PGRN consists of seven full granulin (GRN) domains and one half-domain, connected by linker regions. These domains form a complex structure with β-hairpins stabilized by disulfide bonds . Key features include:
PGRN’s roles span neuroprotection, inflammation, and metabolic regulation:
Lysosomal Function: Regulates lysosomal lipid metabolism, with deficiency linked to reduced triglycerides (TAGs) and altered sphingolipid profiles .
Neurodegeneration: PGRN haploinsufficiency causes frontotemporal dementia (FTD-GRN), associated with TDP-43 pathology .
Anti-Inflammatory Effects: Binds TNFR and DR3, suppressing inflammation in macrophages and Tregs .
Cancer Progression: Elevated PGRN levels correlate with tumor severity in ovarian and brain cancers, driven by mitogenic and angiogenic properties .
Genetic Defects: GRN mutations (e.g., nonsense, splice site) reduce PGRN levels, leading to neurodegeneration. Plasma PGRN thresholds:
Sex and Age Effects: Higher plasma PGRN in women and weak correlation with age .
Biomarker for FTD: Low plasma/CSF PGRN distinguishes GRN mutation carriers from non-carriers .
Alzheimer’s Disease: Elevated PGRN in medial temporal gyrus (MTG) correlates with amyloid-β and tau pathology .
Recombinant Human Progranulin, produced in HEK cells, is a single, glycosylated polypeptide chain. It consists of 1-593 amino acids and has a molecular weight of 74 kDa. The purification of Progranulin is achieved through standard chromatographic techniques.
Sterile Filtered White lyophilized (freeze-dried) powder.
The protein is supplied in a solution containing 1xPBS (phosphate-buffered saline).
To reconstitute the lyophilized Progranulin, it is recommended to dissolve it in sterile 18MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can then be further diluted as needed using other aqueous solutions.
Lyophilized Progranulin remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated at a temperature below -18°C. After reconstitution, PGRN should be stored at 4°C for 2-7 days. For extended storage, it should be kept at -18°C. Adding a carrier protein, such as 0.1% HSA (human serum albumin) or BSA (bovine serum albumin), is advisable for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
The purity of the protein is determined to be greater than 98.0% using SDS-PAGE analysis.
The biological activity of Progranulin is demonstrated by its ability to activate phospho-ERK1/2 in neuronal mouse P19 cells. Additionally, it has been shown to play a regulatory role in food intake and body weight.
Human progranulin is a glycoprotein of approximately 75-80 kDa containing seven and a half granulin modules named p (a half domain), G, F, B, A, C, D, and E . The granulin motif has a unique conformation characterized by a parallel stack of beta-hairpins in the form of a left-handed helix, stabilized by six disulfide bridges . This structural organization is critical for its biological functions and interactions with cellular components. Solution nuclear magnetic resonance analysis initially performed on carp granulin-1 and later on individual human granulin modules revealed this distinct structural arrangement .
Progranulin can undergo proteolytic cleavage that releases individual granulin modules of approximately 6 kDa, sometimes referred to as granulins or epithelins . This processing significantly alters biological activity, as intact progranulin generally exhibits anti-inflammatory properties, while the cleaved granulin peptides tend to promote inflammation . The balance between full-length progranulin and its cleaved products is crucial for maintaining appropriate inflammatory responses. Research methods to study this balance include western blotting with antibodies specific to different regions of the protein and protease inhibition assays to prevent processing in experimental systems.
Progranulin is expressed in multiple cell types including neurons, microglia, and various epithelial cells . In the context of the central nervous system, both neurons and microglia produce progranulin, though microglia appear to be the predominant source . Under inflammatory conditions or following injury, progranulin expression is often upregulated in activated immune cells, particularly microglia in the brain . Cell-type specific expression can be studied using immunohistochemistry, in situ hybridization, and single-cell RNA sequencing techniques.
The most common disease-causing mutations in the GRN gene are nonsense mutations, frameshift mutations, and splice site mutations that lead to haploinsufficiency through degradation of mutant mRNA by nonsense-mediated decay . These mutations result in approximately 50% reduction of functional progranulin, which is sufficient to cause frontotemporal lobar degeneration (FTLD) . Complete loss of progranulin due to homozygous mutations leads to neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disorder . Genetic screening for GRN mutations should include sequencing of all exons and flanking regions, as well as methods to detect large deletions or duplications such as multiplex ligation-dependent probe amplification (MLPA).
Beyond frontotemporal dementia, certain GRN variants that decrease progranulin expression have been identified as risk factors for developing Alzheimer's disease and Parkinson's disease . The exact mechanisms by which reduced progranulin contributes to the pathogenesis of these diseases are not fully understood but may involve impaired lysosomal function, altered neuroinflammatory responses, or disrupted protein clearance pathways . Genome-wide association studies (GWAS) and targeted genetic analyses are required to establish these connections, along with functional assays that measure the impact of specific variants on progranulin expression and activity.
Genetic testing for mutations in the GRN gene is the primary diagnostic approach to identify PGRN-associated FTLD . Additionally, plasma progranulin levels can serve as a biomarker, as patients with GRN mutations typically show significantly reduced circulating progranulin . The GRN-related form of FTLD is characterized by neuronal inclusions containing ubiquitinated and fragmented TDP-43 (encoded by TARDBP), which can be identified in post-mortem brain tissue . Advanced research methods include next-generation sequencing panels targeting multiple FTLD-associated genes, and detailed neuropathological characterization of TDP-43 inclusions using specialized immunostaining protocols.
Progranulin functions as a key regulator of lysosomal function, and its deficiency leads to significant lysosomal abnormalities . These include impaired protein trafficking to lysosomes, dysregulated activity of lysosomal enzymes, and defective lysosomal acidification . In particular, progranulin deficiency affects the degradation of mature cathepsin D (CTSD) by cathepsin B (CTSB) . Complete loss of progranulin in humans results in neuronal ceroid lipofuscinosis, a lysosomal storage disease, further supporting the critical role of progranulin in lysosomal homeostasis . Research methods to evaluate lysosomal function include lysosomal enzyme activity assays, lysosomal pH measurements using specific dyes, and electron microscopy to assess lysosomal morphology.
Progranulin exhibits significant neuro-immunomodulatory properties, generally functioning as an anti-inflammatory factor in contrast to the pro-inflammatory effects of granulin peptides . In progranulin-deficient mouse models, there is evidence of exaggerated neuroinflammatory responses characterized by increased microglial activation and elevated pro-inflammatory cytokine production . This dysregulated neuroinflammation likely contributes to neurodegeneration in FTLD and other conditions associated with progranulin deficiency . Recent research has revealed sex-dependent effects of progranulin deficiency on both peripheral and central immune system regulation, suggesting complex interactions between progranulin, sex hormones, and inflammatory processes . Advanced research approaches include single-cell RNA sequencing of microglia from progranulin-deficient models, multiplex cytokine profiling, and in vivo imaging of neuroinflammation.
For measuring progranulin in biological samples, enzyme-linked immunosorbent assay (ELISA) is the gold standard for quantification in plasma, serum, and cerebrospinal fluid (CSF) . Immunohistochemistry is preferred for tissue localization studies, while western blotting can distinguish between full-length progranulin and its processed forms . When analyzing clinical samples, it's critical to standardize collection and processing procedures, as progranulin levels can be affected by sample handling and storage conditions. For research requiring higher sensitivity, newer techniques such as Single Molecule Array (Simoa) technology can detect progranulin at femtomolar concentrations, which is particularly useful for CSF samples where progranulin concentrations are relatively low.
Several in vitro and in vivo models have been developed to investigate progranulin function. Cellular models include primary neuronal cultures, microglial cultures, and various cell lines with progranulin knockdown or overexpression . For animal models, progranulin knockout (Grn−/−) mice exhibit abnormal behavior and neuropathology reminiscent of human FTLD and are widely used . Additionally, zebrafish models have been valuable for studying progranulin's role in motor neuron diseases, while C. elegans models have provided insights into progranulin's function in Huntington's disease . When selecting a model, researchers should consider species-specific differences in progranulin biology and the particular aspect of progranulin function being investigated. Advanced approaches include conditional knockout models, which allow tissue-specific and temporally controlled deletion of progranulin.
Distinguishing between full-length progranulin and individual granulin peptides requires specialized techniques. These include using domain-specific antibodies that recognize either intact progranulin or specific granulin domains . Recombinant expression systems can be employed to produce either full-length progranulin or individual granulin peptides for functional studies . To investigate processing dynamics, researchers can use protease inhibitors to prevent cleavage of progranulin or engineer cleavage-resistant progranulin variants . Mass spectrometry-based approaches can identify and quantify specific granulin peptides in biological samples. When interpreting results, it's important to consider that commercially available progranulin antibodies may have different specificities for the full-length protein versus its processed forms.
Various therapeutic strategies are being investigated to increase progranulin levels, particularly for treating FTLD and other neurodegenerative diseases. These include gene therapy approaches using viral vectors to deliver the GRN gene, which has shown promising results in animal models of Alzheimer's and Parkinson's disease-like pathologies . Small molecule compounds that can increase progranulin expression, such as trehalose, are also being studied . Additionally, researchers are exploring the potential of recombinant progranulin protein administration and developing antibodies that stabilize progranulin by preventing its degradation or processing . Histone deacetylase inhibitors and other epigenetic modulators have also been investigated for their ability to enhance progranulin expression from the remaining functional GRN allele in patients with FTLD.
Progranulin treatment has shown diverse beneficial effects across multiple neurodegenerative disease models. In zebrafish models of motor neuron disease, progranulin overexpression reverses impaired development of primary motor neurons . In mice, lentiviral delivery of Grn to the brain prevents the development of drug-induced Parkinson's-like phenotypes and genetically-induced Alzheimer's disease-like pathologies . Progranulin has also demonstrated neuroprotective effects in a C. elegans model of Huntington's disease . The mechanisms underlying these beneficial effects likely involve progranulin's neurotrophic and anti-inflammatory properties, as well as its regulation of lysosomal function . When evaluating progranulin as a therapeutic agent, researchers should assess multiple outcome measures including behavioral improvements, biochemical changes, and histopathological alterations.
Development of progranulin-based therapies faces several challenges. A major obstacle is achieving sufficient central nervous system (CNS) penetration, as progranulin is a relatively large protein that does not readily cross the blood-brain barrier . Additional challenges include determining the optimal dosing regimen, understanding potential off-target effects given progranulin's pleiotropic functions, and addressing the potential oncogenic properties of progranulin in certain contexts . The complex processing of progranulin into granulin peptides with opposing inflammatory properties further complicates therapeutic development . Clinical trials must also address patient stratification, as the response to progranulin-enhancing therapies may vary depending on the underlying genetic and pathological mechanisms of disease.
Progranulin regulates both the trafficking of proteins to lysosomes and the activity of lysosomal enzymes . It facilitates lysosomal acidification, which is crucial for optimal functioning of lysosomal enzymes . Specifically, progranulin has been shown to interact with and stabilize cathepsin D (CTSD), maintaining its aspartic-type peptidase activity . In progranulin-deficient states, there is evidence of altered lysosomal enzyme trafficking and activity, contributing to lysosomal dysfunction . Research approaches to study these interactions include lysosomal fractionation techniques, live-cell imaging of fluorescently tagged lysosomal proteins, and enzymatic activity assays under varying pH conditions. Co-immunoprecipitation and proximity ligation assays can help identify specific interactions between progranulin and lysosomal proteins.
Progranulin interacts with several proteins implicated in neurodegeneration. The most well-established relationship is with TDP-43, where progranulin deficiency leads to TDP-43 mislocalization and aggregation . Progranulin may also interact with or influence the processing/aggregation of other disease-related proteins such as amyloid-β and α-synuclein, as suggested by progranulin's protective effects in Alzheimer's and Parkinson's disease models . Additionally, progranulin has been shown to interact with sortilin, a neuronal receptor that regulates progranulin endocytosis and may affect its availability in the CNS . Research techniques to investigate these relationships include co-localization studies using super-resolution microscopy, protein-protein interaction assays, and genetic interaction studies in model organisms.
Progranulin exhibits significant neurotrophic properties, promoting neuronal survival, axonal outgrowth, and maintenance of neuronal integrity . In primary neuronal cultures, progranulin deficiency leads to decreased neural connectivity, while in motor neuron-like cell lines, progranulin overexpression enhances survival under stress conditions . Progranulin exerts anti-apoptotic effects in neurons, with its absence leading to increased caspase-3 activity in response to apoptotic stimuli . In mouse models with targeted overexpression of progranulin in neurons, there is markedly faster axonal regrowth, reformation of neuromuscular junctions, and improved motor function after nerve injury . Research approaches to study these effects include neurite outgrowth assays, growth cone collapse assays, live-cell imaging of axonal transport, and targeted deletion or overexpression of progranulin in specific neuronal populations.
Recent research has revealed significant sex-dependent effects of progranulin deficiency on both peripheral and central immune system regulation . These differences may contribute to the variability in disease presentation and progression observed in progranulin-associated neurodegenerative disorders. Sex hormones likely interact with progranulin signaling pathways, potentially affecting microglial activation states, cytokine production profiles, and neuroinflammatory responses . When designing studies investigating progranulin in neurodegeneration, researchers should include balanced sex representation and analyze data for potential sex differences. Advanced approaches include examining the effects of gonadectomy or hormone replacement on progranulin-dependent phenotypes and investigating potential interactions between progranulin and sex hormone receptors.
Despite carrying the same GRN mutation, affected individuals often show substantial variability in disease onset, clinical presentation, and progression rate . Several factors may contribute to this variability, including genetic modifiers, environmental exposures, and lifestyle factors. Potential genetic modifiers include variants in genes encoding proteins that interact with progranulin or function in the same pathways . Epigenetic modifications affecting the expression of the remaining functional GRN allele may also play a role. Research approaches to investigate this variability include longitudinal studies of GRN mutation carriers, whole genome sequencing to identify potential genetic modifiers, and development of induced pluripotent stem cell (iPSC) lines from patients with different clinical presentations despite identical GRN mutations.
Aging is a major risk factor for neurodegenerative diseases, and age-related changes in progranulin expression and processing may contribute to disease susceptibility. Some studies suggest that progranulin levels in the brain decrease with age, potentially due to altered transcriptional regulation or increased proteolytic processing . Age-related changes in microglial function, a major source of progranulin in the CNS, may also affect progranulin availability and processing . Research methods to investigate age-related changes include analysis of brain tissue samples across the lifespan, longitudinal measurement of plasma or CSF progranulin levels, and examination of age-dependent changes in progranulin processing enzymes. Advanced approaches include single-cell RNA sequencing to examine cell-type specific changes in progranulin expression with aging and investigation of age-related epigenetic modifications of the GRN gene.
Progranulin plays important roles in synaptic function and plasticity, with its deficiency leading to significant alterations in synaptic morphology and transmission. In primary hippocampal cultures from progranulin-deficient mice, there is decreased gross neural connectivity but enhanced transmission at individual synapses, suggesting compensatory mechanisms . Similar increases in the number of synaptic vesicles per synapse have been observed in tissues from progranulin-associated FTLD patients . Progranulin deficiency may affect both excitatory and inhibitory synapses, potentially contributing to network dysfunction and excitotoxicity in neurodegenerative conditions. Research techniques to investigate these effects include electrophysiological recordings, high-resolution imaging of synaptic structures, and optogenetic manipulation of neuronal activity in progranulin-deficient models.
Emerging evidence suggests potential interactions between progranulin and the gut-brain axis in neurodegeneration. Given progranulin's role in immune modulation and inflammation, it may influence gut microbiota composition and intestinal barrier function, which are increasingly recognized as contributors to neuroinflammation and neurodegeneration . Conversely, gut microbiota may affect progranulin expression or processing through production of metabolites or modulation of systemic inflammation. Research approaches to investigate these interactions include analysis of gut microbiome composition in progranulin-deficient models, assessment of intestinal permeability and inflammation, and examination of progranulin expression in enteric neurons and glial cells. Advanced methods include gnotobiotic models with controlled microbiota composition and targeted manipulation of specific bacterial populations.
Progranulin is composed of 7.5 repeats of a highly conserved 12-cysteine granulin/epithelin motif . The precursor protein, progranulin, is cleaved to produce mature granulin peptides, which can be further processed into smaller active peptides named granulin A, B, C, etc. These peptides, along with the intact granulin protein, play crucial roles in regulating cell growth .
Progranulin is involved in a wide range of biological functions:
Progranulin’s role in various diseases has been extensively studied. It is implicated in neurodegenerative diseases such as frontotemporal dementia (FTD) and Alzheimer’s disease. Mutations in the progranulin gene (GRN) can lead to reduced levels of progranulin, contributing to the pathogenesis of these conditions . Additionally, progranulin is involved in cancer biology, where it can act as a growth factor promoting tumorigenesis .