BACE1 Human

Beta-Secretase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of BACE1

BACE1 is a type I transmembrane protein with distinct structural domains:

DomainFunction
Signal peptide (1–21)Directs protein trafficking to the endoplasmic reticulum .
Pro-domain (22–45)Removed by furin in the Golgi apparatus to activate the enzyme .
Catalytic domain (46–460)Contains two aspartic residues (D93, D289) essential for proteolytic activity .
Transmembrane domain (461–477)Anchors BACE1 to membranes, influencing subcellular localization .
Cytoplasmic tail (478–501)Regulates intracellular trafficking and palmitoylation .

BACE1 functions as a dimer, with its catalytic activity dependent on pH (optimal at ~4.5) and substrate accessibility in acidic compartments like endosomes .

Amyloid-Beta Production

BACE1 initiates Aβ generation by cleaving amyloid precursor protein (APP) at the β-site, producing a membrane-bound C99 fragment. Subsequent cleavage by γ-secretase yields Aβ peptides (e.g., Aβ40, Aβ42), which aggregate into plaques in AD . Competing cleavage by α-secretase prevents Aβ formation, producing non-toxic P3 fragments .

Non-APP Substrates and Functions

Recent studies identify over 60 BACE1 substrates, including:

SubstrateRolePathway/Outcome
Neuregulin 1 (NRG1)Myelination of peripheral nerves .Critical for Schwann cell development.
Voltage-gated sodium channel β-subunitsNeuronal excitability .Modulates pain sensitivity .
OccludinBlood-brain barrier (BBB) integrity .Cleavage disrupts tight junctions, causing BBB leakage .
SEZ6, CHL1, APLP1Neurite outgrowth and synaptic plasticity .Implicated in neurodevelopmental processes.

BACE1 also regulates energy metabolism, immune responses, and tumor suppression .

Genetic and Clinical Evidence

  • Elevated BACE1 Activity: Observed in sporadic AD brains, correlating with Aβ plaque burden .

  • Protective Mutation: A single residue mutation (A673T) in APP reduces BACE1 cleavage efficiency, lowering AD risk .

  • Knockout Studies: BACE1−/− mice show reduced Aβ levels but exhibit motor deficits, hyperactivity, and altered emotional behaviors .

Endothelial Dysfunction

Hypertension-induced BACE1 overexpression in cerebral endothelial cells cleaves occludin, triggering caveolin-1-mediated tight junction degradation and nitric oxide synthase (eNOS) inhibition. This cascade contributes to small vessel disease and vascular dementia .

Clinical Trials

CompoundDeveloperPhaseOutcome
Verubecestat (MK-8931)Merck & Co.IIIHalted (2017): No efficacy in mild-to-moderate AD .
Lanabecestat (AZD3293)AstraZeneca/Eli LillyII/IIITerminated (2018): Futility in cognitive endpoints .
LY2886721Eli LillyIIDiscontinued: Hepatotoxicity .

Key Challenges

  • Mechanism-Based Toxicity: BACE1 inhibition impairs muscle spindle function and myelination, causing motor deficits .

  • Off-Target Effects: Early inhibitors (e.g., LY2886721) showed liver toxicity unrelated to BACE1 .

  • Selectivity: BACE2 inhibition risks disrupting melanocyte function (e.g., PMEL processing) .

Reversing Amyloid Pathology

Deleting BACE1 in adult 5xFAD mice reduced Aβ plaques and improved cognitive performance in fear conditioning tests, suggesting that late-stage BACE1 inhibition could still be therapeutic .

Novel Substrate Discovery

Quantitative proteomics identified 68 putative BACE1 substrates, including contactin-2 and L1CAM, highlighting roles in cell adhesion and signaling .

Product Specs

Introduction

BACE1, also known as beta-secretase 1 or beta-site amyloid precursor protein cleaving enzyme 1, is a protein encoded by the BACE1 gene in humans. This protein is primarily found in neurons and plays a vital role in myelin sheath formation in the peripheral nervous system. BACE1 is a transmembrane protein, meaning it is embedded within the cell membrane. It functions as a dimer, with two active sites (aspartate residues) located in its extracellular domain.

Description

This product consists of human BACE1, produced in Sf9 insect cells using a baculovirus expression system. It is a single, glycosylated polypeptide chain with a molecular weight of 49.2 kDa. The recombinant protein encompasses amino acids 22-457 and includes a 6-amino acid His tag at the C-terminus. Purification is achieved through proprietary chromatographic methods.

Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation

The BACE1 protein is supplied in a solution of Phosphate-Buffered Saline (pH 7.4) containing 10% glycerol.

Stability

For optimal storage, keep the vial at 4°C if using within 2-4 weeks. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing of the product.

Purity
Greater than 90% purity as determined by SDS-PAGE analysis.
Biological Activity

The specific activity is greater than 5 pmol/min/µg. One unit of activity is defined as the amount of enzyme required to convert 1.0 picomole of the substrate Mca-SEVNLDAEFRK(Dnp)RR-NH2 to MCA-Pro-Leu-OH per minute at a pH of 3.5 and temperature of 25°C.

Synonyms

Beta-Secretase, Membrane-Associated Aspartic Protease, Beta-Site APP Cleaving Enzyme, Beta-Site APP-Cleaving Enzyme, Aspartyl Protease, EC 3.4.23.46, Memapsin-2, Asp, BACE,  ASP2, Beta-Site Amyloid Beta A4 Precursor Protein-Cleaving Enzyme, Beta-Site Amyloid Precursor Protein Cleaving Enzyme, Transmembrane Aspartic Proteinase Asp2, Beta-Secretase 1 Precursor Variant, Beta-Site APP-Cleaving Enzyme, APP Beta-Secretase, EC 3.4.23, KIAA1149, HSPC104.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

TQHGIRLPLR SGLGGAPLGL RLPRETDEEP EEPGRRGSFV EMVDNLRGKS GQGYYVEMTV GSPPQTLNIL VDTGSSNFAV GAAPHPFLHR YYQRQLSSTY RDLRKGVYVP YTQGKWEGEL GTDLVSIPHG PNVTVRANIA AITESDKFFI NGSNWEGILG LAYAEIARPD DSLEPFFDSL VKQTHVPNLF SLQLCGAGFP LNQSEVLASV GGSMIIGGID HSLYTGSLWY TPIRREWYYE VIIVRVEING QDLKMDCKEY NYDKSIVDSG TTNLRLPKKV FEAAVKSIKA ASSTEKFPDG FWLGEQLVCW QAGTTPWNIF PVISLYLMGE VTNQSFRITI LPQQYLRPVE DVATSQDDCY KFAISQSSTG TVMGAVIMEG FYVVFDRARK RIGFAVSACH VHDEFRTAAV EGPFVTLDME DCGYNIPQTD ESTLMTHHHH HH.

Q&A

What is BACE1 and what is its primary function in humans?

BACE1 is an aspartyl protease of the pepsin family discovered in 1999. It functions as a type I transmembrane protein that serves as the β-secretase enzyme responsible for the initial cleavage of amyloid precursor protein (APP). This cleavage represents the rate-limiting step in the production of amyloid-β (Aβ) peptides implicated in Alzheimer's disease pathology. BACE1 is widely expressed in the brain, particularly in neurons, oligodendrocytes, and astrocytes, with subcellular localization primarily on plasma membranes and in endosomal compartments .

How is BACE1 structurally distinct from other proteases in its family?

BACE1 differs from other peptidases of the pepsin family (such as cathepsin D and E) through its transmembrane domain, making it uniquely suited for membrane protein processing. It shares 59% amino acid sequence homology with BACE2, its closest related protease. The structural domains of BACE1 include an ectodomain containing the catalytic site, a transmembrane domain, and a short cytosolic tail that enables its anchoring to cellular membranes while maintaining proteolytic activity .

What is the distribution pattern of BACE1 in human tissues?

While BACE1 shows highest expression in the brain and pancreas, it is also detected at lower levels in many peripheral tissues. Within the brain, BACE1 is particularly abundant in various neuronal cell types, but also present in glial cells. At the subcellular level, BACE1 localizes to plasma membranes and endosomal compartments, with notable concentration in healthy synaptic terminals. In Alzheimer's disease brains, significant BACE1 accumulation is observed in dystrophic neurites surrounding amyloid plaques .

What transcriptional mechanisms control BACE1 expression?

BACE1 expression is tightly regulated at the transcriptional level through multiple mechanisms. A key regulatory pathway involves NF-κB signaling, where NF-κB physically interacts with the human BACE1 promoter region. Research has demonstrated that increased NF-κB signaling up-regulates BACE1 expression, potentially creating a pathological cycle in inflammatory conditions. Epigenetic modifications also play a significant role, with H3 acetylation facilitating accessibility of the BACE1 promoter. Studies show this acetylation is increased in Alzheimer's disease models, and conversely, decreasing acetylated H3 in the BACE1 promoter regions reduces BACE1 mRNA levels .

How do post-transcriptional mechanisms regulate BACE1?

BACE1 undergoes substantial post-transcriptional regulation, particularly through microRNAs. Specific microRNAs including miR-298 and miR-328 recognize binding sites in the 3' untranslated region (UTR) of BACE1 mRNA and regulate BACE1 protein expression in neuronal cells. These microRNAs typically downregulate BACE1 expression, and their dysregulation has been implicated in elevated BACE1 levels observed in neurodegenerative conditions. This emerging field of transcriptomic signatures offers promising platforms for developing biomarkers, though diagnostic applications remain challenging due to concerns including restricted brain penetration and specificity issues .

What methodological approaches are optimal for measuring BACE1 protein levels?

Quantitative assessment of BACE1 protein levels presents significant technical challenges that researchers have addressed through optimized immunoassay development. ELISA-based approaches require careful antibody selection and validation. One successful strategy employs mouse anti-BACE1 ectodomain antibody (MAB9311) for capture and rabbit anti-BACE1 N-terminus antibody (B0681) for detection, which produces specific and intense signals. Sample preparation is critical—dilution in PBS followed by 10-minute incubation at 50°C significantly improves assay performance by partially unfolding BACE1 before capture. This process has been successfully applied to measure BACE1 in human brain lysates, platelets, plasma, and cerebrospinal fluid from Alzheimer's disease patients .

What are the major substrates of BACE1 beyond APP?

BACE1 cleaves numerous substrates beyond APP, serving diverse physiological functions. Neuronal substrates include SEZ6, L1CAM, leucine-rich repeat neuronal 1 (LRRN1), neurotrimin, and close homolog of L1 (CHL1), all involved in neurite outgrowth and neural development. BACE1 also regulates neuronal sodium channel metabolism through cleavage of voltage-gated sodium channel β2 subunit (Navβ2). Beyond neuronal substrates, BACE1 processes P-selectin glycoprotein ligand-1 (PSGL-1), which functions in immune cell recruitment, and interleukin-1 receptor II (IL-1R2), an inflammatory regulator. This diverse substrate profile explains BACE1's involvement in multiple physiological processes including neuronal development, inflammation, and metabolic regulation .

What experimental approaches are used to identify and validate new BACE1 substrates?

Identification of BACE1 substrates requires a multi-faceted experimental approach combining proteomics with validation studies. The process typically begins with comparative proteomic analysis of membrane proteins from wild-type versus BACE1 knockout tissues, or from tissues treated with BACE1 inhibitors versus controls. Mass spectrometry identifies proteins that accumulate in the absence of BACE1 activity. Candidate substrates then undergo validation through:

  • In vitro cleavage assays using recombinant BACE1 with purified candidate substrate

  • Cell-based assays with BACE1 overexpression or inhibition to confirm cleavage in cellular contexts

  • Analysis of substrate processing in BACE1 knockout animal models

  • Site-directed mutagenesis of predicted cleavage sites to confirm the exact position of BACE1 processing

This systematic approach has expanded our understanding of BACE1's physiological roles through identification of its diverse substrate repertoire .

How does BACE1 contribute to normal physiological processes?

BACE1 functions extend well beyond its pathological role in amyloidogenesis, as evidenced by the phenotype of BACE1 knockout mice. These animals exhibit developmental hypomyelination, suggesting BACE1's importance in normal myelin formation. They also display reduced weight gain and increased energy expenditure, indicating BACE1's role in energy metabolism and homeostasis. BACE1 influences neurite outgrowth and axon guidance through cleavage of neuronal substrates like SEZ6 and CHL1, functions particularly critical during development. The knockout mice show additional phenotypes including memory deficits, seizures, and abnormal EEGs, highlighting BACE1's diverse physiological functions. These roles are particularly pronounced during developmental stages, with axon guidance impairments being more severe when BACE1 is deficient early in life .

How is BACE1 activity altered in Alzheimer's disease?

In Alzheimer's disease, both BACE1 expression and enzymatic activity are significantly elevated. High BACE1 enzymatic activity has been documented in human AD brain extracts, consistent with neurons producing the highest levels of Aβ. A notable pathological finding is the accumulation of BACE1 in neuritic dystrophies surrounding amyloid plaques in both AD mouse models and human AD brains, occurring through a posttranslational mechanism. This localized increase potentially creates a pathological feed-forward loop where initial Aβ deposition leads to increased local BACE1 activity, generating more Aβ and accelerating plaque formation. Research has also shown that dysfunctional autophagy in mutant human neurons augments retention of BACE1 in distal axons, enhancing β-cleavage of APP and subsequent Aβ production .

What evidence supports BACE1's involvement in metabolic diseases?

Multiple experimental findings implicate BACE1 in metabolic disease pathophysiology. BACE1 is expressed in metabolically active tissues including pancreatic β-cells, adipocytes, and hepatocytes. BACE1 knockout mice display reduced weight gain, increased energy expenditure, and enhanced leptin signaling, indicating BACE1's involvement in energy homeostasis regulation. Conversely, BACE1 knock-in mice develop systemic diabetes-like symptoms. In pathological conditions such as type 2 diabetes and obesity, BACE1 expression and activity are increased, potentially driving disease progression. Weight loss observed in clinical trials of BACE1 inhibitors further supports BACE1's role in metabolism. The underlying mechanisms may involve BACE1's influence on glucose homeostasis and insulin signaling pathways through cleavage of various metabolic substrates .

What is known about BACE1's role in vascular pathology?

Emerging evidence indicates BACE1 contributes to vascular pathology, particularly affecting cerebral small vessels. Endothelial BACE1 has been implicated in cerebral small vessel impairment, potentially through mechanisms involving tight junction regulation . BACE1 expression in vascular cells may contribute to cardiovascular disease through proteolytic activity affecting vascular homeostasis. Additionally, BACE1's role in generating Aβ contributes to cerebral amyloid angiopathy, characterized by amyloid deposition in blood vessel walls. BACE1 may also influence vascular function through cleavage of substrates involved in inflammation and immune responses, such as PSGL-1, which affects leukocyte recruitment. These vascular effects represent an important emerging area of BACE1 research beyond its established neuronal functions .

What are the optimal methods for measuring BACE1 enzymatic activity?

Accurate assessment of BACE1 enzymatic activity requires consideration of several methodological factors. For in vitro studies, fluorescence resonance energy transfer (FRET)-based assays using synthetic peptides containing the BACE1 cleavage site from APP provide quantitative activity measurements. When working with tissue samples, sample preparation is critical—tissues should be homogenized in buffers that maintain BACE1 in its native conformation while separating it from potential endogenous inhibitors or activators. Activity measurements should be conducted at acidic pH (4.5-5.5) that mimics the endosomal environment where BACE1 is most active. Controls should include parallel samples treated with specific BACE1 inhibitors to confirm signal specificity. For tissues with lower BACE1 expression, more sensitive activity-based probes that covalently bind to active BACE1 may be preferable. Normalizing activity to BACE1 protein levels can distinguish changes in specific activity versus expression alterations .

How can researchers effectively distinguish between BACE1 and BACE2 activities?

Differentiating between BACE1 and BACE2 activities presents challenges due to their structural similarity (59% sequence homology) and overlapping substrate preferences. Several strategies can address this challenge:

  • Selective inhibitors with substantially higher affinity for one enzyme over the other

  • Substrate-based approaches exploiting different cleavage site preferences (e.g., BACE1 preferentially cleaves the Swedish mutant APP sequence)

  • Genetic approaches using siRNA knockdown or CRISPR-Cas9 genome editing to selectively reduce one enzyme

  • Immunological methods with validated, highly specific antibodies

  • Utilizing expression pattern differences, as some tissues predominantly express one enzyme

Most robust experimental designs employ multiple approaches simultaneously to confidently distinguish between these homologous enzymes' activities .

What are the key considerations for designing BACE1 inhibition studies?

Designing effective BACE1 inhibition studies requires addressing several critical factors. Inhibitor selectivity is paramount—compounds should distinguish between BACE1 and BACE2 to minimize off-target effects. For CNS studies, blood-brain barrier penetration is essential, requiring appropriate physicochemical properties. The timing of intervention requires careful consideration, as developmental versus adult BACE1 inhibition may yield different outcomes due to BACE1's role in developmental processes. Researchers should evaluate dose-dependent effects, as complete versus partial BACE1 inhibition produces different phenotypes, with partial inhibition potentially balancing therapeutic benefits against side effects. Assessment protocols should include both direct Aβ measurements and functional outcomes across multiple domains. Finally, monitoring for potential side effects based on known BACE1 substrates is essential, including effects on myelination, synaptic plasticity, and metabolic parameters .

How do researchers distinguish between direct and indirect effects of BACE1 manipulation?

Differentiating direct from indirect effects of BACE1 manipulation presents significant challenges due to BACE1's numerous substrates and downstream pathways. Several methodological approaches can address this challenge:

  • Substrate-specific knock-in models with mutated BACE1 cleavage sites for specific substrates

  • Rescue experiments restoring specific cleaved products in BACE1-deficient models

  • Temporal control using inducible systems to separate developmental from adult requirements

  • Cell-type specific BACE1 manipulation using Cre-loxP systems

  • Molecular analyses tracking immediate changes in substrate processing following acute BACE1 inhibition

  • Computational approaches modeling the complex network of BACE1 substrates

What explains the apparent contradictions between preclinical and clinical BACE1 studies?

Several factors may explain discrepancies between promising preclinical BACE1 inhibition studies and disappointing clinical trial results:

  • Intervention timing differences—animal studies typically begin treatment before pathology develops, while human trials involve patients with established disease

  • Dose-response relationships may differ between species, with complete BACE1 inhibition potentially causing adverse effects through disruption of physiological functions

  • Species-specific differences in BACE1 substrate profiles and relative importance

  • Disease heterogeneity in humans contrasts with homogeneous pathology in animal models

  • Longer treatment durations in humans may reveal side effects not apparent in shorter animal studies

These factors suggest that more nuanced approaches to BACE1 modulation, perhaps targeting specific pools of BACE1 or particular disease stages, may be required for successful clinical translation .

How might cell-type specific BACE1 functions contribute to disease heterogeneity?

Cell-type specific BACE1 functions likely contribute significantly to disease heterogeneity across pathologies. BACE1 is expressed in neurons, astrocytes, oligodendrocytes, microglia, and various peripheral cell types, with different expression levels and potentially different substrate preferences in each:

  • Neuronal BACE1 primarily contributes to Aβ production in Alzheimer's disease

  • Oligodendrocyte BACE1 influences myelination through processing of neuregulin-1

  • Microglial BACE1 may affect neuroinflammatory processes

  • Pancreatic β-cell BACE1 potentially impacts insulin secretion

  • Adipocyte BACE1 influences metabolic regulation

  • Vascular endothelial BACE1 affects blood vessel function

These diverse roles suggest that diseases might result from dysregulation of BACE1 in specific cell types, explaining why some patients show predominant cognitive symptoms while others present with metabolic or vascular phenotypes .

What are the most promising biomarker approaches for monitoring BACE1 activity in humans?

Developing reliable biomarkers for BACE1 activity in humans remains a significant challenge with several promising approaches:

  • Measurement of BACE1 enzyme levels in cerebrospinal fluid (CSF) using optimized ELISA methods

  • Quantification of BACE1-cleaved products of APP (sAPPβ) and other substrates in CSF

  • Monitoring of BACE1 activity using activity-based probes in accessible tissues

  • Development of PET ligands to visualize BACE1 distribution and activity in the brain

  • Analysis of BACE1-regulated microRNA signatures in blood or CSF

The most effective strategy may involve combining multiple biomarkers to create a comprehensive profile of BACE1 activity. Optimized protocols for sample preparation are critical, including procedures to partially unfold BACE1 before capture in immunoassays, which has been shown to significantly improve detection sensitivity .

What experimental models best capture human BACE1 biology across disease contexts?

The optimal experimental models for studying human BACE1 vary depending on the research question:

  • For neurodegenerative disease research: Transgenic mouse models expressing human APP with mutations enhancing BACE1 cleavage (such as the Swedish mutation)

  • For metabolic disease research: Mice with tissue-specific BACE1 manipulation in pancreas, liver, or adipose tissue

  • For human-specific cellular contexts: Induced pluripotent stem cells (iPSCs) differentiated into various cell types

  • For tissue-level interactions: Organoid models combining multiple cell types

  • For high-throughput screening: Cell lines with stable expression of human BACE1 and reporter substrates

  • For validation in the most relevant context: Post-mortem human tissues

The choice among these models should consider species differences, disease features recapitulated, and the specific BACE1 function being studied .

How might targeting specific BACE1 pools or conformations advance therapeutic approaches?

Emerging research suggests that targeting specific subcellular pools or conformational states of BACE1 may provide more precise therapeutic approaches than global BACE1 inhibition:

  • Targeting endosomal BACE1, where it primarily cleaves APP, while sparing cell surface BACE1 could reduce Aβ production while maintaining processing of some physiological substrates

  • Developing conformation-selective inhibitors that preferentially inhibit BACE1 when bound to specific substrates

  • Creating substrate-selective BACE1 modulators that alter enzyme conformation to preferentially prevent APP processing

  • Targeting BACE1 regulators rather than BACE1 itself, such as proteins that control BACE1 trafficking or activation

  • Developing molecules that selectively inhibit BACE1 in specific cell types where its activity contributes to pathology

These approaches might achieve therapeutic benefits while minimizing adverse effects associated with complete BACE1 inhibition, addressing the challenges that have hindered clinical translation of BACE1-targeted therapies .

Product Science Overview

Structure and Function

BACE1 is a type I membrane protein with a single transmembrane domain. It is synthesized as a precursor protein and undergoes post-translational modifications, including glycosylation and proteolytic cleavage, to become fully active . The enzyme cleaves the amyloid precursor protein (APP) at the β-site, producing a soluble APP fragment and a membrane-bound C-terminal fragment (C99). The C99 fragment is subsequently cleaved by γ-secretase to generate Aβ peptides .

Role in Alzheimer’s Disease

The accumulation of Aβ peptides in the brain is a hallmark of Alzheimer’s disease. These peptides aggregate to form amyloid plaques, which are believed to disrupt cell function and trigger neurodegenerative processes . Genetic mutations in APP or presenilin genes (components of γ-secretase) can increase the production of Aβ, leading to early-onset familial Alzheimer’s disease .

Therapeutic Target

Given its critical role in Aβ production, BACE1 has been a major target for drug development aimed at reducing Aβ levels in the brain. Inhibitors of BACE1 are being investigated as potential treatments for Alzheimer’s disease. However, developing effective BACE1 inhibitors has been challenging due to the enzyme’s broad substrate specificity and the need to avoid off-target effects .

Recombinant BACE1

Recombinant BACE1 is produced using various expression systems, including insect cells and mammalian cells like HEK 293 . These recombinant proteins are used in research to study the enzyme’s structure, function, and interactions with potential inhibitors. They are also employed in high-throughput screening assays to identify new BACE1 inhibitors .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.