acid
  • Ripk1
  • receptor-interacting serine/threonine protein kinase 1
  • ROS
  • reactive oxygen species
  • SAHA
  • suberoylanilide hydroxamic acid
  • SCs
  • Schwann cells
  • SNP
  • single nucleotide polymorphism
  • SORT1
  • sortilin
  • SPs
  • senile plaques
  • TBI
  • traumatic brain injury
  • TDP-43
  • TAR DNA-binding protein 43
  • TFEB
  • transcription factor-EB
  • TNF
  • tumor necrosis factor
  • TRAIL-R3
  • TNF-related apoptosis-inducing ligand receptor 3
  • VCAM-1
  • vascular cell adhesion molecule-1
  • 1 INTRODUCTION

    Dementia refers to an acquired clinical syndrome in which a person's cognitive level drops significantly, thereby interfering with his/her ability to perform activities related to occupation, family, social interaction, and daily life. The global prevalence of dementia is estimated to be 7% in the population aged over 65 years, with a slightly higher prevalence (8%–10%) in developed countries (due to their longer life spans in such countries) (Jia et al., 2020; Prince et al., 2013). With the acceleration of population aging, this condition is predicted to bring huge economic burdens to the world, especially in countries with a high prevalence of this condition.

    Dementia is mainly divided into two categories, namely, neurodegenerative and non-neurodegenerative dementia (Gale et al., 2018). The commonly occurring types of neurodegenerative dementia include Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD), dementia with Lewy bodies (DLB), and Parkinson's disease (PD). In addition to exposure to injury factors, the long-term or serious lack of protective factors is also very important in the pathogenesis of neurodegenerative dementia.

    Progranulin (PGRN), also known as proepithelin (PEPI), granulin-epithelin precursor (GEP), acrogranin, or PC cell-derived growth factor (PCDGF), is a glycosylated protein (Bateman & Bennett, 1998) and is expressed mainly in neurons and microglia in the brain (Almeida et al., 2011). In 2006, it was found that haploinsuffiency induced by heterozygous mutations in the GRN gene is one of the main causes of FTLD (Baker et al., 2006; Cruts et al., 2006). This study has prompted the widespread attentions to the clinical importance of PGRN. Now, the important role of PGRN in neurodegenerative dementia has been widely recognized due to its neuroprotective effects such as anti-inflammatory effects (Martens et al., 2012), promoting neuron survival and neurite growth (Van Damme et al., 2008) and participating in normal lysosomal function (Galimberti et al., 2018; Tanaka et al., 2017).

    2 PGRN IN THE BRAIN

    GRN is located on chromosome 17q21.32 and consists of 12 exons, resulting in three isoforms. PGRN contains 593 amino acids and has a molecular weight of 68.5 kDa (88 kDa with N-linked glycosylation). The full-length form of PGRN is comprised of a secretory N-terminal signal peptide of 17 amino acids, and seven full and one half conserved granulin (GRN) domains connected by short linker regions. GRNs are named as follows, based on their location prior to splicing, from the N to the C terminus: paragranulin (P), GRN1 (G), GRN2 (F), GRN3 (B), GRN4 (A), GRN5 (C), GRN6 (D), and GRN7 (E). After post-translational modifications, a proportion of PGRN remains intracellular because it is trafficked to the lysosome via the trans-Golgi network (TGN). Alternatively, it is secreted into the extracellular space through the TGN and secretory vacuoles. In the extracellular space, several proteases including neutrophil elastase (NE), matrix metalloproteinase 9 (MMP9), MMP12, MMP14, and a disintegrin and metalloproteinase with thrombospondin 7 (ADAMTS7) cleave PGRN into GRNs (Mendsaikhan et al., 2019). Secretory leukocyte protease inhibitor (SLPI), primarily produced by astrocytes in the brain (Suh et al., 2012), is reported to bind to PGRN linker sequences directly, and block proteolysis by elastase (Zhu et al., 2002). Additionally, extracellular PGRN can be transported to lysosomes by endocytosis and may be cleaved by cathepsin L (CTSL) in lysosomes (Lee et al., 2017).

    Brain PGRN is localized to neurons, microglia, and the vasculature (Almeida et al., 2011; Daniel et al., 2003). Neuronal PGRN is widely expressed during early development (Daniel et al., 2003) but is limited to specific groups of neurons later, such as cortical and hippocampal pyramidal neurons and Purkinje cells (Daniel et al., 2000). A study in Grn knockout (KO) mice revealed PGRN expression in neurons and microglia, and this expression was particularly strong in the neurons of the cortex, hippocampus (CA1 and CA3 but weaker in the dentate gyrus [DG]), thalamus, and hypothalamus and less intense in the brain stem (including the substantia nigra) and cerebellar Purkinje neurons (Petkau et al., 2010). PGRN is also found in microglia and in the developing cerebral microvasculature (Almeida et al., 2011; Daniel et al., 2003; Suh et al., 2012).

    Brain PGRN levels are affected by aging, hormones, exercise, and hypoxia. Aging promotes the reduction of PGRN mRNA levels in the brain. PGRN mRNA and protein levels decrease in an age-dependent manner in the cortex, hippocampus, and hypothalamus of male mice (Matsuwaki et al., 2011). Sex hormones, exercise, and hypoxia upregulate PGRN expression. It has been reported that androgen treatment during the perinatal period increases PGRN mRNA levels in the neonatal rat hypothalamus (Suzuki et al., 2009) and that estrogen induces PGRN expression in the neonatal rat hypothalamus (Suzuki et al., 2001) and the adult rat hippocampus (Chiba et al., 2007). Hypoxia and exercise were reported to upregulate PGRN expression in human neuroblastoma cell lines (Piscopo et al., 2010), HT22 mouse hippocampal cells (Luo et al., 2014), and hippocampal pyramidal neurons (Asakura et al., 2011). Microglial PGRN expression depends on the activation state of the microglia (Petkau et al., 2010). Disease-associated microglia (DAM) constitute a recently defined late-activation phenotype of microglia in AD. Studies employing single-cell sequencing technology have shown the upregulation of PGRN mRNA levels in DAM derived from AD mice (Deczkowska et al., 2018; Keren-Shaul et al., 2017).

    The translation of GRN is reported to be under the control of receptor-interacting serine/threonine protein kinase 1 (Ripk1), interleukin-6 (IL-6), transcription factor-EB (TFEB), and several microRNAs (miRNAs). Ripk1 has been reported to be a genetic regulator of Grn in Neuro2a cells, microglial-like BV-2 cells, wild-type primary neurons, and Grn-haploinsufficient primary neurons. It increases both intracellular and extracellular PGRN protein levels by increasing the translation rate of PGRN without affecting mRNA levels (Mason et al., 2017). IL-6 stimulates GRN expression in hepatocellular carcinoma and cholangiocarcinoma cells partly by activating extracellular signal-regulated kinase (Erk)/CCAAT/enhancer-binding protein β (C/EBPβ) signaling ( Frampton et al., 2012; Liu et al., 2016). TFEB is implicated in the regulation of GRN expression through its specific recognition and binding to the E-box consensus sequences (5′-CANNTG-3′) in the GRN promoter region (Belcastro et al., 2011), and its overexpression has been shown to be sufficient to enhance PGRN expression (Holler et al., 2016). Inhibition of class I histone deacetylases (HDACs) upregulates PGRN in human neurons by a mechanism involving TFEB occupancy (She et al., 2017). Increased miRNA 29b (Jiao et al., 2010) and miRNA 15/107 (Wang et al., 2010) lead to reduced PGRN protein levels, and reduced miR-659-3p was correlated with an increase in PGRN (Piscopo et al., 2016). Additionally, miR-922, miR-516a-3p, miR-571, miR-548b-5p, and miR-548c-5p may affect the expression of PGRN; these molecules were reported to be significantly dysregulated in the frontal cortex samples of FTLD-TAR DNA-binding protein 43 (TDP-43) (FTLD-TDP) patients with GRN mutations (Kocerha et al., 2011).

    PGRN secretion is activity dependent (Petoukhov et al., 2013). Neuronal activation by 4-aminopyridine or bicucilline has been reported to result in the accumulation of PGRN in the axons and the secretion of PGRN from synapses; these effects were diminished by the inhibition of neuronal excitation by blocking voltage-gated calcium channels (Petoukhov et al., 2013). Additionally, N-glycosylated PGRN was reported to be secreted in exosomes by neuronal cells (Benussi et al., 2016). The endoplasmic reticulum (ER) chaperone network is important for PGRN secretion (Almeida et al., 2011). PGRN binds to the network of ER Ca2+-binding chaperones including binding immunoglobulin protein (BiP), calreticulin, glucose-regulated protein 94 (GRP94), and four members of the protein disulfide isomerase (PDI) family (PDI and the PDI-related proteins ERp57, ERp72, and ERp5). ERp57 and ERp5 promote PGRN secretion (Almeida et al., 2011).

    Extracellular PGRN enters the cells and functions through its receptors. Sortilin 1 (SORT1), a VPS10 family protein that is highly expressed in neurons, acts as a neuronal receptor for PGRN. It rapidly endocytoses and delivers PGRN to lysosomes (Hu et al., 2010; Paushter et al., 2018). Additionally, several other potential PGRN receptors have been reported, including tumor necrosis factor (TNF) receptor (TNFR) (Tang et al., 2011), Toll-like receptor 9 (Park et al., 2011), the prosaposin (PSAP)/cation-independent mannose-6-phosphate receptor (CI-M6PR) complex (Zhou et al., 2015), the PSAP/low-density lipoprotein receptor-related protein 1 (LRP1) complex (Paushter et al., 2018), and the ephrin type-A receptor 2 (EphA2) (Neill et al., 2016).

    3 GRN MUTATIONS IN NEURODEGENERATION

    The brain regions that express PGRN are mainly involved in emotion, memory, and recognition (Petkau et al., 2010). PGRN deficiency, usually caused by GRN mutations, contributes to neurodegeneration in these brain regions.

    3.1 GRN mutations in neurodegenerative diseases

    3.1.1 FTLD

    Heterozygous GRN mutations lead to PGRN haploinsufficiency in FTLD, which is a clinically incurable heterogeneous neurodegenerative disease characterized by progressive atrophy of the frontal and temporal lobes. FTLD patients often present with progressive deficits in behavior, executive function, or language (Xu et al., 2019). The inclusion bodies in FTLD neurons have different histopathological features, for example, positive for tau protein (FTLD-Tau), ubiquitinated TDP-43 (FTLD-TDP), or fused in sarcoma protein (FTLD-FUS) (Bang et al., 2015; Mackenzie & Neumann, 2016). GRN mutations are primarily associated with the FTLD-TDP subtype (Finch et al., 2009; Schymick et al., 2007). More than 60 disease-causing GRN mutations have been identified (Mackenzie & Neumann, 2016), accounting for 20%–25% of familial FTLD cases and about 10% of all FTLD cases, and all reduce PGRN levels or result in loss of PGRN function (Paushter et al., 2018; Schymick et al., 2007). FTLD is the second leading cause of early onset dementia after AD, particularly in patients younger than 65 years of age (Bang et al., 2015).

    3.1.2 AD

    AD is the most common type of senile dementia and is characterized by the deposition of extracellular senile plaques (SPs) containing Aβ, formation of intracellular neurofibrillary tangles (NFTs) consisting of hyperphosphorylated tau, and progressive neuron losses in the memory-associated brain regions (Tan et al., 2019; Wright & Harding, 2019). GRN mutations associated with reduced PGRN levels may be linked to AD (Perry et al., 2013). A single nucleotide polymorphism (SNP) located in the 3′-untranslated region (3′-UTR) of GRN designated as rs5848 has been found to cause AD (Sheng et al., 2014; Xu et al., 2017). Additionally, other SNPs such as rs4792939 (located in intron 2) and rs850713 (located in intron 5) have been shown to increase the susceptibility for developing AD (Jing et al., 2016; Viswanathan et al., 2009).

    3.1.3 Neuronal ceroid lipofuscinosis (NCL)

    NCL is a progressive neurodegenerative disease usually seen in children with cognitive decline, progressive cerebellar atrophy, retinopathy, and myoclonic epilepsy. Despite having diverse underlying biochemical etiologies, NCL is thought to result from the excessive accumulation of autofluorescent storage material (lipofuscin) in neurons and other cells (Nita et al., 2016). Homozygous GRN mutations lead to complete PGRN loss and cause NCL (Smith et al., 2012). Furthermore, it was confirmed that PGRN haploinsufficiency in FTLD cases with a GRN mutation leads to NCL-like features in humans, some occurring before the onset of dementia (Almeida et al., 2016; Götzl et al., 2014; Valdez et al., 2017; Ward et al., 2017); this supports the contention that lysosomal dysfunction is a key pathophysiological process in GRN-associated FTLD and GRN-associated NCL.

    3.1.4 Hippocampal sclerosis of aging (HS-Aging)

    HS-Aging is a common neurodegenerative condition associated with dementia and is diagnosed post-mortem when neuron loss and astrocytosis are observed in the hippocampus, independent of SPs and NFTs (Nelson et al., 2013). HS-Aging affects up to 25% of the ‘oldest-old’ persons and is associated with substantial cognitive impairments (Nelson et al., 2011, 2013). TDP-43 pathology is a pathological biomarker of HS-Aging (Amador-Ortiz, Lin, et al., 2007). Two SNPs associated with HS-Aging are linked to FTLD-TDP, namely, the GRN gene variants rs5848 and rs1990622 (near TMEM106B) (Dickson et al., 2010; Murray et al., 2014). The status of the SNP rs5848 (which confers GRN risk) correlates with variations in cerebrospinal fluid (CSF) proteins (e.g., increased tau), with the risk allele (T) associated with increased levels of AXL receptor tyrosine kinase (AXL), TNF-related apoptosis-inducing ligand receptor 3 (TRAIL-R3), vascular cell adhesion molecule-1 (VCAM-1), and clusterin (CLU) (Fardo et al., 2017).

    3.1.5 Amyotrophic lateral sclerosis (ALS) with dementia (ALS-D)

    ALS is also known as motor neuron disease with the degeneration of upper and lower motor neurons. ALS-D is characterized by both frontotemporal degeneration and motor neuron disease. Hallmarks of both ALS-D and FTLD are the toxic cytoplasmic inclusions of the prion-like C-terminal fragments of TDP-43 C-terminal domain (TDP-43 CTD), which are present in ∼50% of FTLD and ∼97% of ALS patients (Ling et al., 2013). Mutations in the GRN (rs9897526, rs34424835, and rs850713), leading to haploinsufficiency and diminished function of PGRN, are strongly linked to ALS (Sleegers et al., 2008). However, in an ALS Italian cohort, no major PGRN genetic variability was found contribute to disease etiopathogenesis (Del Bo et al., 2011).

    3.2 Animal models with PGRN insufficiency

    As GRN mutations appear to act through a loss-of-function mechanism, PGRN-deficient mice (Grn± and Grn-/-) are the suitable disease models to simulate the disease process of FTLD.

    3.2.1 Behavioral changes

    PGRN-deficient mice have increased depression- and disinhibition-like behaviors, enhanced aggressiveness, elevated anxiety-related behaviors, decreased ejaculation incidence, moderate abnormalities in social interactions, motor coordination, and novel object recognition, from a relatively young age ( Kayasuga et al., 2007; Petkau et al., 2012; Yin, Dumont, et al., 2010); such mice also show impaired spatial learning and memory in their old age (18 months old) (Yin, Dumont, et al., 2010). Furthermore, mice with Grn mutations display self-grooming, an obsessive-compulsive disorder (OCD)-like behavior (Krabbe et al., 2017).

    3.2.2 Pathological changes in the brain

    A development of neuropathology occurs in the brains of PGRN-deficient mice, including the increased loss and vacuolation of neurons (Ahmed et al., 2010), ubiquitination and accumulation of phosphorylated TDP-43 (Yin, Dumont, et al., 2010), enhanced activation of microglia and astrocytes (Ahmed et al., 2010; Tanaka et al., 2013; Yin, Banerjee, et al., 2010; Yin, Dumont, et al., 2010), impaired synaptic connectivity and plasticity (Petkau et al., 2012), lysosomal impairments ( Paushter et al., 2018; Zhou, Sun, Bracko, et al., 2017), inflammation (Krabbe et al., 2017; Menzel et al., 2017), and abnormal intraneuronal lipofuscin accumulation and tissue vacuolation, with focal neuronal loss and severe gliosis apparent in the oldest mice (Ahmed et al., 2010; Petkau et al., 2016; Tanaka et al., 2014; Yin, Dumont, et al., 2010). Besides neuron loss, in the hippocampus, midbrain, brainstem, and thalamus of Grn-/- mice, age-associated, progressive, and increased neuronal lipofuscin aggregations were also observed due to the dysfunction of endosomes and lysosome-related cellular degradation (Ahmed et al., 2010). These lipofuscin accumulations were seen in aged wild-type mice and in Grn± mice but in accelerated amounts in Grn-/- mice particularly at old age. Additionally, the medium spiny neurons in the nucleus accumbens have been reported to display hyperexcitability in Grn-/- mice (Krabbe et al., 2017).

    Grn deletion upregulates the expression of many microglial genes and lysosomal genes. Greater activation of microglia with age in PGRN-deficient mice was also observed (Yin, Banerjee, et al., 2010). Mice with insufficient PGRN have exaggerated inflammation in the brain, which is characterized by increased transcription of the pro-inflammatory cytokines TNFα, IL-1β, and IL-6, and decreased transcription of the anti-inflammatory cytokine IL-10 (Menzel et al., 2017). A recent study indicated that PGRN deficiency promotes microglial transition from a homeostatic to disease-specific state that causes endolysosomal dysfunction and neurodegeneration (Zhang et al., 2020). Grn deletion microglia was shown neurotoxic and promoted TDP-43 proteinopathy (Zhang et al., 2020).

    Transmembrane protein 106B (TMEM106B) is an important lysosomal protein implicated in FTLD. TMEM106B variants have been shown to act as disease modifiers in FTLD. Loss of TMEM106B exacerbates FTLD pathologies and causes motor deficits in PGRN-deficient mice. Compared to Grn-/- mice, Tmem106b-/-Grn-/- mice have exacerbated FTLD-related phenotypes with a much earlier onset, including microgliosis, astrogliosis, ubiquitin, and phosphorylated TDP43 inclusions, as well as worsening of lysosomal and autophagic deficits (Feng et al., 2020; Werner et al., 2020; Zhou et al., 2020). They also show a strongly reduced life span and severe motor deficits (Werner et al., 2020; Zhou et al., 2020).

    4 PGRN and GRNs

    4.1 PGRN acts as a growth factor

    PGRN is a growth factor that promotes neurogenesis and brain development. PGRN stimulates adult neurogenesis in the DG of the hippocampus (Asakura et al., 2011; Chiba et al., 2007; Suzuki et al., 2009), the proliferation of PC12 cells (Daniel et al., 2000), and the male-specific differentiation of the neonatal hypothalamus and contributes to normal brain development (Suzuki et al., 1998, 2000). Moreover, it regulates neurite outgrowth and enhances neuronal survival (Van Damme et al., 2008; Gao et al., 2010; Ryan et al., 2009). In a previous study, neurons with Grn deletion showed significantly decreased neurite outgrowth and branching, and PGRN overexpression and exogenous PGRN was able to rescue this phenotype (Gass, Lee, et al., 2012). The neurite outgrowth promoting activity of PGRN has been shown to depend on GRN(E) (De Muynck et al., 2013; Suzuki et al., 2000). Both PGRN and GRN(E) stimulate the proteolytic activity of CTSD in vitro to promote axonal outgrowth and neuronal survival (Beel et al., 2017; Van Damme et al., 2008).

    The expression of PGRN in the developing cerebral microvasculature (Daniel et al., 2003) and a profound disruption of the blood-brain barrier (BBB) in Grn knockout (KO) mice after brain ischemia-reperfusion (Jackman et al., 2013) raised the possibility that PGRN is involved in maintaining the structure and function of cerebral microvessels. Under the same injury, the lack of PGRN in the brain increases the severity of the injury. Mice with Grn deletion displayed exaggerated axonal injury in the brain following traumatic brain injury (TBI) (Suzuki et al., 1998). Loss of PGRN increases the susceptibility of the brain to BBB breakdown (Jackman et al., 2013).

    PGRN is required for the expression of several receptors in the brain. Estrogen receptor α (ERα) is a classic well-studied ER subtype and is expressed in both neurons and astrocytes (Fang et al., 2018). In Grn-KO mice, PGRN deficiency resulted in a lack of expression of ERα in astrocytes, but did not affect ERα expression in neurons (Doke et al., 2016). The serotonin 1A receptor (5-HT1A) is involved in the inhibition of aggression and anxiety. Hippocampal 5-HT1A mRNA levels were found to be significantly reduced in PGRN-deficient mice after aggressive encounters, suggesting that GRN plays a role in establishing sexual dimorphic behaviors at least partially, by modulating the brain serotonergic system (Kayasuga et al., 2007).

    The mechanism of action of PGRN as a growth factor and neurotropic factor has not been fully elucidated. Although SORT1 is regarded as a neuronal receptor for PGRN, PGRN has been reported to regulate neuronal outgrowth independent of sortilin (Gass, Lee, et al., 2012). Furthermore, the cleavage of full-length PGRN into GRNs is required for increased neuronal outgrowth, suggesting that certain GRNs are involved in the neurotrophic functions of PGRN (Gass, Lee, et al., 2012). Other related receptors and signals involved in this regard need to be investigated further.

    4.2 PGRN/GRNs in neuroinflammation

    Inflammation is a physiological protective response to pathogen exposure, cell injury, and stress. However, dysregulation of the degree and duration of the inflammatory response results in tissue damage. Alterations in PGRN levels are associated with many chronic inflammatory conditions such as rheumatoid arthritis (RA) (Cerezo et al., 2015; Liu, 2011), osteoarthritis (Wei et al., 2017), and inflammatory bowel disease (Thurner et al., 2014), as well as conditions involving acute inflammation such as acute lung injury (Luo et al., 2020; Xie et al., 2018), septic shock (Yu et al., 2016), and acute brain injury (Kanazawa et al., 2015).

    PGRN deficiency may promote or amplify the inflammatory response. Elevated brain inflammation in Grn-KO mice was confirmed by increased transcription of pro-inflammatory cytokines and decreased transcription of anti-inflammatory cytokines (Menzel et al., 2017). When exposed to bacterial lipopolysaccharide, Grn-deficient macrophages produce more proinflammatory cytokines and chemokines including CCL2, CXCL1, IL-6, IL-12p40, and TNF-α and less of the anti-inflammatory cytokine IL-10 (Yin, Banerjee, et al., 2010). Moreover, microglia in PGRN-deficient mice showed greater cytotoxicity than normal microglia under the stimulation of inflammatory factors (Yin, Banerjee, et al., 2010). In models of brain injury, Grn-KO aggravated the neuroinflammatory response, axonal injury, astrogliosis, and neuron loss (Martens et al., 2012; Menzel et al., 2017). Most recently, microglial toxicity has been proven to be a key driving factor that promotes neurodegeneration in PGRN deficiency (Zhang et al., 2020). PGRN deficiency promotes microglial toxicity, resulting in endolysosomal dysfunction and neurodegeneration. Deleting C1qa and C3 mitigated this microglial toxicity and rescued TDP-43 proteinopathy and neurodegeneration (Zhang et al., 2020).

    PGRN has been shown to inhibit inflammatory responses in brain (Jian et al., 2018; Mendsaikhan, Tooyama, & Walker, 2019). Among the multiple pro-inflammatory cytokines, TNF-α is at the peak of the inflammatory cascade, and the increase of TNF-α correlates with the manifestation of an inflammatory reaction (Bradley, 2008). PGRN is found to directly bind to Types 1 and 2 receptors of TNF (TNFR1 and TNFR2) and offset TNF-α-mediated inflammatory signaling (Tang et al., 2011; Tian et al., 2014).

    PGRN and GRNs have been found to possess opposing inflammatory functions. GRNs are pro-inflammatory and may neutralize the anti-inflammatory activities of full-length PGRN (Zhu et al., 2002). Microglia, the resident immune cells in the brain (especially those that have become reactive), produce and secrete high levels of PGRN. In a rat model of cerebral ischemia, the amount of microglial PGRN and the activity of NE increased at an early stage after cerebral ischemia, resulting in the production of GRNs. The inhibition of NE was reported to suppress GRNs production as well as the increase in pro-inflammatory cytokines, suggesting that an increase in PGRN cleavage by NE (to produce GRNs) is involved in the inflammatory response (Horinokita et al., 2019).

    4.3 PGRN/GRNs in lysosomal functions

    The presence of intracellular aggregates in the brains of dementia patients implies a failure in the protein clearance mechanism and notably, a profound disruption of lysosome function (Chung et al., 2019; Götzl et al., 2016; Hwang et al., 2019; Lie & Nixon, 2019).

    PGRN localizes to the lysosomes (Lee et al., 2017) and promotes lysosomal biogenesis and function (Paushter et al., 2018). SORT1 is a high-affinity binding partner of PGRN, and SORT1-mediated sorting is likely to occur both at TGN in the biosynthetic pathway and at the plasma membrane in the process of endocytic trafficking to the lysosomes (Paushter et al., 2018). Like SORT1, the soluble lysosomal protein PSAP is able to transport PGRN from the secretory pathway as well as from the extracellular space. In Sort1-KO mice, PGRN is still successfully transported to neuronal lysosomes (Hu et al., 2010; Zhou et al., 2015), which might be mediated indirectly by the combination of PGRN and PSAP. The transport of PGRN by PSAP depends on CI-M6PR or LRP1, both of which are trafficking receptors of PSAP (Paushter et al., 2018). When transported into lysosomes, PGRN co-localizes with the active CTSL, which is an intracellular PGRN protease within the lysosome (Lee et al., 2017). It has been reported that GRNs, rather than PGRN, are the predominant stable species present in lysosomes and that the GRNs levels are regulated by SORT1 and TMEM106B (Holler et al., 2017).

    PGRN facilitates lysosomal clearance (Kao et al., 2017), possibly by controlling lysosomal acidification (Tanaka et al., 2017) and by acting as a chaperone of degradation enzymes (Beel et al., 2017; Evers et al., 2017; Valdez et al., 2017). In neurons, PGRN co-localizes with cathepsin D (CTSD), an aspartyl protease degrading protein present in the lysosomes (Beel et al., 2017), and increases the activity of CTSD but not that of cathepsin B (CTSB) or CTSL (Valdez et al., 2017). PGRN also stimulates the maturation of the CTSD precursor (proCTSD) at an acidic pH (Butler et al., 2019). PGRN-insufficient neurons exhibit impaired lysosomal proteolysis and decreased CTSD activity (Valdez et al., 2017). Elevated CTSD protein levels were observed in the brains of Grn-KO mice (Tanaka et al., 2014) and in FTLD patients with GRN mutations (FTLD-GRN) (Götzl et al., 2014). However, CTSD activity was significantly decreased in a range of tissue lysates from Grn deletion mice (Zhou, Paushter, et al., 2017). Notably, the addition of recombinant PGRN rescued the decreased activity of CTSD in the brain lysates of geriatric Grn-KO mice (Beel et al., 2017). GRNs are localized in the endolysosomal compartment; they cause impairment of lysosomal functions and induce a compensatory HLH-30/TFEB transcriptional response (Butler et al., 2019c). However, the recombinant peptides GRN(BAC) and GRN (CDE) were reported to induce a significant destabilizing effect on proCTSD. This destabilization has been correlated with enhanced CTSD maturation and activity at an acidic pH (Butler et al., 2019). GRN(E) was found to increase the proteolytic activity of recombinant CTSD (Beel et al., 2017; Valdez et al., 2017).

    Another lysosomal enzyme regulated by PGRN or GRNs is glucocerebrosidase (GBA), a β-glucosidase that cleaves glucocerebroside into glucose and ceramide. As a co-chaperone of heat shock protein 70 (HSP70), PGRN recruits the GBA trafficking receptor, lysosome integral membrane protein 2 (LIMP-2), and GBA (Jian et al., 2016). Thus, PGRN, mainly through the C-terminal GRN(E) region, is required for the lysosomal localization of GBA, and its deficiency leads to cytoplasmic accumulation and activity reduction of GBA (Jian, Tian, et al., 2016; Jian, Zhao, et al., 2016; Zhou et al., 2019). GBA deficiency has been reported in the brains of patients with Gaucher disease (GD) (Stirnemann et al., 2017), lysosomal storage disease (LSD), and sporadic PD (Gegg & Schapira, 2018). Significantly decreased serum PGRN levels have been reported in GD patients (Jian, Chen, et al., 2018; Jian, Zhao, et al., 2016). Impaired GBA was observed in the brains of FTLD-GRN patients; this was associated with lower levels of mature GBA protein and the accumulation of insoluble, incompletely glycosylated GBA (Arrant et al., 2019). GBA activity was significantly reduced in tissue lysates from Grn-KO mice (Zhou et al., 2019). In addition, GRN-mutant neurons showed lipid accumulation and increased insoluble α-synuclein (Valdez et al., 2020). A recent study using FTLD-GRN patient-derived cortical neurons differentiated from induced pluripotent stem cells (iPSCs) as well as post-mortem tissue from patients with FTLD-GRN showed that PGRN haploinsufficiency resulted in the impaired processing of PSAP to saposin C, a critical activator of GBA (Valdez et al., 2020).

    TMEM106B, a highly expressed neuronal protein that is mainly located in the late endosome/lysosome compartments, participates in the regulation of lysosomal morphology and lysosomal trafficking (Schwenk et al., 2014; Stagi et al., 2014). TMEM106B deficiency results in defects in lysosome size, mobility, and lysosomal trafficking (Schwenk et al., 2014; Werner et al., 2020). In mice, Tmem106b deletion leads to accumulation of lysosome vacuoles at the distal end of the axon initial segment in motor neurons and the development of FTLD-related pathology during aging (Feng et al., 2020). However, overexpressing TMEM106B resulted in the accumulation of enlarged lysosomes, delayed the degradation of endocytic cargoes, and exacerbated lysosomal abnormalities caused by the loss of PGRN (Brady et al., 2013; Zhou et al., 2017). PGRN and TMEM106B may be linked synergistically. PGRN deficiency leads to increased TMEM106B protein levels in the mouse cortex with aging and is accompanied by exaggerated lysosomal abnormalities and increased lipofuscin accumulation (Zhou, Sun, Brady, et al., 2017). Thus, PGRN is thought to promote TMEM106B degradation to maintain the proper level of TMEM106B on lysosomal membranes in the aged brain. In mice, TMEM106B deficiency exacerbates PGRN deficit-related FTLD pathologies (Feng et al., 2020; Werner et al., 2020; Zhou et al., 2020). Paradoxically, in another study, loss of TMEM106B ameliorated lysosomal and FLTD phenotypes in Grn-KO mice (Klein et al., 2017).

    4.4 PGRN/GRNs in tau aggregation

    Tau is a microtubule-associated protein. Tauopathies are a heterogeneous group of neurodegenerative dementias (including AD and FTLD) characterized by the accumulation of abnormal tau, leading to the formation of aggresomes such as NFTs (Almansoub et al., 2019; Mroczko et al., 2019; Papanikolopoulou & Skoulakis, 2020). Independent of the aggregation and destabilization of microtubules, phosphorylated tau increased Aβ toxicity and led to the impairment of synaptic function and memory formation in a mouse model of AD (Ittner et al., 2010). How the abnormal tau protein evades the monitoring of intracellular protein control and metabolic system and finally forms these aggresomes has not been fully clarified (Papanikolopoulou & Skoulakis, 2020). To a large extent, tauopathies may result from mutations in the tau gene (MAPT), dysregulation of alternative splicing, and post-translational modifications or truncation.

    In humans, GRN is located on chromosome 17q21, 1.7 Mb centromeric of MAPT (Cruts et al., 2006; Gass et al., 2006). PGRN reduction exacerbates tau pathology. The total level of tau protein was found to be reduced in the brain (Papegaey et al., 2016) but increased in the plasma (Foiani et al., 2018) of FTLD patients. In P301L tau transgenic mice, PGRN protein reduction might contribute to tau hyperphosphorylation and accumulation by upregulating the activity of cyclin-dependent kinases (Hosokawa et al., 2015; Takahashi et al., 2017). In a study of a mouse with the R504X mutation of GRN KI, synapse pathology was specifically associated with tau phosphorylation at Ser203 (corresponding to human Ser214) in the early-stage pathology of FTLD. Specifically, PGRN blocked the activation of Tyro3 and suppressed the activation of its downstream MAPK signaling; PGRN deficiency de-repressed Tyro3 resulting in the activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, the mislocalization of tau to the dendritic spines, and spine loss (Fujita et al., 2018). However, in the middle temporal gyrus samples of AD patients, increased PGRN protein levels correlating with tau phosphorylation at serine 205 were detected, though PGRN-positive NFTs were not observed (Mendsaikhan, Tooyama, Bellier, et al., 2019).

    4.5 PGRN/GRNs in Aβ aggregation

    In addition to NFTs, another major feature of AD pathology is the deposition of extracellular SPs composed of aggregated Aβ; Aβ is formed by the amyloidogenic cleavage of the amyloid precursor protein (APP) (Fagiani et al., 2019; Lloret et al., 2019; Wang et al., 2020). Several pathophysiological roles of APP have been reported in neurodevelopmental diseases (autism, fragile X syndrome, and Lesch-Nyhan disease), neurodegenerative disorders [AD, ALS], and multiple sclerosis) and metabolic disorders (diabetes) (Nguyen, 2019). Aβ is toxic to neurons in various ways including promoting apoptosis, causing synaptic loss, disrupting the cytoskeleton, disrupting cellular calcium balance, and loss of membrane potential. There is abundant evidence showing that the soluble oligomeric Aβ generated extracellularly and intracellularly is the primary noxious form (Fagiani et al., 2019; Wang et al., 2020).

    In the middle temporal gyrus of the human brain, Aβ plaques with PGRN deposits were identified in non-demented cases with low/smaller plaques, suggesting that this is an early event in plaque formation (Mendsaikhan, Tooyama, Bellier, et al., 2019). In the brains of AD mice, PGRN protein levels, but not mRNA levels, were significantly reduced when there were no or fewer SPs (Minami et al., 2014). However, GRNs have been found to colocalize with Aβ plaques in the brains of AD patients (Pickford et al., 2011) and transgenic AD mice (Pereson et al., 2009). GRN(B) binds to monomeric and oligomeric Aβ42 to promote rapid fibril formation. As Aβ oligomers are well-established neurotoxins, the rapid promotion of fibrils by GRN(B) mitigates Aβ42-induced cellular apoptosis (Bhopatkar et al., 2019).

    Selective reduction of microglial PGRN in AD mice impaired phagocytosis, increased plaque load threefold, and exacerbated cognitive deficits (Minami et al., 2014). In contrast, PGRN deficiency significantly reduced Aβ deposition in APP mice and APP/PS1 mice (Hosokawa et al., 2018; Takahashi et al., 2017). It has been reported that global PGRN reduction induces the expression of microglial TYROBP network genes (TNG) and increases AD risk by exacerbating neuronal injury and tau pathology rather than by accelerating Aβ pathology (Takahashi et al., 2017). All these reports suggest that neuronal PGRN and microglia PGRN might have different roles in Aβ production and toxicity.

    4.6 PGRN/GRNs in TDP-43 aggregation

    TDP-43 is a DNA/RNA binding protein that controls the expression of thousands of genes and is a major component of neuronal and glial aggregates in the brains of tau-negative FTLD, AD, DLB, HS, ALS-D, and TBI patients with dementia (Amador-Ortiz et al., 2007; Arai et al., 2006; Flanagan et al., 2018; Heyburn et al., 2019; Ling et al., 2013; Neumann et al., 2006). It is also associated with Huntington's disease (HD) and stroke (Hatsuta et al., 2019; Heyburn et al., 2019).

    Pathological modifications of TDP-43 involve both the loss of its normal function in the nucleus and the toxicity of its cytoplasmic components. Pathological TDP-43 is hyperphosphorylated, ubiquitinated, cleaved into fragments comprising of TDP-43 CTD, and is mislocalized to cytoplasmic protein inclusions and the mitochondria (Bayram et al., 2019; Cascella et al., 2019; Huang et al., 2018). The degree of TDP-43 pathology correlates with the degree of neuronal loss (Mackenzie et al., 2013). Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity (Zhang et al., 2009). Two C-terminal fragments of TDP-43, a 25-kDa fragment (TDP-25) and a 35-kDa (TDP-35) fragment, are generated by caspases and exist in the neurons of patients with ALS and FTLD (Neumann et al., 2006) or other neurodegenerative diseases (Bayram et al., 2019; Gao et al., 2018). To date, the toxicities of TDP-25 and TDP-35 remain unclear. TDP-35 retains both the RRM1 and RRM2 sequences responsible for the interaction with RNA, thus maintaining its ability to regulate RNA maturation (Kitamura et al., 2016). However, this truncated form has a defective nuclear localization signal and accumulates in the cytoplasm, where it is highly prone to aggregate formation (Bozzo et al., 2016). TDP-25, which is defective in RNA-binding motifs, is not able to bind mRNAs (Kitamura et al., 2016). Both full-length TDP-43 and fragmented TDP-43 accumulate in the cytoplasm and form insoluble inclusions by coacervation with RNA in the cytoplasm and are further toxic to mammalian neurons (Weskamp et al., 2020). TDP-43 also induces mitochondrial damage by decreasing the mitochondrial membrane potential, inhibiting mitochondrial complex I activity, reducing mitochondrial ATP synthesis, and elevating the production of reactive oxygen species (ROS) (Wang et al., 2019). Recently, it was reported that TDP-43 pathology contributes to dementia status and progression in a variety of ways in different phosphorylation states involving both neurons and glia, independent of age and classic AD-related pathologies (Buciuc et al., 2020).

    TDP-43 pathology is associated with GRN mutations (Baker et al., 2006; Cruts et al., 2006; Ghidoni et al., 2008; Mackenzie, 2007; Sleegers et al., 2009). PGRN mediates the proteolytic cleavage of TDP-43 to generate TDP-25 and TDP-35 and reduces insoluble TDP-43 levels in mutant TDP-43 mice (Beel et al., 2018). Suppression of PGRN expression leads to caspase-dependent accumulation of TDP-43 fragments (Zhang et al., 2007). PGRN-deficient mice are reported to display greater activation of microglia and astrocytes and more neuronal cytosolic phosphorylated TDP-43 accumulation than wild-type mice (Yin, Banerjee, et al., 2010). Furthermore, mutations or genetic variants of PGRN underlie the abnormal deposition of TDP-43 and the co-occurrence of abnormal deposition of TDP-43 and tau in neurodegeneration pertaining to dementia (Arai et al., 2009; Baker et al., 2006; Cruts et al., 2006). It is still unknown as to how PGRN deficiency causes TDP-43 aggregation. The derailment of lysosomal degradation pathways with reduced clearance of TDP-43 may be involved (Götzl et al., 2014). Cells lacking PGRN show reduced autophagic flux (Chang et al., 2017) and lysosomal dysfunction (Kao et al., 2017; Paushter et al., 2018). Consequently, Grn deletion neurons more readily accumulate pathological forms of TDP-43 (Caccamo et al., 2009; Chang et al., 2017; Paushter et al., 2018). Additionally, PGRN deficiency also affects autophagy and lysosome-independent pathways to induce TDP-43 pathology and neurodegeneration such as impairing Ran-mediated nuclear import of cytoplasmic TDP-43 (Ward et al., 2014). It has been confirmed that PGRN overexpression reduces insoluble TDP-43 levels in TDP-43 (A315T) mice (Beel et al., 2018).

    Different from PGRN, specific GRNs may exacerbate TDP-43 cytotoxicity and increase TDP-43 levels (Salazar et al., 2015). Recently, it was shown that GRN(B) and GRN(C) interact with TDP-43 CTD and differentially modulated TDP-43 CTD aggregation in vitro; further, it has been reported that GRN(B) promotes the formation of insoluble aggregates of the TDP-43 CTD, while GRN(C) mediates liquid-liquid phase separation (Bhopatkar et al., 2020).

    4.7 PGRN/GRNs in neuronal loss and synaptic deficit

    PGRN has multiple neurotrophic effects including stimulating neurite growth, enhancing synaptic connectivity, and promoting neuron survival (Van Damme et al., 2008). PGRN deficiency reduces neural connectivity and leads to increased neuronal loss following injury (Gass, Lee, et al., 2012; Martens et al., 2012). In rat primary hippocampal cultures, downregulation of PGRN decreased neuronal arborization and length, as well as synapse densities. However, the number of synaptic vesicles per synapse and the frequency of mEPSCs increased, suggesting an increase in release at the remaining synapses. The number of vesicles per synapse was also found to be increased in post-mortem brain sections from FTLD-GRN patients with PGRN haploinsufficiency (Tapia et al., 2011). In 10- to 12-month-old Grn-KO male mice, altered synaptic connectivity and impaired synaptic plasticity were observed. In the hippocampal CA1 region of Grn-KO male mice, apical dendrites in pyramidal cells displayed an altered morphology, and decreased spine density was observed (Petkau et al., 2012). GluN2B-containing NMDAR (NR2B) and tau phosphorylation induced by NR2B are important for the regulation of the structural plasticity of neurons (Laurier-Laurin et al., 2014; Tan et al., 2017). PGRN deficiency increased tau AT8 and AT180 pathologies in P301L tau mice (Takahashi et al., 2017). In contrast, PGRN decline diminished the dendritic arborization of primary cortical neurons by reducing the levels and density of NR2B as well as tau phosphorylation (at serine 396/404 and serine 262); this effect was prevented by acute NR2B stimulation (Longhena et al., 2017). In APP/PS1 mice, PGRN-deficient mice exhibited reduced diffuse Aβ plaque growth and less severe axonal dystrophy but enhanced C1q complement deposition at the synapses (Takahashi et al., 2017). PGRN deficiency led to an age-dependent, progressive increase in complement production and enhanced synaptic pruning in microglia (Lui et al., 2016). During aging, Grn-KO mice showed profound microglial infiltration and preferential elimination of inhibitory synapses in the ventral thalamus, which lead to hyperexcitability in the thalamocortical circuits and obsessive-compulsive disorder (OCD)-like grooming behaviors. Remarkably, deletion of the C1qa gene significantly reduced synaptic pruning by Grn-KO microglia and mitigated neurodegeneration, behavioral phenotypes, and premature mortality in Grn-KO mice (Lui et al., 2016). Thus, the upregulation of microglial synaptic pruning might be a key factor that promotes synaptic deficit in PGRN deficiency.

    In different pathological models, PGRN overexpression has different effects on neuronal morphology and connections. In TDP-43 mutant mice, PGRN reduced insoluble TDP-43 levels, slowed axonal degeneration, and prolonged survival (Beel et al., 2018). In AD mice, the overexpression of PGRN in hippocampus significantly reduced Aβ plaque burden and revised synaptic atrophy (Van Kampen & Kay, 2017). Fragile X syndrome is an inheritable form of intellectual disability and autistic behavior caused by the loss of the Fragile X mental retardation protein (FMRP, encoded by FMR1) (Salcedo-Arellano et al., 2020), which is an RNA-binding protein that regulates the local translation of numerous mRNAs at the synapses (Prieto et al., 2020). The absence of FMRP causes the overexpression of PGRN. PGRN mRNA and protein were upregulated in the medial prefrontal cortex of Fmr1-KO mice, causing insufficient dendritic spine pruning and late-phase long-term potentiation (LTP). In Fmr1-KO mice, the partial downregulation of PGRN restored spine morphology and reversed behavioral deficits including impaired fear memory, hyperactivity, and motor inflexibility (Zhang et al., 2017). It is noteworthy that FMRP and TDP-43 are associated with ribonuclear protein particles and share mRNA targets in neurons (Ferro et al., 2018; Majumder et al., 2016).

    4.8 PGRN/GRNs in the extracellular fluid of patients with dementia

    The levels of PGRN and GRNs in the extracellular fluid (CSF or serum/plasma) have diagnostic and predictive value for AD and FTLD. However, they are not helpful in discriminating diseases because a variety of diseases with inflammation also present changes in the levels of PGRN/GRNs.

    Mutations in GRN have been considered to be among the most common causes of familial FTLD (Mackenzie et al., 2011; Schymick et al., 2007). Both PGRN and multiple GRNs are haploinsufficient in the brains of FTLD-GRN carriers. It was reported that GRNs were significantly reduced (by ~60%) compared to controls, while PGRN was reduced by ∼40% in soluble lysates of the frontal cortex (Brodmann Area 9) brain tissue from FTLD-GRN cases (Holler et al., 2017). In primary human fibroblasts from FTLD-GRN patients, PGRN and GRNs were significantly decreased by ~50% (Holler et al., 2017). The PGRN levels of FTLD-GRN patients decreased by ~50% in the plasma and CSF (Finch et al., 2009; Ghidoni et al., 2012; Goossens et al., 2018; Meeter et al., 2016). The associations between the decreased levels of CSF PGRN and impairments in categoric and letter fluency, naming, and overall cognition, as well as olfactory dysfunction were observed in FTLD cases (Körtvélyessy et al., 2015). Recently, it was confirmed that plasma PGRN levels predict GRN mutations even in pre-symptomatic carriers more than four decades before disease onset. Although plasma PGRN does not correlate with age at onset and FTLD phenotypes, stable plasma PGRN levels constitute a reliable and cost-effective marker that is suitable as a screening tool in patients with FTLD and more broadly in patients without a family history or with atypical presentations of the disease (Sellami et al., 2020).

    A common rs5848 allele in the 3′UTR of GRN is associated with decreased serum and brain PGRN levels and an increased risk of developing AD (Hsiung et al., 2011; Perry et al., 2013; Sheng et al., 2014). PGRN localizes around SPs in the post-mortem brain tissue of AD patients (Gliebus et al., 2009; Mendsaikhan, Tooyama, Bellier, et al., 2019; Pereson et al., 2009). CSF PGRN increases in the course of AD and is associated with soluble triggering receptors expressed on myeloid cells 2 (sTREM2), neurodegeneration, and cognitive decline (Suárez-Calvet et al., 2018). PGRN mRNA is elevated in the peripheral blood of patients with AD and mild cognitive impairment (MCI) (Cooper et al., 2018). In a study involving 107 AD patients and 107 healthy controls, there were no differences in plasma PGRN levels between patients with AD and healthy controls. However, a positive correlation between plasma PGRN levels and age was observed in female AD patients who had a higher PGRN level than male AD patients (Piscopo et al., 2013). Collectively, these data suggest that PGRN has limited diagnostic utility for AD (Morenas-Rodríguez et al., 2016; Piscopo et al., 2013).

    5 INCREASING PGRN LEVEL TO PREVENT NEURODEGENERATION IN DEMENTIA

    As an effective neuroinflammatory regulator and autocrine neurotrophic factor, PGRN is important for long-term neuronal survival and for improving brain availability. Although the mechanisms linking PGRN deficiency with neurodegeneration in dementia are not yet fully understood, it is clear that PGRN deficiency contributes to neurodegeneration, occurs in the very early stage of dementia, and lasts stably for a long time. These characteristics suggest that PGRN may be an important therapeutic target, and restoring PGRN levels may be an effective way to prevent and treat dementia. (Cui et al., 2019; Elia et al., 2020; Galimberti et al., 2018; Gass et al., 2012; Malik et al., 2019).

    The overexpression of PGRN by gene delivery is an alternative therapeutic strategy for neurodegenerative disorders due to GRN mutations. Such a strategy may help ameliorate neuronal and microglial pathology and improve lysosomal dysfunction. Adeno-associated virus (AAV) vector delivery of GRN to the medial prefrontal cortex rescued the social behavioral deficits in Grn +/− mice (Arrant et al., 2017). In Grn-KO mice with features of NCL and FTLD, PGRN overexpression in neurons reduced lipofuscinosis and microgliosis and improved lysosomal function even at low doses (Arrant et al., 2018). In AD mice, lentivirus-mediated PGRN overexpression in microglia prevented spatial memory deficits and hippocampal neuronal loss by inhibiting Aβ deposition and toxicity (Minami et al., 2014). In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice, PGRN overexpression in nigrostriatal neurons preserved both dopamine content and locomotor function and was accompanied by reduced inflammation and apoptosis (Van Kampen et al., 2014). It was further found that PGRN conferred therapeutic benefits by interacting with TDP-43 to regulate polyglutamine toxicity in a Caenorhabditis elegans model of Huntington′s disease (HD) (Tauffenberger et al., 2013). Passage Bio Inc. and Prevail Therapeutics Inc. are committed to the application of AAV gene therapy drugs, including nucleic acid vectors encoding PGRN and its variants, derivatives, or functional fragments in the treatment of PGRN deficit-related brain diseases.

    Developing small-molecule drugs to increase PGRN levels is a promising way to treat dementia. Suberoylanilide hydroxamic acid (SAHA), an HDAC inhibitor approved for use in cancer treatment, upregulates PGRN transcription. Combined with the ERK1/2 blocker selumetinib, SAHA was able to significantly inhibit cytosolic TDP-43 accumulation (Cenik et al., 2011). FRM-0334, also an HDAC inhibitor, is in phase II clinical trials for the amelioration of GRN mutation-induced PGRN insufficiency. This drug crosses the blood-brain barrier more easily than SAHA (Tsai & Boxe r, 2016). Knocking down Ripk1 increases both intracellular and extracellular PGRN protein levels (Mason et al., 2017); thus, targeting RIPK1 may be a therapeutic strategy for PGRN deficit-related dementia (Degterev et al., 2019; Mifflin et al., 2020; Yuan et al., 2019). Denali Therapeutics Inc. is vigorously developing small-molecule inhibitors of RIPK1. Alkalizing drugs (chloroquine, bepridil, and amiodarone), vacuolar ATPase inhibitors (bafilomycin A1, concanamycin A, archazolid B, and apicularen A), and the channel blocker nimodipine are also potential pharmacological upregulators of PGRN production (Capell et al., 2011). Trehalose, a natural disaccharide, dose-dependently increased PGRN mRNA as well as intracellular and secreted PGRN in both mouse and human cell lines independent of TFEB. Moreover, trehalose rescued PGRN deficiency in human fibroblasts and neurons derived from iPSCs generated from GRN mutation carriers. Finally, the oral administration of trehalose to PGRN haploinsufficient mice significantly increased PGRN expression in the brain (Holler et al., 2016).

    SORT1 is a potential target to correct PGRN reduction (Miyakawa et al., 2020). The therapeutic potential of SORT1 was developed by generating and characterizing monoclonal antibodies that downregulate SORT1. Alector Inc. received the Orphan Drug Designation from the US FDA for AL001, a monoclonal antibody targeting SORT1 for the treatment of FTLD.

    The adverse and the off-target effects of PGRN increasing need to be further evaluated. More recently, researchers have used AAV9 to deliver GRN to the lateral ventricles of brains in Grn-KO mice. Surprisingly and unexpectedly, despite a global increase in PGRN, this overexpression resulted in dramatic and selective hippocampal toxicity and degeneration affecting neurons and glia, which was preceded by T cell infiltration and perivascular cuffing (Amado et al., 2019). Furthermore, GRN overexpression in wild-type animals also induced T-cell infiltration (Amado et al., 2019). The effects of using recombinant human PGRN protein (rhPGRN), PGRN coding gene, and small molecule drugs to upregulate PGRN levels seem be unpredictable. Therefore, it is needed to understand the details of PGRN metabolism in different tissues, such as the receptors associated with its neuroprotections, its intracellular, and extracellular degradations, and the actions of different GRNs. Overexpressed PGRN was reported in the cancer tissues, and the upregulation of PGRN promotes the invasion and metastasis of cancer cells (Liu et al., 2020). At the same time, the upregulation of PGRN enhances the secretion of several inflammatory cytokines. Therefore, the risks of cancer and inflammation caused by long-term upregulation of PGRN should be considered.

    6 CONCLUSIONS

    Although the mechanism of neurodegeneration in dementia remains unclear, a long-term or serious lack of protective factors is important in the pathogenesis of dementia. The important role of PGRN in dementia is highlighted by its multiple neuroprotective functions such as promoting neuron survival and neurite growth, as well as anti-inflammatory and lysosomal chaperone properties. Mutations in GRN have been found in several neurodegenerative dementias including FLTD, AD, NCL, and HS-Aging. Herein, GRN mutations, PGRN deficiency, and GRNs in the neuropathological changes related to dementia including aggregation of tau, Aβ, and TDP-43, neuroinflammation, lysosomal dysfunction, neuronal death, and synaptic deficit were summarized. Details of several therapeutic strategies targeting PGRN that have been developed using various models have also been presented. These strategies highlight PGRN as a potential anti-neurodegeneration target in dementia.

    ACKNOWLEDGMENTS

    This work was supported by a grant from the National Natural Science Foundation of China (82071478).

      CONFLICT OF INTEREST

      The authors declare that they have no conflicts of interest (financial or otherwise) related to the information presented in this manuscript.

      DATA AVAILABILITY STATEMENT

      Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

        The full text of this article hosted at iucr.org is unavailable due to technical difficulties.