Fact-checked by Grok 2 weeks ago

Glucocerebrosidase

Glucocerebrosidase, also known as β-glucosylceramidase or GCase, is a lysosomal enzyme encoded by the GBA1 gene on chromosome 1q22 that catalyzes the of (GlcCer), a intermediate, into glucose and within the acidic environment of lysosomes. This enzymatic activity is essential for metabolism, preventing the accumulation of GlcCer in cells, particularly macrophages, which can otherwise lead to lysosomal storage disorders. Mutations in GBA1 result in deficient or dysfunctional GCase, causing , an autosomal recessive disorder characterized by GlcCer buildup in organs like the , liver, and . The precursor protein consists of 536 and functions as a membrane-associated protein in lysosomes, with its involving catalytic residues such as Glu340 and Glu235 that facilitate the β-glucosidic bond cleavage. GCase is synthesized in the , undergoes in the Golgi apparatus, and is trafficked to lysosomes via mannose-6-phosphate receptors, where it achieves optimal activity at pH 5.5. Beyond , heterozygous GBA1 mutations are a significant genetic risk factor for , as reduced GCase activity impairs α-synuclein clearance and promotes lysosomal dysfunction in neurons. Therapeutically, recombinant GCase enzymes like imiglucerase and velaglucerase alfa are used for enzyme replacement therapy in type 1, while substrate reduction therapies such as inhibit GlcCer synthesis to alleviate substrate accumulation. Ongoing research explores small-molecule chaperones to stabilize mutant GCase and approaches to restore GBA1 expression; as of 2025, AAV-based gene therapies such as FLT201 are in clinical trials for . This highlights the enzyme's critical role in both metabolic and neurodegenerative contexts.

Gene and Expression

Genomic Organization

The GBA1 gene, officially symbolized as GBA1 (also known as GBA, GCB, or GLUC), encodes the lysosomal enzyme glucocerebrosidase and is located on the long arm of at cytogenetic band 1q22. In the GRCh38.p14 assembly, the gene occupies genomic positions 155,234,452 to 155,244,627 on the complementary strand, spanning approximately 10 kb of DNA. This positioning places GBA1 within a gene-rich region of , flanked by nearby genes such as MTX1. The GBA1 genomic structure comprises 11 exons and 10 introns, with the coding sequence distributed across these exons to produce a mature mRNA of about 1.9 kb after , which can yield multiple transcript variants. A highly homologous , denoted GBAP1 or GBA (GBAP), lies approximately 16 kb downstream of GBA1 on the same , sharing over 96% sequence identity but containing deletions and point mutations that render it non-functional. This close proximity facilitates unequal recombination and gene conversion events between GBA1 and GBAP1, increasing the risk of structural variants and complicating . More than 300 pathogenic variants have been identified in GBA1, predominantly missense mutations, but also including , frameshift, splice-site, and complex rearrangements arising from interactions. Key examples include the c.1226A>G (p.N409S or N370S) variant in 9, the most prevalent in non-neuronopathic and classified as mild due to residual enzyme activity of 15-20%; and the c.1448T>C (p.L483P or L444P) variant in 10, associated with severe forms and near-complete loss of function. Variants are broadly categorized by severity—severe (e.g., L444P, with <5% activity), mild (e.g., N370S), and complex/risk types—based on their biochemical impact and recombination involvement, as established through functional assays and clinical correlations. The GBA1 gene demonstrates strong evolutionary conservation across metazoans, with orthologs identifiable in vertebrates such as mice (Gba) and zebrafish (gba.1), reflecting its essential role in lysosomal lipid metabolism. At the protein level, glucocerebrosidase belongs to glycoside hydrolase family 30 (GH30) in the CAZy classification, exhibiting sequence and structural homology to other lysosomal beta-glucosidases and retaining catalytic residues conserved from bacterial ancestors, which underscores its ancient origin as a sphingolipid-degrading enzyme.34152-3/fulltext)

Expression Patterns

The GBA1 gene, encoding , displays ubiquitous mRNA expression across human tissues, with notably elevated levels in macrophage-rich sites such as the spleen, liver, bone marrow, and leukocytes, reflecting its role in glycolipid degradation within the mononuclear phagocyte system. In contrast, expression is lower in neural tissues like the brain and in fibroblasts, consistent with reduced lysosomal demands in these cell types. Data from the GTEx consortium indicate median transcripts per million (TPM) values ranging from approximately 10-15 in brain regions to 30-40 in spleen and liver, demonstrating at least a 2- to 4-fold variation across tissues. Similarly, RNA-seq analyses in the confirm this pattern, with highest RNA detection in hematopoietic and hepatic samples. GeneCards integration of GTEx data further highlights overexpression in whole blood by about 6.3-fold relative to average tissue levels. Transcriptional regulation of GBA1 involves the transcription factor EB (TFEB), which binds to coordinated lysosomal expression and regulation (CLEAR) motifs in the gene's promoter, facilitating coordinated upregulation of lysosomal genes under nutrient or stress conditions. The gene is also responsive to lysosomal stress pathways, particularly the unfolded protein response (UPR), where the transcription factor CHOP induces GBA1 expression to mitigate endoplasmic reticulum stress and support lysosomal biogenesis. These mechanisms ensure adaptive expression in response to physiological demands, such as inflammation or metabolic shifts. Post-transcriptional control contributes to GBA1 expression dynamics, with developmental upregulation observed during macrophage differentiation; for instance, enzyme activity is highest in human monocyte-derived macrophages compared to other cell lines, underscoring enhanced translation in immune cells. RT-PCR and RNA-seq studies across cell types reveal up to 10-fold differences in mRNA abundance between leukocytes and fibroblasts, emphasizing cell-specific posttranscriptional fine-tuning for lysosomal function.

Protein Structure and Properties

Primary and Tertiary Structure

Glucocerebrosidase, also known as acid β-glucosidase or GCase, is a lysosomal enzyme encoded by the GBA1 gene on chromosome 1q22. The mature protein comprises 497 amino acid residues following cleavage of the N-terminal signal peptide, with a calculated polypeptide molecular weight of approximately 55.6 kDa that increases to about 59.7 kDa due to post-translational modifications, primarily N-linked glycosylation. The enzyme possesses five consensus N-linked glycosylation sequons (Asn-X-Ser/Thr), but occupation typically occurs at four sites—Asn19, Asn59, Asn146, and Asn270—with the fifth at Asn462 remaining unoccupied in mammalian cells; these modifications are essential for proper folding, stability, and trafficking to the lysosome. The tertiary structure of glucocerebrosidase is characterized by a compact, globular fold divided into three distinct domains, as revealed by . Domain I (residues 1–27 and 383–414) consists of a three-stranded antiparallel β-sheet flanked by short loops, contributing to the overall scaffold. Domain II (residues 28–259) adopts a mainly α-helical architecture with eight α-helices arranged in a bundle. Domain III (residues 260–382 and 415–497) forms the catalytic core as a (β/α)8 , where alternating β-strands and α-helices create a cleft housing the . This tri-domain organization positions the at the interface of Domain III's barrel, accessible via a narrow pocket lined by hydrophobic residues. Within the of Domain III, the catalytic machinery consists of a dyad formed by Glu235, which functions as the general acid/base catalyst, and Glu340, serving as the nucleophilic residue that forms a covalent glycosyl-enzyme during . Structural studies indicate that binding of the activator Saposin C induces conformational changes, particularly in flexible loops surrounding the active site (such as those involving residues 396–399 and 311–318), which widen the substrate-binding pocket and enhance catalytic efficiency on membrane-embedded glucosylceramide. Glucocerebrosidase exhibits a propensity for dimerization in , which can influence its stability and activity, though the functional predominates in the . This dimerization is modulated by intramolecular bonds that stabilize the tertiary fold, including the pairs Cys4–Cys16 and Cys18–Cys23 in Domain I, along with a conserved free Cys342 near the ; mutations disrupting these bonds lead to misfolding and reduced stability. A recent 2025 cryo-TEM structure of the enzyme in complex with its lysosomal transporter LIMP-2 at 3.7 Å resolution further illuminates these dynamics, revealing minimal large-scale conformational shifts but highlighting flexibility in the loops and the role of hydrophobic interactions in partner binding.

Physicochemical Characteristics

Glucocerebrosidase exhibits optimal enzymatic activity in an acidic environment, with a range of 4.7–5.9 that aligns with lysosomal conditions. This dependence ensures efficient of glucosylceramide within the , where the enzyme functions as a retaining β-glucosidase. The enzyme demonstrates moderate , with a temperature (Tm) of approximately 55°C for recombinant forms, indicating vulnerability to elevated temperatures; active site occupancy by inhibitors or substrates can enhance this stability by up to 21°C. Its half-life is notably short, on the order of minutes at temperatures slightly above physiological levels, but lysosomal half-life extends to 32–48 hours, which can be prolonged by such as saposin C or phospholipids that prevent proteolytic degradation. contributes to overall stability, shielding the protein from denaturation, though this is secondary to active site protection. The of glucocerebrosidase is approximately 6.0–7.0 for the precursor form, shifting to more acidic values (4.3–5.0) upon sialylation, which influences its in aqueous buffers at neutral . The is generally soluble in lysosomal-like buffers but prone to aggregation, particularly for mutant variants, unless stabilized by pharmacological chaperones that promote proper folding and prevent misfolding-induced precipitation. Spectroscopically, glucocerebrosidase absorbs light maximally at 280 nm, attributable to its 12 residues, enabling concentration determination down to micromolar levels. or binding often quenches intrinsic or induces a shift in emission spectra, reflecting conformational changes near the . Key inhibitors include the irreversible agent conduritol B , with an of 4.3–9.5 μM, which covalently modifies the catalytic Glu340. Reversible inhibitors, such as isofagomine, bind competitively with low micromolar affinity, stabilizing the without permanent inactivation.

Enzymatic Function

Catalytic Mechanism

Glucocerebrosidase catalyzes the of (Glcβ1-1'Cer), a β-glycosidic linkage between glucose and , to yield free glucose and as products. This reaction occurs optimally at acidic around 5.5, consistent with the lysosomal environment. As a member of the glycoside hydrolase family 30 (GH30), glucocerebrosidase employs a retaining catalytic that preserves the β-configuration at the anomeric carbon of the substrate. The process follows a classical double-displacement involving two sequential steps. In the first step (), the catalytic Glu340 deprotonates and attacks the anomeric C1 carbon of , displacing the aglycone and forming a covalent β-glucosyl-enzyme intermediate; concurrently, Glu235 serves as the general acid to protonate the departing leaving group. In the second step (deglycosylation), Glu235, now acting as the general base, deprotonates and activates a molecule to perform a nucleophilic attack on the C1 carbon of the intermediate, hydrolyzing the glycosyl-enzyme bond and releasing β-glucose while regenerating the . The enzyme adheres to Michaelis-Menten kinetics for substrate binding and turnover. Representative kinetic parameters for the natural substrate include a K_m of approximately 100-200 μM and a k_{cat} of about 50 s⁻¹ at pH 5.5, reflecting efficient under lysosomal conditions.

Regulation and Modulators

Glucocerebrosidase (GCase) activity is primarily activated by saposin C, a small lysosomal activator protein that facilitates substrate presentation through lipid transfer mechanisms. Saposin C binds to GCase and anionic phospholipids such as 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), promoting membrane destabilization and enhancing enzyme-substrate interactions; this results in up to a 10-fold increase in the catalytic rate (kcat) under optimal conditions. The activation requires the presence of negatively charged phospholipids to anchor the complex to lysosomal membranes, ensuring efficient of glucosylceramide. Endogenous inhibitors of GCase include aggregates, which directly interact with the enzyme and impair its lysosomal trafficking and activity. In primary neuron models and postmortem (PD) brain tissue, overexpression or accumulation reduces GCase activity by approximately 50% in lysosomal fractions, exacerbating substrate buildup and contributing to pathological loops in synucleinopathies. Pharmacological modulators, such as , act as chemical chaperones that stabilize mutant GCase forms, increasing enzymatic activity and protein levels in patient-derived fibroblasts and mouse models of (GD) and PD. Allosteric modulation of GCase occurs through sites distinct from the catalytic pocket, influencing dimerization and . For instance, quinazoline-based small molecules like JZ-4109 bind at the dimer interface (e.g., near Lys346), promoting conformational shifts that enhance activity without competing at the ; this effect is pH-dependent, with dimer formation predominant at lysosomal 5.0. GCase exhibits optimal activity in the acidic range of 4.5–5.5, aligning with its lysosomal localization. Recent 2025 structural studies using cryo-electron microscopy (cryo-EM) have revealed the GCase-LIMP-2 transporter complex at 3.7 resolution, identifying key interaction sites that inform the design of small-molecule activators. These activators target allosteric pockets to stabilize the active conformation, preserving LIMP-2 binding while boosting lysosomal function, with potential applications in and therapies currently advancing in preclinical trials.

Physiological Role

Role in Lipid Metabolism

Glucocerebrosidase, also known as acid , serves as a critical in the lysosomal of glycosphingolipids, primarily hydrolyzing glucosylceramide (GlcCer) into and glucose. This process is essential for preventing the accumulation of GlcCer, which arises from the continuous turnover of cellular membranes and the degradation of more complex glycosphingolipids during . In cells with high membrane dynamics, such as macrophages, glucocerebrosidase ensures efficient recycling of , maintaining lysosomal and supporting overall balance. Within the broader pathway, glucocerebrosidase functions downstream of glucosylceramide (UGCG), the responsible for GlcCer in the Golgi apparatus from and UDP-glucose. GlcCer serves as a precursor for complex glycosphingolipids, including gangliosides, which are sequentially degraded in lysosomes through exoglycosidase actions; the pathway culminates with glucocerebrosidase's of GlcCer as the final step before breakdown. Downstream products, such as , are further metabolized to and fatty acids, with phosphorylatable to (S1P), a bioactive involved in signaling pathways. This integration positions glucocerebrosidase at a pivotal node, linking glycosphingolipid and degradation to sphingolipid-mediated cellular signaling. Glucocerebrosidase contributes significantly to turnover in macrophages, where it processes substantial GlcCer loads from phagocytosed material and recycling. Imbalances in its activity, as observed in recent studies on , lead to elevated hexosylceramides (including GlcCer), disrupting metabolic flux and promoting neuroinflammatory responses. The enzyme also interacts with upstream hydrolases in glycosphingolipid pathways, notably acting after acid , which converts lactosylceramide to GlcCer during the of gangliosides like GM1. This sequential interplay ensures coordinated of complex , with glucocerebrosidase's efficiency influencing the overall flux through interconnected lysosomal pathways.

Cellular Localization and Trafficking

Glucocerebrosidase (GCase), encoded by the GBA1 gene, is targeted to the via interaction with lysosomal 2 (LIMP-2, also known as SCARB2), bypassing the canonical mannose-6-phosphate receptor pathway used by most lysosomal hydrolases. This binding occurs in the () and is essential for proper sorting, as demonstrated in cellular models where LIMP-2 deficiency leads to mislocalization of GCase to the secretory pathway and reduced lysosomal enzyme activity. Although early studies suggested potential mannose-6-phosphate involvement, subsequent research confirmed the LIMP-2-dependent mechanism as the primary route, with LIMP-2 itself potentially utilizing mannose-6-phosphate for its own trafficking. The N-linked sites on GCase contribute to this process by promoting stable complex formation with LIMP-2. The trafficking itinerary of GCase begins in the , where it folds and associates with LIMP-2, followed by transport through the Golgi apparatus to late endosomes and ultimately the . Within the acidic environment of the (pH ~4.5-5.0), protonation of specific residues disrupts the GCase-LIMP-2 interaction, releasing the enzyme into the lysosomal for activity. This pathway is supported by phosphatidylinositol 4-kinases, which generate lipids necessary for vesicle formation and transport efficiency. While the exact of transit vary by , studies in fibroblasts indicate rapid progression from synthesis to lysosomal maturation, with functional detectable within hours. In steady-state conditions, GCase is predominantly distributed within lysosomes, where it associates with the inner membrane and lumen to access its substrates. Biochemical and studies reveal that the majority of mature GCase resides in this compartment, with minor fractions detectable in endosomal intermediates or occasionally at the plasma membrane due to incomplete sorting or . mechanisms involving endosomal-lysosomal help maintain this distribution, allowing reutilization of the before proteolytic by cathepsins, resulting in a relatively short lysosomal . Recent investigations into lysosomal have highlighted disruptions in GCase localization under pathological conditions. In ATP13A2 knockout models, which mimic aspects of and Kufor-Rakeb syndrome, lysosomal accumulation of polyamines (e.g., ) elevates luminal by ~0.2 units and induces electrostatic interference between GCase and lysosomal like bis(monoacylglycero)phosphate, thereby impairing function without altering total protein levels or overt trafficking defects. These findings, observed in human induced pluripotent stem cell-derived neurons and mouse brain tissue, underscore polyamines as modulators of lysosomal efficacy, with potential implications for neurodegenerative disorders linked to GBA1 variants.

Disease Associations

Gaucher Disease

Gaucher disease is an autosomal recessive lysosomal storage disorder resulting from pathogenic variants in the GBA1 gene, which encodes the enzyme glucocerebrosidase, leading to deficient activity of this lysosomal . This deficiency impairs the breakdown of , a derived from degradation. The disorder is classified into three main types based on clinical presentation and neurological involvement: type 1 (non-neuronopathic), which accounts for more than 90% of cases and primarily affects visceral organs; type 2 (acute neuronopathic), a severe infantile form with rapid neurological deterioration; and type 3 (subacute neuronopathic), which features progressive neurological symptoms developing in childhood or later. Common mutations associated with the disease include N370S, which is particularly prevalent in Ashkenazi Jewish populations. The pathophysiology of Gaucher disease centers on the accumulation of undegraded within lysosomes of macrophages, transforming these cells into characteristic Gaucher cells with a "crinkled " appearance due to lipid-laden . These lipid-engorged macrophages infiltrate and proliferate in the , liver, and , causing such as and , as well as hypersplenism that contributes to and . In bone tissue, Gaucher cell infiltration leads to , crises (acute episodes of severe pain and inflammation), osteonecrosis, and pathological fractures, resulting from disrupted remodeling and vascular compromise. Type 1 disease spares the , while types 2 and 3 involve neuronal accumulation, leading to , neuronal loss, and symptoms like oculomotor abnormalities and seizures. The global incidence of is estimated at 1 in 40,000 to 60,000 live births, with higher rates in certain populations due to founder effects. In Ashkenazi Jewish individuals, the carrier frequency is approximately 1 in 15, substantially elevating disease compared to the general population. Epidemiological data from , based on the national Gaucher Disease Registry, indicate 706 confirmed cases diagnosed between 1980 and 2024, with an incidence of 0.21 per 1,000,000 person-years. Diagnosis of typically begins with clinical suspicion based on symptoms like unexplained or , followed by confirmation through activity measuring glucocerebrosidase levels in peripheral leukocytes, cultured fibroblasts, or dried spots, where activity below 15% of is diagnostic. identifies biallelic GBA1 variants, aiding in subtype classification and carrier screening, particularly in high-risk populations. Elevated chitotriosidase activity, produced by activated macrophages, serves as a sensitive for monitoring and response, often increased up to 1,000-fold in affected individuals. Glucocerebrosidase (GCase), encoded by the GBA1 gene, represents the most common genetic factor for (), with variants conferring a substantially elevated lifetime compared to the general . Severe GBA1 mutations are associated with odds ratios for PD development ranging from approximately 10 to 21, while milder variants yield odds ratios of 3 to 5. Heterozygous carriers of GBA1 variants face a lifetime PD estimated at 5-30%, depending on the specific mutation and , far exceeding the 1-2% baseline in non-carriers. This genetic predisposition highlights GBA1's role in PD susceptibility, independent of full enzymatic deficiency. Reduced GCase activity due to GBA1 variants disrupts lysosomal function and promotes the aggregation of , a hallmark protein in . Lysosomal dysfunction impairs the degradation of , leading to its accumulation in neurons and exacerbating proteotoxic stress. Recent 2025 studies have further elucidated this pathway, demonstrating that elevated hexosylceramides—substrates of GCase—in brains induce neuronal gene upregulation mimicking pathogen responses, which in turn dysregulates GBA1 expression and perpetuates lipid imbalance. These mechanisms underscore a feed-forward loop linking partial GCase deficiency to progression. GBA1 variants also show associations with other neurodegenerative disorders, including (DLB). In DLB, GBA1 mutations increase disease risk similarly to , with carriers exhibiting accelerated cognitive decline and pathology. Biomarker research in 2024 has advanced non-invasive diagnostics, with blood-based assays measuring reduced GCase activity alongside elevated levels identifying PD subtypes with high specificity. The relationship between GCase and is bidirectional, as aggregated inhibits GCase activity, amplifying lysosomal impairment and severity. This inhibitory effect, observed in cellular models, forms a pathogenic feedback loop. Recent 2025 models using variants in induced pluripotent stem cell-derived neurons confirm this interaction, showing how oligomers disrupt GCase trafficking and function, thereby worsening neurodegeneration.

Therapeutic Developments

Enzyme Replacement Therapies

Enzyme replacement therapy (ERT) for glucocerebrosidase deficiency in began with alglucerase (Ceredase), derived from purified human placental glucocerebrosidase and approved by the FDA in 1991 as the first such treatment. Alglucerase was discontinued in the early due to concerns over risks from human-derived sources and limited supply, paving the way for recombinant alternatives. It was succeeded by imiglucerase (Cerezyme), a recombinant form of the produced in ovary (CHO) cells, which received FDA approval in 1994 and demonstrated comparable efficacy to alglucerase in treating type 1 . Imiglucerase is administered intravenously at a standard dose of 60 units per kg body weight every two weeks. The mechanism of imiglucerase relies on its pattern, featuring exposed residues that facilitate uptake by macrophages via -mediated endocytosis, targeting the primary site of accumulation in . Once internalized in lysosomes, the hydrolyzes into glucose and , alleviating substrate buildup in affected tissues. This macrophage-directed delivery enhances therapeutic efficiency, as these cells express high levels of the , facilitating uptake followed by lysosomal routing. In patients with type 1 , imiglucerase treatment typically reduces spleen volume by 30-50% within the first year, alongside improvements in liver volume and hematologic parameters such as hemoglobin levels and platelet counts, often evident within 6-12 months. Long-term therapy maintains these gains, with many patients achieving normalized blood counts and reduced , though may respond more slowly. However, ERT has limited efficacy in neuronopathic forms (types 2 and 3) due to poor blood-brain barrier penetration, restricting its benefits to peripheral manifestations. Approved alternatives include velaglucerase alfa (VPRIV), a recombinant produced via gene activation in a human fibroblast line and approved by the FDA in 2010, which offers similar macrophage targeting and dosing to imiglucerase with a favorable safety profile in clinical trials. Another option is taliglucerase alfa (Elelyso), expressed in plant s derived from suspension cultures and approved in 2012, providing an innovative method that avoids mammalian contaminants while achieving comparable reductions in spleen and liver volumes. Both alternatives have been optimized in the 2020s for manufacturing scalability and immunogenicity monitoring, serving as effective switches for patients intolerant to imiglucerase.

Novel Approaches

Substrate reduction therapy (SRT) represents a key novel approach for managing glucocerebrosidase (GCase) deficiencies by targeting upstream enzymes to decrease the accumulation of , the substrate hydrolyzed by GCase. , an iminosugar inhibitor of glucosylceramide synthase, was the first SRT approved for type 1 (GD) in patients unable to receive enzyme replacement therapy, demonstrating reductions in liver and volume as well as improvements in levels in clinical studies. Eliglustat, a more selective and potent glucosylceramide synthase inhibitor, was approved in 2014 as a first-line oral therapy for adults with type 1 GD, showing non-inferiority to imiglucerase in maintaining disease stability over 12 months, with comparable reductions in volume and improvements in platelet counts. These therapies reduce substrate load without directly augmenting GCase activity, offering a convenient alternative for long-term management, particularly in non-neuronopathic forms of GD. Pharmacological chaperones aim to stabilize misfolded GCase variants, promoting proper lysosomal trafficking and enhancing enzymatic activity in GD and GBA-associated (). , a mucolytic agent repurposed as a chaperone, binds to GCase to facilitate folding and increase lysosomal activity; preclinical studies showed up to a 35% elevation in GCase protein levels in the following . Phase 2 trials in GBA-PD patients confirmed ambroxol's safety and tolerability at high doses (up to 1,260 mg/day), with evidence of penetration and modest increases in GCase activity in , though motor and cognitive outcomes were not significantly altered over 18 months. Ongoing phase 2 trials as of 2025 continue to evaluate ambroxol's potential to slow progression in GBA variant carriers by boosting residual GCase function by approximately 20-30% in responsive genotypes. Allosteric activators of GCase, guided by structural insights, offer another investigational strategy to enhance enzyme function in both GD and PD. Recent cryo-electron microscopy (cryo-TEM) studies in 2025 resolved high-resolution structures of GCase in complex with its lysosomal transporter LIMP-2, enabling the design of small molecules that modulate trafficking to enhance GCase function and counteract lysosomal dysfunction in PD models. These preclinical activators, identified through , increase GCase catalytic efficiency by stabilizing the enzyme's active conformation and promoting Saposin C-mediated activation, demonstrating restored glucosylceramide clearance in cellular models of and reduced α-synuclein pathology in PD neurons. Early data suggest potential for oral delivery, with lead compounds showing dose-dependent activity enhancements in animal models without off-target effects on related lysosomal hydrolases. Gene therapy approaches seek durable restoration of GCase expression through viral vectors or , addressing root causes of GCase deficits in GD and GBA-PD. (AAV) vectors encoding wild-type GBA1, such as AAV9-GBA1, have been optimized for delivery via intrathecal or intracisternal administration, restoring enzymatic activity in preclinical and models of PD and type 2/3 GD; 2025 studies reported up to 50% normalization of brain GCase levels and suppression of α-synuclein accumulation following single-dose infusion. Phase 1/2 trials initiated in 2025 for GBA-PD patients demonstrate feasibility and tolerability, with interim data indicating sustained GCase expression in up to 12 months post-administration. For type 1 , liver-directed AAV therapies like FLT201 have entered phase 1 trials, demonstrating increased systemic GCase activity in early 2025 data. Complementing this, /Cas9 editing of GBA1 variants in induced pluripotent stem cells (iPSCs) from GD patients enables correction of pathogenic mutations, yielding isogenic lines with restored GCase activity; 2025 validations confirmed efficient in iPSC-derived neurons, reducing glucosylceramide accumulation and improving lysosomal function without off-target edits. These edited iPSCs provide platforms for personalized modeling and potential autologous cell therapies.

References

  1. [1]
    GBA1 glucosylceramidase beta 1 [ (human)] - NCBI
    Sep 9, 2025 · This gene encodes a lysosomal membrane protein that cleaves the beta-glucosidic linkage of glycosylceramide, an intermediate in glycolipid metabolism.
  2. [2]
    GBA1 - Lysosomal acid glucosylceramidase - Homo sapiens (Human)
    Glucosylceramidase that catalyzes, within the lysosomal compartment, the hydrolysis of glucosylceramides/GlcCers (such as beta-D-glucosyl-(1<->1')-N-acylsphing ...
  3. [3]
    Glucocerebrosidase and its relevance to Parkinson disease - PMC
    Aug 29, 2019 · Mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase, are among the most common known genetic risk factors for the development of ...
  4. [4]
    Gaucher Disease - StatPearls - NCBI Bookshelf
    Nov 12, 2023 · Gaucher disease results from mutations in the GBA1 gene, leading to deficient glucocerebrosidase activity within lysosomes.
  5. [5]
    A Review of Gaucher Disease Pathophysiology, Clinical ...
    Gaucher disease is caused by a deficiency in glucocerebrosidase (GCase) (or β-glucosidase), which leads to an accumulation of GlcCer.
  6. [6]
    Clinical, mechanistic, biomarker, and therapeutic advances in GBA1 ...
    Sep 12, 2024 · GBA1 encodes the lysosomal enzyme glucocerebrosidase (GCase), which is responsible for breaking down glucosylceramide (GlcCer) [11]. In this ...
  7. [7]
    Mechanism of glucocerebrosidase activation and dysfunction in ...
    The absence of fully functional GCase leads to the accumulation of its lipid substrates in lysosomes, causing Gaucher disease, an autosomal recessive disorder ...<|control11|><|separator|>
  8. [8]
    Cryo-TEM structure of β-glucocerebrosidase in complex with its ...
    Mar 30, 2025 · Mutations within the GCase-encoding gene GBA1 that impair enzyme function are the monogenic cause for Gaucher's disease (GD). In addition ...
  9. [9]
    Next-Generation Sequencing Analysis of GBA1 - Frontiers
    GBA1 is located in a gene-rich region on chromosome 1q21 that encompasses seven genes and two pseudogenes within an 85-kb region (Winfield et al., 1997). GBA1 ...
  10. [10]
    Entry - *606463 - GLUCOSIDASE, BETA, ACID; GBA - OMIM
    ... gene and the 3-prime end of the pseudogene. The location of a pseudogene near the functional gene for GBA on chromosome 1q may be the basis of disease ...
  11. [11]
    Association study of GBA1 variants with MSA based on ... - Nature
    Jul 18, 2024 · GBA1 consists of 11 exons spanning 7.6 kb, and a highly homologous pseudogene (GBA1LP, HGNC:4178) is located approximately 16 kb apart from the ...Missing: organization | Show results with:organization
  12. [12]
    Classification of GBA1 Variants in Parkinson's Disease: The GBA1 ...
    Jan 4, 2023 · GBA1 variants can be classified into three categories based on their role in Gaucher's disease (GD) or PD: severe, mild, and risk variant (for PD).
  13. [13]
    Classification of GBA1 variants in Parkinson's disease
    GBA1 variants can be classified into three categories based on their role in Gaucher's Disease (GD) or PD: severe, mild, and risk variant (for PD).
  14. [14]
    [PDF] GBA1 Variants and Parkinson's Disease
    7,8 The most common GD-causing genetic variants are the point mutations p.N409S (N370S) and p.L483P (L444P). In. GD, β-glucocerebrosidase activity is ...
  15. [15]
    GBA1 Gene - Glucosylceramidase Beta 1 - GeneCards
    A related pseudogene is approximately 12 kb downstream of this gene on chromosome 1. Alternative spl... See more... Aliases for GBA1 Gene. Aliases for GBA1 Gene.
  16. [16]
    Bulk tissue gene expression for GBA (ENSG00000177628.16)
    The Genotype-Tissue Expression (GTEx) project is an ongoing effort to build a comprehensive public resource to study tissue-specific gene expression and ...Missing: levels human
  17. [17]
    Tissue expression of GBA1 - Summary - The Human Protein Atlas
    The value ranges from 0 and 1, where 0 indicates identical expression across all cells/tissue types, while 1 indicates expression in a single cell/tissue type. ...
  18. [18]
    Direct and indirect regulation of β-glucocerebrosidase by ... - Nature
    Oct 22, 2024 · Like many other lysosomal genes, the GBA1 promoter contains two Coordinated Lysosomal Expression and Regulation (CLEAR) motifs, DNA ...
  19. [19]
    UPR activation and CHOP mediated induction of GBA1 transcription ...
    The results showed UPR activation in all tested cells. They also indicated that transcription of the GBA1 gene is upregulated through activation of the UPR- ...
  20. [20]
    Cultured Macrophage Models for the Investigation of Lysosomal ...
    Figure 1 shows that GBA1 activity is highest in HMDM cell lysate. A similar trend is observed for GAA, GALC, HEXA&B and GUSB activities. Strikingly, PMA matured ...<|control11|><|separator|>
  21. [21]
    Comparative physicochemical and biological characterization of the ...
    The calculated molecular mass of the amino acid sequence of imiglucerase is 55,574 Da, and the distribution of intact enzyme proteoforms around 60 kDa for ...
  22. [22]
    Human acid beta-glucosidase. N-glycosylation site occupancy and ...
    The five potential N-glycosylation sites (sequons) of human acid beta-glucosidase were individually mutated to determine site occupancy and the effect of site ...Missing: positions | Show results with:positions
  23. [23]
    X-ray structure of human acid-β-glucosidase, the defective enzyme ...
    Domain II, which is an immunoglobulin-like domain, is shown in green. The catalytic domain (domain III), which is a TIM barrel, is shown in blue, and the ...
  24. [24]
    The biochemical basis of interactions between Glucocerebrosidase ...
    Jan 22, 2020 · The five N-glycosylation sites (Asn 19, Asn 59, Asn 146, Asn 270, and Asn 462) are shown as purple spheres. The free cysteine residues are ...
  25. [25]
    Stabilization of Glucocerebrosidase by Active Site Occupancy
    May 9, 2017 · ABP 5 labeled various molecular weight forms of GBA in the range 58–66 kDa, stemming from modifications in the enzyme's N-linked glycans. (41) ...
  26. [26]
    Dissociation of glucocerebrosidase dimer in solution by its co-factor ...
    The structures of GCase and Sap C are shown in Fig. 1. Sap C is one of four saposin proteins resulting from proteolytic cleavage of the 70 kDa precursor ...
  27. [27]
    Glucocerebrosidase: Functions in and Beyond the Lysosome - NIH
    The storage of GlcCer in GD patients occurs almost exclusively in macrophages residing in the spleen, liver, bone marrow, lymph nodes, and lung (Figure 1C) [131] ...
  28. [28]
    Stabilization of Glucocerebrosidase by Active Site Occupancy - PMC
    This suggests again that ABP labeling markedly stabilizes GBA in vivo, since the half-life of unlabeled GBA is reported to be around 32–48 h. Lipophilic ABPs 4 ...
  29. [29]
    Remodeling the oligosaccharides on β‐glucocerebrosidase using ...
    Dec 13, 2011 · Oligosaccharides play a critical role in protein folding, solubility, stability, activity, and serum half-life (Kobata, 1992; Nagai et al., 1997 ...
  30. [30]
    Function of oligosaccharide modification in glucocerebrosidase, a ...
    Glucocerebrosidase is synthesised as a 62.5-kDa precursor with high-mannose-type oligosaccharide chains and an apparent native isoelectric point of 6.0–7.0.
  31. [31]
    Glucocerebrosidase and its relevance to Parkinson disease
    Aug 29, 2019 · Mutations in GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase, are among the most common known genetic risk factors for the development of ...
  32. [32]
    Conduritol B epoxide | Glycosylases - Tocris Bioscience
    Conduritol B epoxide is an irreversible glucocerebrosidase (GBA) inhibitor (K i = 53 μM; IC 50 = 4.28 - 9.49 μM). Also inhibitis lysosomal α-glucosidase
  33. [33]
    Isofagomine Induced Stabilization of Glucocerebrosidase
    Aug 7, 2025 · The iminosugar isofagomine (IFG), a competitive inhibitor and a potential pharmacological chaperone of GCase, is currently undergoing clinical ...Missing: ifosfamide | Show results with:ifosfamide
  34. [34]
    A Guided Tour of the Structural Biology of Gaucher Disease: Acid-β ...
    Mutations in both acid-β-glucosidase (GCase) and saposin C lead to Gaucher disease, the most common lysosomal storage disorder.
  35. [35]
    Molecular imaging of membrane interfaces reveals mode of ... - PNAS
    ... saposin C (sapC) t ... There is a 10-fold increase in the initial rate of hydrolysis at 2.5 μM sapC, which is ...
  36. [36]
    The role of saposin C in Gaucher disease - PMC - PubMed Central
    Sap C has an isoelectric point of pH 4.3–4.4 [4]. At pH 5.4 it undergoes a conformational change in which the α-helix bundle opens to a V-shaped, or ...
  37. [37]
  38. [38]
    Ambroxol effects in glucocerebrosidase and α‐synuclein transgenic ...
    Nov 18, 2016 · This study investigated the effect of ambroxol treatment on glucocerebrosidase activity and on α‐synuclein and phosphorylated α‐synuclein protein levels in ...
  39. [39]
    β-Glucocerebrosidase Modulators Promote Dimerization of β ... - NIH
    Our newly discovered allosteric site and observed GCase dimerization provide a new mechanistic insight into GCase and its noniminosugar modulators and ...Missing: disulfide Cys18 Cys23
  40. [40]
    Glucocerebrosidase: Functions in and Beyond the Lysosome - MDPI
    A recent study in the United Kingdom revealed that 5–25% of patients with PD carry glucocerebrosidase gene mutations, and 10–30% of glucocerebrosidase carriers ...Glucocerebrosidase... · 3.1. Gcase Protein And Life... · 4. Gaucher Disease...
  41. [41]
    Elevated hexosylceramides in Parkinson's disease cause gene ...
    Aug 30, 2025 · Reduced glucocerebrosidase is associated with increased alpha-synuclein in sporadic Parkinson's disease. ... Glucocerebrosidase activity ...<|control11|><|separator|>
  42. [42]
    Prevalence, Genetics, and Inheritance - Know Gaucher Disease
    There are three types of GD. Type 1 Gaucher disease (GD1) is the most prevalent form of the disease, accounting for more than 90% of all cases. Unlike type 2 ...
  43. [43]
    Gaucher disease: the origins of the Ashkenazi Jewish N370S ... - NIH
    Type 1 disease is panethnic but is more prevalent in individuals of Ashkenazi Jewish (AJ) descent. Of the causative GBA mutations, N370S is particularly ...Missing: global | Show results with:global
  44. [44]
    Gaucher Disease - GeneReviews® - NCBI Bookshelf
    The diagnosis of GD is established in a proband by the finding of 0%-15% of normal glucocerebrosidase enzyme activity in peripheral blood leukocytes (or other ...
  45. [45]
    Clinical manifestations and management of Gaucher disease - PMC
    Gaucher disease is a rare multi-systemic metabolic disorder caused by the inherited deficiency of the lysosomal enzyme β-glucocerebrosidase.
  46. [46]
    Gaucher Disease in Bone: From Pathophysiology to Practice - PMC
    Jun 24, 2019 · GD is caused by the inherited deficiency of the acid hydrolase glucocerebrosidase causing an accumulation of its substrate glucosylceramide.
  47. [47]
    Gaucher Disease: Progress and Ongoing Challenges - PMC
    Now, deficient glucocerebrosidase was found to be responsible for all three types of Gaucher disease.
  48. [48]
    Gaucher Disease Type 1
    Gaucher disease type 1 (pronounced go-SHAY) is the most prevalent form of the disease in western countries, making up about 95 percent of cases there.
  49. [49]
    Type 1 Gaucher Disease Prevalence| Information for patients and ...
    1 IN 17. Gaucher disease is one of the most common genetic disorders in the. Ashkenazi population and has a carrier frequency of 1 in 17.
  50. [50]
    Epidemiology of Gaucher Disease in France: Trends in Incidence ...
    May 9, 2025 · Incidence, Prevalence and Mortality. In June 2024, the FGDR contained data of 706 patients with confirmed GD in France between 1980 and 2024 ...
  51. [51]
    Patient centered guidelines for the laboratory diagnosis of Gaucher ...
    Dec 21, 2022 · An enzyme activity result of less than 15% of normal activity in homogenates of leukocytes or fibroblasts is diagnostic of GD [129]. Residual ...
  52. [52]
    Marked elevation of plasma chitotriosidase activity. A novel hallmark ...
    We report the finding of a very marked increase in chitotrisidase activity in plasma of 30 of 32 symptomatic type 1 GD patients studied.
  53. [53]
    Alglucerase injection (Ceredase®) – Gaucher disease
    Dec 3, 2024 · The recombinant product Cerezyme®, was approved by the FDA in 1994, and shown to be as effective for the treatment of Type 1 Gaucher disease as ...
  54. [54]
    Imiglucerase in the treatment of Gaucher disease: a history and ...
    This article reviews the developmental history of enzyme therapy for Gaucher disease and discloses the authors' perceptions of the principles most likely to ...
  55. [55]
    Imiglucerase in the management of Gaucher disease type 1
    Oct 14, 2016 · Modification in the mannose residues improved its targeting to tissue macrophages and in 1990, Barton et al reported the first results with ...
  56. [56]
    Long-term hematological, visceral, and growth outcomes in children ...
    One year of imiglucerase greatly improved hemoglobin, platelets, and liver and spleen volumes. Improvements were maintained or grew further through 5 years of ...3. Results · 3.3. Hematologic Outcomes · 4. Discussion
  57. [57]
    [PDF] VPRIV (velaglucerase alfa for injection) - accessdata.fda.gov
    The active ingredient of VPRIV is velaglucerase alfa, which is produced by gene activation technology in a human fibroblast cell line. Velaglucerase alfa is ...Missing: method | Show results with:method
  58. [58]
    Taliglucerase alfa: safety and efficacy across 6 clinical studies in ...
    Feb 23, 2018 · Taliglucerase alfa is an enzyme replacement therapy (ERT) approved for treatment of adult and paediatric patients with Type 1 Gaucher ...
  59. [59]
    Velaglucerase alfa in the treatment of Gaucher disease type 1 - NIH
    Velaglucerase alfa has been approved as a safe and effective form of enzyme replacement therapy for the treatment of symptomatic individuals with Gaucher ...
  60. [60]
    Substrate reduction therapy: miglustat as a remedy for symptomatic ...
    Enzyme replacement therapy is proven to be safe and effective in the treatment of GD type 1, establishing imiglucerase as the current standard of care.
  61. [61]
    Enzyme Replacement or Substrate Reduction? A Review of ... - NIH
    Both miglustat and eliglustat demonstrate efficacy in reducing symptoms of Gaucher disease in patients who either cannot tolerate or refuse treatment with ...
  62. [62]
    Ambroxol in Parkinson's: results of phase 2 trial published
    The drug was also found to cross the blood-brain barrier; it achieved a good level of penetrance into the brain, and there was a 35% increase overall in levels ...Missing: chaperone 2024
  63. [63]
    Ambroxol as a disease-modifying treatment to reduce the risk of ...
    Nov 24, 2023 · In PD, two phase II trials that studied the effects of ABX have been completed. ... Pilot study using ambroxol as a pharmacological chaperone in ...
  64. [64]
    Study protocol of the GRoningen early-PD Ambroxol treatment ...
    May 1, 2024 · This trial is a single-center, double-blind, randomized, placebo-controlled study, including 80 PD patients with a GBA mutation, receiving either ambroxol 1800 ...
  65. [65]
    High-throughput screening for small-molecule stabilizers of ...
    Glucocerebrosidase (GCase) is implicated in both a rare, monogenic disorder (Gaucher disease, GD) and a common, multifactorial condition (Parkinson's disease, ...Missing: cryo- EM
  66. [66]
    AAV gene therapy for GBA-PD and Gaucher Disease - bioRxiv
    Jun 18, 2025 · Here, we report an AAV-mediated GBA1 replacement strategy to treat GD and GBA-PD by a one-time infusion via intravenous (GD Type 1) or intra-CSF ...
  67. [67]
    Investigational Gene Therapies for Parkinson's Disease - PMC
    Jul 10, 2025 · Intracisternal administration of AAV9-GBA1 (PR001, renamed LY3884961) gene therapy is currently being investigated in an open-label phase 1/2a ...
  68. [68]
    Efficient GBA1 editing via HDR with ssODNs by ... - Frontiers
    Apr 29, 2025 · A study of 240 patients with Gaucher Disease found that 12% of alleles at GBA1 were recombinant alleles, most arising from gene conversion, ...
  69. [69]
    Study Validates New Models to Study GD-Linked GBA1 Variants
    Jul 7, 2025 · In this study, researchers confirmed that CRISPR-edited isogenic induced pluripotent stem cell (iPSC) lines carrying pathogenic GBA1 variants ...