CTSD Human

Cathepsin-D Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

  • Gene: The CTSD gene is located on chromosome 11 (11p15.5) and spans ~12 kb. It produces a single precursor protein (pre-pro-CTSD) that undergoes post-translational modifications to form a mature enzyme .

  • Protein: Mature CTSD is a dimer composed of a 34 kDa heavy chain and a 14 kDa light chain, linked by hydrophobic interactions. The catalytic site includes two aspartic residues (Asp33 and Asp231) .

PropertyDetails
Gene ID1509
Protein Size337 amino acids (mature form)
Post-Translational ProcessingCleavage of signal peptide (20 aa) and propeptide (44 aa) to form mature enzyme
Key DomainsHeavy chain (196 aa), light chain (141 aa), catalytic aspartates (D33, D231)

Biological Functions

CTSD exhibits dual roles in cellular processes:

  • Lysosomal Degradation: Cleaves proteins and activates precursors in lysosomes .

  • Apoptosis Regulation: Mature CTSD promotes apoptosis via cleavage of pro-apoptotic factors like BID .

  • Extracellular Signaling: Secreted pro-CTSD acts as a ligand to enhance cell proliferation and metastasis in cancers .

Neurodegenerative Disorders

  • Neuronal Ceroid Lipofuscinosis (NCL10): Biallelic CTSD mutations (e.g., F229I, W383C) cause early-onset neurodegeneration due to impaired lysosomal function .

  • Alzheimer’s Disease (AD): Elevated CTSD levels correlate with amyloid-beta aggregation, though AD-linked variants (e.g., G149V) show normal enzymatic activity .

  • Parkinson’s Disease (PD): The A239V mutation increases enzymatic activity, enhancing α-synuclein degradation and potentially slowing disease progression .

Cancer

  • Breast Cancer: CTSD overexpression is linked to tumor invasion and estrogen receptor signaling .

  • Colorectal Cancer (CRC): Upregulated CTSD promotes metastasis and survival under stress (e.g., serum deprivation) .

Mechanistic Studies

  • Monocyte-Derived Pro-CTSD: In diabetic models, monocytes secrete pro-CTSD, which increases cerebrovascular permeability via caveolin-1-dependent transcytosis .

  • Therapeutic Potential: Targeting CTSD maturation or secretion could mitigate neurodegenerative and oncogenic pathways .

Therapeutic Implications

  • Enzyme Replacement Therapy: Investigated for NCL10 but limited by blood-brain barrier penetration .

  • Small-Molecule Activators: Designed to enhance CTSD activity in PD models .

  • Cancer Therapeutics: Inhibitors (e.g., pepstatin A) reduce tumor growth in preclinical CRC models .

Product Specs

Introduction
Cathepsin D, initially a 54kDa precursor, undergoes proteolytic processing to a 48kDa single chain intermediate. This matures into a more stable two-chain form consisting of 34kDa and 14kDa units. This estrogen-regulated lysosomal protease is believed to aid cancer cell migration and invasion through the digestion of the basement membrane, extracellular matrix, and connective tissue. Due to its mitogenic and proteolytic properties, it's considered a potential prognostic marker for various tumor types. Cathepsin D is found in both epithelial cells and macrophages.
Description
Produced in HEK293 cells, CTSD is a single, glycosylated polypeptide chain. It comprises 398 amino acids (21-412 a.a.) and has a molecular weight of 43.4kDa. The protein is expressed with a 6 amino acid His tag at the C-terminus and undergoes purification using proprietary chromatographic methods.
Physical Appearance
A clear, sterile filtered solution.
Formulation
CTSD is supplied at a concentration of 1mg/ml in a buffer composed of 50mM MES (pH 5.5), 100mM NaCl, and 20% Glycerol.
Stability
For long-term storage, keep the vial at temperatures between -20°C and -80°C. This protein remains stable for 12 months when stored at the recommended temperature. Repeated freeze-thaw cycles should be avoided.
Purity
Purity of CTSD is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Cathepsin D, EC 3.4.23.5, CTSD, CPSD, CLN10, MGC2311.
Source

HEK293 Cells

Amino Acid Sequence

LVRIPLHKFT SIRRTMSEVG GSVEDLIAKG PVSKYSQAVP AVTEGPIPEV LKNYMDAQYY GEIGIGTPPQ CFTVVFDTGS SNLWVPSIHC KLLDIACWIH HKYNSDKSST YVKNGTSFDI HYGSGSLSGY LSQDTVSVPC QSASSASALG GVKVERQVFG EATKQPGITF IAAKFDGILG MAYPRISVNN VLPVFDNLMQ QKLVDQNIFS FYLSRDPDAQ PGGELMLGGT DSKYYKGSLS YLNVTRKAYW QVHLDQVEVA SGLTLCKEGC EAIVDTGTSL MVGPVDEVRE LQKAIGAVPL IQGEYMIPCE KVSTLPAITL KLGGKGYKLS PEDYTLKVSQ AGKTLCLSGF MGMDIPPPSG PLWILGDVFI GRYYTVFDRD NNRVGFAEAA RL-HHHHHH

Enzymatic Activity

> 20 pmol/min/ug, defined as the amount of enzyme which cleaves 1pmol of Mca-PLGLDpa-AR-NH2/min at pH-3.5 at 25C.

Q&A

What is the normal maturation process of human CTSD?

The maturation of human Cathepsin D involves multiple sequential steps across cellular compartments. CTSD is initially synthesized as an inactive precursor (proCTSD, ~52 kDa) comprising 412 amino acids. Following synthesis, the N-terminal signal peptide is cleaved within the endoplasmic reticulum (ER). The resulting inactive proCTSD undergoes glycosylation and is transported to the Golgi compartment, where mannose residues are phosphorylated to facilitate targeting to lysosomes via the mannose-6-phosphate pathway. Upon reaching the acidic environment of endosomes, proteolytic processing removes the 44-amino acid pro-peptide, generating an active intermediate form (~48 kDa). In lysosomes, this intermediate is further processed into a mature double-chain form consisting of an N-terminal light chain (~14 kDa) and a C-terminal heavy chain (~34 kDa) that remain non-covalently associated . This complex maturation pathway highlights the vulnerability of CTSD to mutations that may affect processing, localization, or enzymatic function.

How is CTSD expression distributed in human tissues?

While CTSD is ubiquitously expressed throughout human tissues, research indicates particularly high abundance in the brain compared to other tissues. Experimental comparisons between human cell lines demonstrate that neural-like cells (SH-SY5Y neuroblastoma and H4 neuroglioma) exhibit significantly higher levels of endogenous CTSD compared to non-neural cells such as human embryonic kidney cells (HEK293T). Importantly, these neural cell lines show similar CTSD expression levels to human dopaminergic neurons derived from induced pluripotent stem cells (iPSn), making them suitable models for studying CTSD function in neurological contexts . This brain-enriched expression pattern aligns with the prominent neurological manifestations observed in CTSD-related disorders.

What are the primary substrates of CTSD in the central nervous system?

CTSD functions as a lysosomal aspartic protease responsible for degrading numerous protein substrates within the central nervous system. Among its most clinically relevant substrates is α-synuclein (a-syn), a protein centrally implicated in Parkinson's disease pathogenesis. Both in vitro and in vivo mouse studies have demonstrated that a-syn undergoes proteolysis mediated by CTSD, while CTSD dysfunction facilitates α-synuclein accumulation and toxicity . Beyond a-syn, CTSD plays crucial roles in degrading various proteins involved in neuronal homeostasis, with its dysfunction leading to substrate accumulation in lysosomes. This accumulation is particularly detrimental to postmitotic cells like neurons, explaining why lysosomal storage disorders frequently present with neurological impairments and why many neurodegenerative diseases feature lysosomal dysfunction as a pathological hallmark .

What cell models are most appropriate for studying CTSD function?

Several cell models have proven valuable for investigating CTSD function in a research context. Human neuroblastoma (SH-SY5Y) and neuroglioma (H4) cell lines are particularly suitable due to their neural-like properties and high endogenous CTSD expression levels comparable to human dopaminergic neurons. For advanced studies, CRISPR/Cas9 technology has been employed to generate CTSD knockout (KO) lines in both SH-SY5Y and H4 backgrounds, providing essential null backgrounds for mutation analysis . Additionally, human induced pluripotent stem cells (iPSCs) differentiated into midbrain dopaminergic neurons offer physiologically relevant models, especially for studying CTSD in the context of Parkinson's disease. Patient-derived fibroblasts have also been utilized to analyze CTSD maturation and activity in cells carrying disease-associated mutations. For optimal experimental design, researchers should select models based on their specific research questions, with neural-derived cells being preferred for neurodegenerative disease investigations .

How is CTSD enzymatic activity accurately measured in experimental settings?

Measuring CTSD enzymatic activity requires careful attention to assay conditions and appropriate controls. The standard methodology employs fluorogenic peptide cleavage assays under acidic conditions (pH 4.5). In a typical protocol, cell lysates are prepared in a Triton-based buffer (50 mM sodium acetate, 0.1 M NaCl, 1 mM EDTA, 0.2% Triton X-100, pH 4.5) through incubation for 1 hour at 4°C. After centrifugation, the lysates are immediately used for activity determination, where 2 μl of cell extract is incubated with lysis buffer containing 0.1 μM quenched fluorogenic peptide and 0.05 mM Leupeptin at 37°C for 30 minutes . Critical controls include recombinant CTSD as a positive control and samples treated with the CTSD-specific inhibitor pepstatin A as negative controls. For comparative studies, activity measurements should be normalized to CTSD protein levels as determined by immunoblotting. To ensure assay specificity, researchers should also measure other lysosomal enzymes such as Cathepsin B and β-Glucocerebrosidase (GCase) to distinguish CTSD-specific effects from general lysosomal dysfunction .

What techniques are used to assess CTSD cellular localization?

Multiple complementary techniques are employed to accurately determine CTSD cellular localization. Immunofluorescence microscopy represents the primary approach, utilizing antibodies specific to CTSD alongside markers for different cellular compartments (lysosomes, endoplasmic reticulum, Golgi apparatus). This enables visualization of CTSD distribution throughout the cell and co-localization with organelle-specific proteins . Subcellular fractionation provides a biochemical approach to separate cellular components based on density, allowing quantitative assessment of CTSD levels in different fractions. Western blotting analysis of CTSD maturation forms also serves as an indirect indicator of localization, as the presence of mature CTSD (light and heavy chains) indicates successful trafficking to lysosomes where final processing occurs. For more precise localization studies, confocal microscopy with Z-stack imaging and quantitative co-localization analysis should be employed. Researchers investigating CTSD variants should always include wild-type CTSD and known localization-defective mutants (such as NCL10-associated mutations) as reference controls .

How do NCL10-associated CTSD mutations differ functionally from AD/PD-associated variants?

Detailed biochemical characterization reveals fundamental functional differences between NCL10-associated and AD/PD-associated CTSD variants. NCL10-associated mutations (including A58V, S100F, G149V, F229I, Y255X, W383C, and R399H) demonstrate severe defects in protein maturation and/or enzymatic activity. Most of these variants fail to undergo proper lysosomal trafficking, accumulating instead within the secretory pathway and exhibiting significantly reduced or absent mature CTSD forms . This profound loss of function aligns with the severe clinical manifestations observed in NCL10 patients, including early-onset neurodegeneration, progressive psychomotor retardation, and often premature death. In contrast, AD/PD-associated CTSD variants show relatively normal maturation, lysosomal localization, and enzymatic function in experimental analyses. The AD-associated A58V variant, located within the pro-peptide region that is cleaved during maturation, shows normal processing and activity levels comparable to wild-type CTSD . Interestingly, one PD-associated variant (A239V) actually exhibits enhanced enzymatic activity and improved α-synuclein degradation, suggesting potential neuroprotective properties rather than loss of function .

How does CTSD dysfunction contribute to α-synuclein accumulation in Parkinson's disease?

CTSD plays a critical role in α-synuclein homeostasis through direct proteolytic degradation, making CTSD dysfunction a potential contributor to pathological α-synuclein accumulation in Parkinson's disease. In vitro and in vivo studies have demonstrated that CTSD-mediated proteolysis represents a major pathway for α-synuclein clearance, with complete loss of CTSD function leading to significant α-synuclein accumulation and enhanced neurotoxicity . This relationship has been experimentally verified using CTSD knockout cell models as well as cells expressing NCL10-associated CTSD variants with severe loss of function. The importance of this degradation pathway is further underscored by research showing that even partial reduction in CTSD activity can accelerate α-synuclein aggregation under conditions of increased α-synuclein expression . Interestingly, the PD-associated A239V variant with enhanced enzymatic activity demonstrates improved α-synuclein degradation capacity, suggesting that natural CTSD variants with increased function may exist as potential protective factors against PD pathogenesis. These findings collectively highlight CTSD activation as a potential therapeutic strategy for reducing α-synuclein burden in PD .

What methodological approaches are used to distinguish between CTSD-specific effects and general lysosomal dysfunction?

Distinguishing CTSD-specific effects from general lysosomal impairment requires a multi-faceted methodological approach. Primary strategies include parallel assessment of multiple lysosomal enzymes alongside CTSD. Researchers typically measure the activities of other lysosomal hydrolases such as Cathepsin B and β-Glucocerebrosidase (GCase) using specific fluorogenic substrates, normalizing results to wild-type controls . Lysosomal mass quantification using markers like dextran blue (DexBlue) provides insight into whether alterations in lysosomal abundance might account for observed CTSD activity changes. Complementary approaches include lysosomal pH measurements using ratiometric probes to ensure that apparent CTSD dysfunction isn't simply due to altered lysosomal acidification, which would affect multiple acid hydrolases . For genetic studies, rescue experiments with wild-type CTSD cDNA in CTSD-deficient backgrounds can confirm phenotype specificity. Additionally, inhibitor studies using the CTSD-specific inhibitor pepstatin A alongside broader lysosomal inhibitors help delineate CTSD-dependent processes. These combined approaches enable researchers to confidently attribute observed phenotypes to CTSD dysfunction rather than general lysosomal impairment .

How do researchers address contradictory findings regarding CTSD A58V association with Alzheimer's disease risk?

The association between the CTSD A58V variant and Alzheimer's disease risk remains controversial, with conflicting results across multiple studies. To address these contradictions, researchers employ several methodological approaches. Meta-analyses combining data from multiple independent studies provide greater statistical power to detect true associations while minimizing the impact of sampling variability. Stratification by ethnicity, age, gender, and apolipoprotein E (APOE) genotype helps identify potential subgroup-specific effects that might explain discrepant findings . Functional studies examining the biochemical properties of the A58V variant complement genetic association analyses, with findings showing that A58V exhibits normal maturation and enzymatic activity despite its controversial disease association . Researchers also examine potential modifier genes that might interact with CTSD to influence disease risk. The scientific controversy is well-documented, with supporting studies (Papassotiropoulos et al., 2000; Mariani et al., 2006; Albayrak et al., 2010; Schuur et al., 2011; Sayad et al., 2014) and contradicting studies (Bagnoli et al., 2002; Mateo et al., 2002; Li et al., 2004; Mo et al., 2014) cited in the literature, emphasizing the need for larger, well-controlled studies with consistent methodologies .

What approaches are used to analyze the structural impact of CTSD mutations?

Structural analysis of CTSD mutations employs multiple complementary computational and experimental approaches. Molecular dynamics simulation (MDS) represents a powerful computational method for predicting structural changes induced by amino acid substitutions, as demonstrated in the analysis of the PD-associated A239V variant . This approach revealed increased flexibility in a loop adjacent to the catalytic center, potentially explaining the enhanced enzymatic activity observed with this variant. Homology modeling based on crystal structures of human CTSD provides a foundation for mapping mutations onto three-dimensional protein structures and predicting their impact on folding, stability, and substrate interactions . Experimental approaches include circular dichroism spectroscopy to assess changes in secondary structure elements and thermal shift assays to evaluate protein stability. For mutations affecting protein-protein interactions or substrate binding, surface plasmon resonance or isothermal titration calorimetry can quantify binding kinetics and affinity changes. Additionally, hydrogen-deuterium exchange mass spectrometry offers insights into conformational dynamics and solvent accessibility alterations induced by mutations. These combined approaches provide mechanistic understanding of how specific mutations affect CTSD structure and function .

What are the critical controls needed when studying CTSD variants in overexpression systems?

Rigorous experimental design for studying CTSD variants in overexpression systems requires multiple crucial controls. First, wild-type CTSD must be included as a positive control for normal maturation, localization, and enzymatic activity. The catalytically inactive D97S mutant serves as an essential negative control for enzymatic function while maintaining proper folding . For cellular studies, both CTSD-expressing and CTSD knockout backgrounds should be utilized to distinguish variant-specific effects from background activity. When analyzing disease-associated mutations, including known NCL10-associated variants with established loss of function provides important reference points for the severity spectrum . Expression level controls are particularly critical, as varying expression can confound functional assessments; therefore, expression should be quantified by immunoblotting and normalized across samples. For localization studies, co-staining with markers for different cellular compartments (lysosomes, ER, Golgi) is essential. When measuring CTSD activity, samples treated with the specific inhibitor pepstatin A establish baseline readings due to non-specific activity. For α-synuclein degradation assays, α-synuclein levels should be monitored both with and without lysosomal inhibition to confirm the lysosomal nature of the degradation process .

How should researchers interpret inconsistencies between patient fibroblast studies and overexpression models?

Interpreting discrepancies between findings in patient fibroblasts and overexpression models requires careful consideration of several methodological factors. First, expression level differences must be addressed, as overexpression systems typically produce substantially higher protein levels than endogenous expression in patient cells, potentially masking subtle functional deficits or creating artificial effects through protein aggregation or mislocalization . Genetic background variations represent another critical factor, as patient fibroblasts contain the complete patient genome including potential modifier genes that interact with CTSD variants, while overexpression models typically introduce variants into standardized cell lines lacking this genomic context . Cell type-specific effects must also be considered, as CTSD function and the impact of mutations may differ between fibroblasts and neural cells more relevant to neurodegenerative disease. Additionally, compound heterozygosity in patients (carrying different mutations on each allele) versus the study of individual mutations in overexpression models can yield different results. To reconcile these inconsistencies, researchers should employ multiple complementary approaches, including gene editing to introduce mutations at endogenous loci, testing variants in neural cell models, and conducting comprehensive phenotypic analyses beyond basic enzymatic activity measurements .

What methodological approaches help distinguish between primary CTSD defects and secondary consequences?

Distinguishing primary CTSD defects from secondary consequences requires sophisticated experimental design and analytical approaches. Time-course experiments represent a fundamental strategy, enabling researchers to establish the temporal sequence of cellular events following CTSD dysfunction. Inducible expression systems for CTSD variants allow precise control over the timing of CTSD manipulation and subsequent observation of cellular responses . Rescue experiments provide another powerful approach, where wild-type CTSD is reintroduced into CTSD-deficient or mutant-expressing cells to determine which phenotypes are directly reversible by restoring normal CTSD function . Dose-response relationships between CTSD activity levels and observed phenotypes help establish causality, with proportional relationships suggesting direct effects. Multi-omics approaches, including proteomics to identify accumulated substrates and metabolomics to detect altered metabolic pathways, provide comprehensive views of cellular changes. Single-cell analyses can identify cell-to-cell variability in responses to CTSD dysfunction, helping distinguish primary from adaptive changes. Finally, in vivo models with tissue-specific or temporally controlled CTSD manipulation allow researchers to isolate primary defects in complex organismal contexts .

How might CTSD activation be developed as a therapeutic strategy for neurodegenerative diseases?

The development of CTSD activation as a therapeutic strategy for neurodegenerative diseases represents a promising frontier, particularly given the enhanced α-synuclein degradation capacity observed with the PD-associated A239V variant . Several approaches warrant investigation. Structure-based drug design utilizing insights from molecular dynamics simulations of the A239V variant could guide the development of small molecule activators that mimic the conformational changes induced by this mutation, particularly focusing on increasing flexibility in the loop adjacent to the catalytic center . Allosteric modulation represents another promising avenue, where compounds binding to sites distant from the active center could enhance CTSD activity without directly affecting substrate binding. Gene therapy approaches delivering optimized CTSD variants with enhanced activity (such as engineered versions based on A239V) to affected brain regions could provide long-term therapeutic effects. Additionally, strategies targeting CTSD regulation through modulation of transcription, translation, or post-translational modifications might increase endogenous CTSD levels or activity. For any therapeutic development, researchers must carefully balance enhanced CTSD activity against potential off-target effects, given CTSD's role in degrading multiple substrates beyond disease-specific proteins like α-synuclein .

What are the challenges in developing cellular models that accurately reflect CTSD-related pathology?

Developing cellular models that faithfully recapitulate CTSD-related pathology faces several significant challenges. The long-term nature of neurodegenerative diseases presents a fundamental difficulty, as cellular models often cannot sustain pathological states for periods matching the years or decades of human disease progression . Cell type heterogeneity in the brain further complicates modeling efforts, as CTSD dysfunction may affect different neural populations distinctly, requiring multiple cell type-specific models. Additionally, the complex interplay between neurons and glia in disease pathogenesis necessitates co-culture systems or organoid approaches rather than single cell type models . Age-related factors significantly impact neurodegenerative disease development, yet many cellular models lack age-associated cellular changes that may influence CTSD function and substrate accumulation. Technical challenges include achieving physiologically relevant expression levels of CTSD variants, as overexpression may not accurately reflect the gradual accumulation of dysfunction in patients. To address these limitations, researchers should consider advanced approaches such as patient-derived induced pluripotent stem cell (iPSC) models, brain organoids incorporating multiple cell types, microfluidic systems enabling long-term culture, and gene editing to introduce mutations at endogenous loci rather than relying on overexpression systems .

How might systems biology approaches advance understanding of CTSD in neurodegeneration?

Systems biology approaches offer powerful frameworks for integrating multiple data types to comprehensively understand CTSD's role in neurodegeneration. Multi-omics integration combining proteomics, transcriptomics, metabolomics, and lipidomics data from CTSD-deficient or mutant models would provide unprecedented insights into the cellular pathways affected by CTSD dysfunction . Network analysis could identify key interacting partners and pathway connections that modulate CTSD function or respond to its impairment, potentially revealing novel therapeutic targets beyond CTSD itself. Mathematical modeling of CTSD maturation, trafficking, and enzymatic activity could predict how specific mutations affect these processes and how cellular systems compensate for CTSD dysfunction . Single-cell approaches would elucidate cell type-specific responses to CTSD impairment, helping explain the selective vulnerability of certain neural populations in CTSD-related diseases. Longitudinal in vivo studies coupled with systems modeling could track disease progression and identify critical transition points where therapeutic intervention might be most effective. Additionally, comparative analyses across different neurodegenerative diseases with CTSD involvement (NCL10, AD, PD) could reveal common and disease-specific mechanisms. These integrated approaches would substantially advance understanding of how CTSD dysfunction contributes to neurodegeneration and guide the development of targeted therapeutic strategies .

Product Science Overview

Structure and Function

Cathepsin-D is composed of a protein dimer of disulfide-linked heavy and light chains, both produced from a single protein precursor . The enzyme is ubiquitously distributed in lysosomes and is involved in the breakdown of various proteins, including amyloid-β protein (Aβ) and the microtubule-associated protein tau . These proteins are significant in the context of neurodegenerative diseases such as Alzheimer’s disease.

Role in Disease

Cathepsin-D has been implicated in the pathogenesis of several diseases, including breast cancer and Alzheimer’s disease . In Alzheimer’s disease, Cathepsin-D degrades both Aβ and tau, which accumulate pathognomonically in the disease . Genetic deletion of Cathepsin-D in mice has been shown to trigger large increases in cerebral Aβ and tau, leading to prominent tauopathy . This suggests that Cathepsin-D plays a major role in the proteostasis of these proteins in vivo.

Recombinant Human Cathepsin-D

Recombinant human Cathepsin-D is produced using various expression systems, such as HEK 293 cells . This recombinant form is used in research to study the enzyme’s function and its role in disease. The recombinant protein retains the same structure and function as the native enzyme, making it a valuable tool for scientific studies.

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 2024 Thebiotek. All Rights Reserved.