CTSF Human, Sf9

Cathepsin-F Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production and Purification

CTSF Human is expressed in Sf9 cells infected with recombinant baculovirus. Post-infection, the protein is purified via chromatographic techniques .

Protocol Overview:

  • Expression System: Sf9 insect cells infected with baculovirus encoding CTSF .

  • Purification: Affinity chromatography using the C-terminal His-tag .

  • Formulation: Supplied in phosphate-buffered saline (pH 7.4) with 40% glycerol .

Biological Activity

CTSF exhibits endopeptidase activity, hydrolyzing substrates like Z-Phe-Arg-AMC.

Activity Metrics:

  • Specific Activity: >5 pmol/min/µg at pH 5.0 and 37°C .

  • Functional Role: Degrades intracellular proteins, contributing to lysosomal proteolysis and antigen presentation .

Applications in Research

CTSF Human, Sf9 is utilized in:

  1. Cancer Biology: Studies on tumor progression due to CTSF overexpression in cancer cell lines .

  2. Lysosomal Function: Mechanistic insights into protein degradation and lysosomal storage disorders .

  3. Drug Development: Screening protease inhibitors targeting lysosomal pathways .

Comparison of CTSF Production Systems

ParameterSf9 Baculovirus E. coli
GlycosylationYesNo
Molecular Mass52.5 kDa26–27.4 kDa
Expression YieldHigh (post-translational)Moderate (no modifications)
ActivityNative-likeRequires refolding

Research Findings

  • Sf9 System Robustness: Widely adopted for recombinant protein production due to scalability and post-translational modification capabilities .

  • Glycosylation Impact: Sf9-derived CTSF’s glycosylation enhances stability and functional activity compared to bacterial systems .

  • Therapeutic Relevance: Sf9 systems are FDA-approved for gene therapy products, underscoring their reliability .

Product Specs

Introduction

Cathepsin F (CTSF), a member of the peptidase C1 family, is a cysteine proteinase belonging to the papain family. These proteinases constitute a significant part of the lysosomal proteolytic system. CTSF exhibits ubiquitous expression, and its gene is located on chromosome 11q13, in proximity to the gene encoding cathepsin W. This proteinase participates in the regular breakdown of proteins. Moreover, CTSF is implicated in certain degradative processes associated with tumor progression, as its expression is elevated in specific cancer cell lines.

Description

Produced in Sf9 Baculovirus cells, CTSF is a single, glycosylated polypeptide chain with a molecular weight of 52.5kDa. It comprises 474 amino acids (20-484.a.a). The protein is expressed with a 6 amino acid His tag at the C-Terminus and purified using proprietary chromatographic methods.

Physical Appearance

Sterile filtered, colorless solution.

Formulation

The CTSF protein solution (0.25mg/ml) is supplied in Phosphate buffered saline (pH7.4) with 40% glycerol.

Stability

For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the protein frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. It is crucial to avoid repeated cycles of freezing and thawing.

Purity

Purity exceeds 90.0% as assessed by SDS-PAGE.

Biological Activity

The specific activity, defined as the amount of enzyme required to hydrolyze 1 picomole of Z-Phe-Arg-AMC to Z-Phe-Arg and AMC per minute at a pH of 5.0 and a temperature of 37°C, is greater than 5 pmol/min/ug.

Synonyms

CTSF, CATSF, CLN13.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADLAPAQPRA ASFQAWGPPS PELLAPTRFA LEMFNRGRAA GTRAVLGLVR GRVRRAGQGS LYSLEATLEE PPCNDPMVCR LPVSKKTLLC SFQVLDELGR HVLLRKDCGP VDTKVPGAGE PKSAFTQGSA MISSLSQNHP DNRNETFSSV ISLLNEDPLS QDLPVKMASI FKNFVITYNR TYESKEEARW RLSVFVNNMV RAQKIQALDR GTAQYGVTKF SDLTEEEFRT IYLNTLLRKE PGNKMKQAKS VGDLAPPEWD WRSKGAVTKV KDQGMCGSCW AFSVTGNVEG WFLNQGTLL SLSEQELLDC DKMDKACMGG LPSNAYSAIK NLGGLETEDD YSYQGHMQSC NFSAEKAKVY INDSVELSQN EQKLAAWLAK RGPISVAINA FGMQFYRHGI SRPLRPLCSP WLIDHAVLLV GYGNRSDVPF WAIKNSWGTD WGEKGYYYLH RGSGACGVNT MASSAVVDHH HHHH

Q&A

What is the functional significance of CTSF protein in human cellular processes?

Cathepsin F (CTSF) is a lysosomal cysteine protease belonging to the peptidase C1 family that plays essential roles in intracellular protein degradation and turnover. This 484-amino acid protein contains two key functional domains: a peptidase inhibitor I29 domain and a peptidase C1 domain, which work together to regulate its catalytic activity. CTSF has particularly important functions in neuronal health, as homozygous mutations in the CTSF gene cause adult-onset neuronal ceroid lipofuscinosis (ANCL or Kufs disease), a progressive neurodegenerative lysosomal storage disorder. The protein's enzymatic activity contributes to normal lysosomal function, and disruption of this activity leads to accumulation of storage material and subsequent cellular dysfunction. Recent evidence suggests it may also have roles in frontotemporal dementia when specific mutations are present in the heterozygous state .

Why is Sf9 insect cell expression preferred for recombinant CTSF production?

Sf9 insect cells represent an optimal compromise between proper protein folding and reasonable yield when expressing complex mammalian proteins like CTSF. Unlike bacterial expression systems that often produce misfolded proteins requiring refolding procedures, Sf9 cells provide eukaryotic post-translational modifications essential for proper CTSF folding and activity. While mammalian cell systems (such as HEK293) offer the most authentic human-like modifications, they typically have lower yields and higher costs. Sf9 cells also grow at room temperature, which can reduce protein aggregation problems common with complex proteases. Additionally, the baculovirus expression system used with Sf9 cells enables high-level expression of proteins that might be toxic when expressed in mammalian systems, making it particularly valuable for enzymes like CTSF that have proteolytic activity .

How do CTSF mutations impact lysosomal function in neurodegenerative disorders?

CTSF mutations disrupt normal lysosomal function through multiple mechanisms, ultimately leading to neurodegenerative phenotypes. Homozygous mutations in CTSF, such as p.Ile404Thr found in a Belgian family with ANCL, impair enzymatic function by affecting the peptidase C1 domain. This disruption leads to incomplete protein degradation and accumulation of storage material within lysosomes, visible as autofluorescent lipopigments on microscopic examination. The progressive accumulation of this material causes lysosomal dysfunction, impaired autophagy, and eventual neuronal death. Interestingly, even heterozygous mutations like p.Arg245His may contribute to frontotemporal dementia with a progressive supranuclear palsy phenotype, suggesting dosage-sensitive roles for CTSF in certain neuronal populations. The remarkable phenotypic variability seen even within families carrying identical mutations indicates complex interactions with genetic modifiers and environmental factors. The close connection between CTSF dysfunction and frontotemporal presentations highlights the importance of lysosomal health in maintaining neuronal integrity, particularly in frontal brain regions .

How can researchers design experiments to distinguish between pathogenic and benign CTSF variants?

Designing experiments to distinguish between pathogenic and benign CTSF variants requires a multimodal approach combining biochemical, cellular, and computational methods. First, researchers should conduct comparative enzymatic activity assays using purified recombinant wild-type and variant CTSF proteins expressed in Sf9 cells. This requires measuring full enzyme kinetics (Km, Vmax, kcat/Km) with multiple substrates under varying pH conditions (pH 4.0-7.0) to detect subtle changes in catalytic efficiency or pH optima. Second, researchers should develop cellular models expressing these variants, preferably in neuronal cells or patient-derived iPSCs differentiated into neurons, to assess lysosomal morphology, autophagy flux (LC3-II/I ratios), and accumulation of storage material. Third, structural analysis using circular dichroism or, ideally, X-ray crystallography can reveal how variants affect protein folding and substrate binding. Fourth, comparative protein stability assays (thermal shift assays, limited proteolysis) can identify variants that primarily affect protein stability rather than catalytic function. Finally, researchers should validate findings by examining heterozygous carriers of variants to determine if there are subclinical phenotypes that might indicate partial loss of function. This comprehensive approach helps differentiate truly pathogenic variants from benign polymorphisms or variants of uncertain significance .

How should researchers resolve contradictory findings between in vitro CTSF activity and in vivo phenotypes?

Resolving contradictions between in vitro CTSF activity and in vivo phenotypes requires systematic investigation of multiple factors that may explain the discrepancy. First, researchers should evaluate whether the in vitro conditions accurately reflect the cellular environment where CTSF functions. This includes testing activity across a range of pH values (4.0-7.0), with various co-factors, and in the presence of potential regulatory proteins. Second, researchers should consider that some mutations might primarily affect protein stability, trafficking, or half-life rather than catalytic activity. Time-course studies in cellular systems can reveal degradation rates and accumulation patterns. Third, compensatory mechanisms may mask phenotypes in vivo; therefore, researchers should examine expression of other cathepsins and lysosomal enzymes that might functionally compensate for CTSF deficiency. Fourth, tissue-specific factors might modulate CTSF function, explaining why some mutations predominantly affect neuronal tissues despite CTSF's broader expression pattern. Fifth, researchers should investigate whether CTSF has non-enzymatic roles (scaffold functions, protein-protein interactions) that contribute to phenotypes independent of catalytic activity. Finally, patient-derived cellular models (fibroblasts, iPSC-derived neurons) provide valuable systems to bridge in vitro and in vivo findings by allowing manipulation of CTSF in disease-relevant genetic backgrounds .

What are the optimal conditions for measuring CTSF enzymatic activity in research applications?

Optimal conditions for measuring CTSF enzymatic activity require careful attention to buffer composition, pH, temperature, and substrate selection. The recommended buffer system is 100 mM sodium acetate with 1-5 mM DTT or 2-10 mM β-mercaptoethanol (essential for maintaining the catalytic cysteine in reduced form) and 1 mM EDTA to inhibit metalloproteases. The optimal pH range is 4.5-5.5, reflecting the lysosomal environment, but researchers should perform a pH profile (pH 4.0-7.0) to identify potential shifts in pH optima for mutant proteins. Temperature should be maintained at 37°C for physiological relevance. Pre-activation by incubating the enzyme at acidic pH (4.0-4.5) for 10-15 minutes before substrate addition is critical for full activity. Z-Phe-Arg-AMC is the most commonly used fluorogenic substrate, but researchers should consider multiple substrates to detect changes in substrate specificity. Enzyme concentration should be optimized to ensure linear reaction kinetics throughout the measurement period (typically 0.5-5 nM final concentration). Reactions should be monitored continuously if possible, or at multiple time points, to ensure linearity. Controls should include specific inhibitors (E-64, leupeptin) and measurements with related cathepsins to confirm specificity. For accurate kinetic parameter determination, substrate concentrations should span at least 0.2-5× Km .

How can researchers effectively distinguish between direct effects of CTSF mutations and secondary cellular adaptations?

Distinguishing direct effects of CTSF mutations from secondary cellular adaptations requires temporal and mechanistic approaches. First, researchers should develop inducible expression systems for both wild-type and mutant CTSF, allowing time-course analysis from the initial expression through long-term adaptation. This permits identification of primary effects (occurring within hours of expression) versus secondary adaptations (developing over days to weeks). Second, researchers should implement parallel approaches comparing acute versus chronic models: acute knockdown/knockout with immediate rescue versus stable cell lines with constitutive alteration of CTSF function. Third, pathway analysis using transcriptomics or proteomics at multiple time points can reveal the sequence of cellular changes following CTSF dysfunction. Fourth, pharmacological separation of effects can be achieved using specific inhibitors that target suspected secondary pathways to determine if blocking these pathways prevents downstream consequences of CTSF mutation. Fifth, researchers should conduct comparative studies across multiple cell types with different baseline expression levels of compensatory pathways to identify cell type-specific versus universal effects of CTSF mutations. Finally, cross-rescue experiments, where defects caused by CTSF mutation are rescued by manipulating other proteins or pathways, can distinguish direct effects from adaptive responses by revealing mechanistic relationships. This systematic approach allows researchers to construct causality chains from primary CTSF dysfunction to ultimate cellular phenotypes .

What cellular models are most appropriate for studying CTSF-related neurological disorders?

Selecting appropriate cellular models for studying CTSF-related neurological disorders requires balancing disease relevance, technical feasibility, and research questions. Patient-derived fibroblasts provide a valuable starting point, maintaining the disease-causing mutations in their genomic context, but may not exhibit pronounced neurological phenotypes. iPSC technology allows reprogramming of patient cells and differentiation into neurons or mixed neural cultures, providing the most disease-relevant human cellular model with the authentic genetic background. These can be matured into 3D cerebral organoids to study developmental aspects and cell-cell interactions. For precise genetic manipulation, CRISPR-edited neuronal cell lines (SH-SY5Y, ReNcell) offer controlled genetic backgrounds where specific CTSF mutations can be introduced with appropriate isogenic controls. Primary neurons from animal models carrying equivalent mutations provide authentic neuronal properties for electrophysiological and morphological studies. For high-throughput screening, neuroblastoma cell lines overexpressing wild-type or mutant CTSF are more practical. Most informative studies employ multiple complementary models, for example, combining iPSC-derived neurons for disease relevance with CRISPR-edited lines for mechanistic studies. Researchers should select models based on whether they are studying cell-autonomous effects, which can be addressed in simpler systems, or complex neuron-glia interactions, which require mixed cultures or organoids .

What experimental controls are essential when overexpressing CTSF in cellular systems?

Designing rigorous controls for CTSF overexpression experiments is essential for valid interpretation of results. First, empty vector controls are mandatory to account for transfection/transduction effects, with matched vector backbones and selection markers. Second, catalytically inactive CTSF mutants (typically active site cysteine to alanine substitution) distinguish between effects requiring enzymatic activity versus protein presence alone. Third, expression level controls are critical—researchers should use western blotting to confirm comparable expression levels between wild-type and mutant constructs, and consider fluorescent tags or inducible expression systems to monitor expression. Fourth, subcellular localization controls should include versions of CTSF with altered targeting signals to determine if effects depend on lysosomal localization versus protein expression regardless of location. Fifth, related cathepsin overexpression (cathepsin L or B) helps determine specificity of observed effects to CTSF. Sixth, time-course sampling distinguishes immediate effects from adaptive responses. Seventh, rescue experiments in knockout or knockdown backgrounds comparing wild-type versus mutant CTSF confirm specificity and test functionality of mutants. Finally, dosage controls using titration of expression levels help identify potential threshold effects and distinguish toxic overexpression artifacts from physiologically relevant phenotypes. These comprehensive controls ensure that observed phenotypes are specifically attributable to CTSF function rather than experimental artifacts .

Known CTSF Mutations and Associated Clinical Phenotypes

MutationProtein DomainDisease AssociationInheritance PatternClinical ManifestationsAge of Onset
p.Ile404ThrPeptidase C1ANCL/Kufs DiseaseAutosomal recessiveProgressive behavioral abnormalities, dementia, motor dysfunction26-50 years
p.Arg245HisNear Inhibitor I29Frontotemporal dementiaHeterozygousProgressive supranuclear palsy phenotype, behavioral changes65-76 years
Other CTSF mutationsVariousANCL/Kufs Disease Type BAutosomal recessiveProgressive behavioral abnormalities, dementiaVariable

The p.Ile404Thr mutation in CTSF was identified in a Belgian family with autosomal recessive adult-onset neuronal ceroid lipofuscinosis, with remarkably variable onset ages ranging from 26 to 50 years despite identical mutations. One patient presented with myoclonic epilepsy characteristic of Kufs Disease Type A, while others showed typical Type B presentations, challenging the conventional clinical classification. The p.Arg245His mutation was found in heterozygous state in two unrelated patients with frontotemporal dementia presenting with progressive supranuclear palsy phenotypes, suggesting potential roles for CTSF in broader neurodegenerative contexts beyond classic lysosomal storage disorders .

Optimal Expression Conditions for CTSF in Sf9 System

ParameterOptimal ConditionTechnical Considerations
VectorpFastBac with C-terminal His-tagAllows secretion and simplified purification
Cell Density2 × 10^6 cells/ml at infectionDensity critical for protein quality
MOI (Multiplicity of Infection)1-3Higher MOI may reduce protein quality
Expression Temperature27°CLower temperature improves folding
Expression Duration72-96 hoursLonger times increase yield but may reduce quality
Harvest TimingMonitor viability; harvest at ~70-80% viabilityBalance between yield and quality
Lysis BufferpH 7.4 phosphate buffer with protease inhibitorsPrevents self-digestion during purification
PurificationIMAC followed by size exclusion chromatographyTwo-step purification enhances purity

Expressing CTSF in Sf9 cells requires careful optimization of infection parameters and harvest timing to balance protein yield with quality. The addition of protease inhibitors during all purification steps is essential to prevent self-digestion. Low-temperature expression (27°C) significantly improves the proportion of correctly folded protein compared to standard 28-30°C conditions. For functional studies, the purified protein requires activation at acidic pH before activity measurement .

Troubleshooting Guide for CTSF Activity Assays

IssuePossible CausesSolutions
Low/No ActivityIncomplete activationPre-incubate at pH 4.0-4.5 for 15 minutes
Oxidized active siteIncrease DTT concentration to 5-10 mM
Protein aggregationOptimize purification; add glycerol to storage buffer
Improper pHTest activity across pH 4.0-7.0
High BackgroundContaminating proteasesAdd selective inhibitors; improve purification
Auto-hydrolysis of substrateReduce incubation time; lower temperature
Inconsistent ResultsTemperature fluctuationsMaintain strict temperature control
Freeze-thaw cyclesAliquot enzyme; avoid repeated freeze-thaw
Buffer composition variationsPrepare fresh buffers; standardize protocol

Successful CTSF activity measurement requires attention to protein quality, assay conditions, and proper controls. The enzyme is particularly sensitive to oxidation of its catalytic cysteine, requiring reducing conditions throughout purification and storage. Activity is highly dependent on pH, with optimal activity in the acidic range mimicking lysosomal conditions. When comparing wild-type and mutant proteins, matching purification protocols and assay conditions is essential for valid comparisons .

Cellular Phenotypes Associated with CTSF Dysfunction

PhenotypeDetection MethodSignificance in CTSF Disorders
Lysosomal enlargementLysoTracker staining, LAMP1 immunofluorescencePrimary indicator of lysosomal storage
Autofluorescent storage materialConfocal microscopy (ex: 405 nm, em: 500-550 nm)Characteristic of neuronal ceroid lipofuscinosis
Impaired autophagyLC3-II/I ratio, p62 accumulationSecondary consequence of lysosomal dysfunction
Mitochondrial dysfunctionMitoTracker, Seahorse analysisTertiary effect leading to energy deficits
Astrogliosis and microglial activationGFAP and Iba1 staining (in vivo models)Neuroinflammatory response to neuronal dysfunction
Neurite abnormalitiesMAP2/Tau staining, neurite tracingMay precede neurodegeneration

CTSF dysfunction leads to a cascade of cellular abnormalities beginning with primary lysosomal defects and progressing to impaired autophagy, mitochondrial dysfunction, and ultimately neuronal death. The accumulation of autofluorescent storage material (lipofuscin) is a hallmark finding, particularly relevant for diagnosis. Advanced imaging techniques combined with biochemical assays can track disease progression and response to potential therapeutics in cellular models. The temporal sequence of these phenotypes provides insight into disease mechanisms and potential intervention points .

What therapeutic strategies show promise for CTSF-related neurological disorders?

Current research identifies several promising therapeutic approaches for CTSF-related disorders, though each presents unique challenges. Enzyme replacement therapy, delivering recombinant CTSF protein to affected tissues, faces blood-brain barrier penetration challenges that may require specialized delivery strategies such as engineered antibodies or nanoparticles. Gene therapy approaches using AAV vectors show promise in preclinical models of lysosomal storage disorders, though optimal serotypes for targeting affected neuronal populations must be determined. Small molecule chaperones that improve folding and stability of mutant CTSF proteins represent a particularly promising approach for missense mutations that affect protein stability rather than catalytic function. These could be identified through high-throughput screening using thermal shift assays with recombinant proteins. Substrate reduction therapy, targeting biosynthetic pathways for accumulated substrates, presents another option, though specific CTSF substrates require better characterization. Regardless of approach, early intervention will likely be crucial, as neurodegeneration may become irreversible once advanced. Combined therapeutic strategies may ultimately prove most effective, perhaps using gene therapy for long-term correction while employing enzyme replacement or small molecules for immediate effect .

How can multi-omics approaches advance our understanding of CTSF function in health and disease?

Multi-omics approaches provide powerful tools to comprehensively understand CTSF function and dysfunction. Proteomics can identify the natural substrates of CTSF through comparative analysis of wild-type versus CTSF-deficient cells, while degradomics specifically profiles proteolytic events altered by CTSF dysfunction. Lipidomics reveals changes in membrane composition resulting from altered lysosomal function, potentially identifying biomarkers for disease progression. Transcriptomics identifies compensatory mechanisms and stress responses activated by CTSF deficiency, helping explain phenotypic variability between patients with identical mutations. Metabolomics can detect alterations in cellular energetics and small molecule metabolism secondary to lysosomal dysfunction. Interactomics using proximity labeling approaches can map the CTSF protein interaction network, revealing potential regulatory mechanisms and additional therapeutic targets. Integration of these multi-omics datasets through systems biology approaches can construct comprehensive models of how CTSF dysfunction propagates through cellular networks to cause disease. Most importantly, applying these approaches to patient-derived cells and comparing different mutations can explain the remarkable clinical heterogeneity observed even within families carrying identical CTSF mutations .

Product Science Overview

Structure and Expression

Cathepsin-F is produced as a single, glycosylated polypeptide chain containing 474 amino acids, with a molecular mass of approximately 52.5 kDa . The recombinant form of Cathepsin-F is often expressed in Sf9 Baculovirus cells, which are derived from the fall armyworm, Spodoptera frugiperda . This recombinant protein includes a 6-amino acid His tag at the C-terminus, which facilitates purification using chromatographic techniques .

Function and Activity

Cathepsin-F plays a role in normal protein catabolism and is involved in various degradative processes, including those occurring during tumor progression . It is highly expressed in some cancer cell lines, indicating its potential role in cancer biology . The enzyme exhibits specific activity, defined as the amount of enzyme that hydrolyzes 1 pmole of Z-Phe-Arg-AMC to Z-Phe-Arg and AMC per minute at pH 5.0 at 37°C .

Stability and Storage

The recombinant Cathepsin-F protein is typically formulated in a phosphate-buffered saline solution (pH 7.4) with 40% glycerol . For short-term storage, it can be kept at 4°C, while for long-term storage, it should be frozen at -20°C. To maintain its stability, it is recommended to add a carrier protein, such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA), and to avoid multiple freeze-thaw cycles .

Biological Significance

Cathepsin-F is ubiquitously expressed in various tissues, including the heart, skeletal muscle, brain, testis, and ovary . It has moderate expression levels in the prostate, placenta, liver, and colon, but no detectable expression in peripheral leukocytes and thymus . The enzyme’s activity is optimal at a pH range of 5.2 to 6.8, and it is relatively unstable at cytosolic pH (7.2), with a half-life of approximately 2 minutes . This suggests that Cathepsin-F functions primarily in acidic cellular compartments, such as lysosomes .

Research and Applications

Recombinant Cathepsin-F produced in Sf9 cells is used extensively in laboratory research to study its role in protein degradation and its involvement in diseases such as cancer . The enzyme’s activity and stability make it a valuable tool for investigating the mechanisms of lysosomal proteolysis and the development of potential therapeutic interventions targeting cathepsin activity.

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.