CASP2 Human

Caspase 2 Apoptosis-Related Cysteine Peptidase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

CASP2 is synthesized as an inactive zymogen (pro-caspase-2) containing:

  • CARD domain: Facilitates protein interactions (e.g., with RAIDD, PIDD, and DEFCAP) .

  • Proteolytic subunits: Processed into p19 (large) and p12 (small) subunits during activation. The active enzyme forms a dimer of p18:p12 heterodimers .

  • Isoforms: Alternative splicing generates variants with substitutions in the prodomain or catalytic regions, including isoforms lacking nuclear localization signals .

Biological Functions and Mechanisms

CASP2 operates through two primary pathways:

  • Apoptosis: Activated in the PIDDosome complex (with PIDD1 and CRADD) to cleave substrates like BID and ICAD, triggering mitochondrial apoptosis .

  • Proteostasis: Acts as a deubiquitinase under proteotoxic stress, reducing ubiquitinated aggregates (e.g., TDP-43 and TBK1) to maintain cellular homeostasis .

Key findings include:

  • Ubiquitin regulation: CASP2 deficiency increases ubiquitinated proteins and accelerates pathological aggregation in ALS models .

  • Non-apoptotic roles: Modulates synaptic plasticity and dendritic spine stability, with knockout mice showing anxiety-like behaviors .

Neurodegenerative Disorders

  • Amyotrophic lateral sclerosis (ALS): CASP2-deficient mice exhibit motor defects, neuromuscular denervation, and TDP-43 aggregation, mimicking ALS pathology .

  • Alzheimer’s disease: Elevated CASP2 correlates with amyloid-β toxicity, suggesting a role in neuronal death .

Neurodevelopmental Disorders

  • Lissencephaly: Biallelic truncating CASP2 variants cause fronto-temporal pachygyria and developmental delay, similar to CRADD and PIDD1 mutations .

Metabolic Diseases

  • Nonalcoholic steatohepatitis (NASH): Casp2 inhibitors (e.g., LJ2a) show therapeutic potential by reducing hepatocyte apoptosis .

Table 2: Recombinant CASP2 Proteins

SupplierExpression HostTagPurityApplications
OriGene E. coliN-terminal His>80%Enzyme assays, antibody production
R&D Systems E. coliNone>95%Kinetic studies, drug screening

Caspase-2 Inhibitors

  • LJ2a: Irreversible inhibitor with 5,500,000 M⁻¹s⁻¹ selectivity for CASP2 over Caspase-3 .

  • LJ3b: Demonstrates 765-fold lower efficacy compared to LJ2a, highlighting stereochemical sensitivity .

Clinical Implications and Future Directions

  • Therapeutic targeting: CASP2 activation may alleviate protein aggregation in ALS, while inhibition could mitigate apoptosis in NASH .

  • Biomarker potential: Ubiquitination patterns in CASP2-deficient models may inform diagnostics for neurodegenerative diseases .

Product Specs

Introduction
Caspase 2, also known as Caspase 2 Apoptosis-Related Cysteine Peptidase (CASP2), is a protein belonging to the caspase family. These enzymes are crucial in the process of apoptosis, which is a form of programmed cell death. CASP2 is involved in the activation cascade of caspases, a series of events that ultimately lead to the dismantling of a cell. It also plays a role in inactivating proteins that are essential for a cell's survival. A variety of signals that trigger apoptosis can induce the breakdown of CASP2, indicating its involvement in the cell death pathway.
Description
Recombinant human CASP2, expressed in E.coli, is available as a single, non-glycosylated polypeptide chain. It consists of 126 amino acids (residues 348-452) and has a molecular weight of 14.1 kDa. The protein is engineered with a 21 amino acid His-tag at the N-terminus to facilitate purification, which is achieved through proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The provided CASP2 solution has a concentration of 1mg/ml and is formulated in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), store the CASP2 vial at 4°C. For longer storage, it is recommended to freeze the solution at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advised. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of CASP2 is determined to be greater than 80% using SDS-PAGE analysis.
Synonyms
Caspase-2, CASP-2, CASP2, Caspase 2 Apoptosis-Related Cysteine Peptidase, Neural precursor cell expressed developmentally down-regulated protein 2, NEDD-2, Protease ICH-1, ICH1, NEDD2, Caspase-2 subunit p18, Caspase-2 subunit p13, Caspase-2 subunit p12, Caspase 2 isoform 1, PPP1R57.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAGKEKLPKM RLPTRSDMIC GYACLKGTAA MRNTKRGSWY IEALAQVFSE RACDMHVADM LVKVNALIKD REGYAPGTEF HRCKEMSEYC STLCRHLYLF PGHPPT.

Q&A

What is the primary function of CASP2 in human cells?

CASP2 is a highly conserved cysteine-aspartyl protease traditionally associated with apoptosis but now recognized as having multiple non-apoptotic functions. It plays critical roles in regulating cell cycle events, DNA damage repair, protection against ferroptosis, protein quality control through deubiquitination activity, and neurodevelopment. These functions are frequently independent of its canonical role in programmed cell death . CASP2 operates within a protein complex called the PIDDosome, which includes PIDD (p53-induced protein with a death domain) and RAIDD (RIP-associated ICH-1 homologous protein with a death domain), serving as the activation platform for CASP2 .

How does the PIDDosome complex regulate CASP2 activation?

The PIDDosome complex forms the primary activation platform for CASP2 and consists of three key components: CASP2, PIDD, and RAIDD. PIDD is a p53 target gene, creating a functional link between p53 signaling and CASP2 activation . When investigating PIDDosome assembly, researchers should use co-immunoprecipitation assays to detect protein-protein interactions between these components. Experimental approaches should include both overexpression systems and endogenous detection following cellular stress stimuli, particularly DNA damage inducers. Western blotting using specific antibodies against CASP2 (such as clone 11B4), PIDD1, and RAIDD allows for verification of complex formation . Genetic approaches using CRISPR/Cas9 to delete individual PIDDosome components have revealed that disruption of this complex in certain contexts (such as NPM1c+ acute myeloid leukemia cells) significantly impairs cellular proliferation and survival .

How does CASP2 influence cell cycle progression?

CASP2 functions as a cell cycle regulator, primarily during G1 and S phases. In response to replication stress, CASP2 protects stalled replication forks from collapsing and prevents subsequent DNA damage accumulation. Methodologically, researchers can analyze this function by using flow cytometry with BrdU incorporation and propidium iodide staining to track cell cycle progression. CASP2-knockout cells display accelerated proliferation rates and impaired cell cycle arrest following DNA damage . To experimentally demonstrate CASP2's cell cycle regulatory function, researchers should compare wild-type and CASP2-deficient cells using pulse-chase experiments with thymidine analogs to assess S-phase progression and exit timing. Chromosomal aberration assays reveal that CASP2 loss leads to S-phase-specific chromosomal abnormalities that can be visualized through metaphase spread preparation and analysis .

What role does CASP2 play in the DNA damage response?

CASP2 protects cells from DNA damage accumulation, particularly during replication stress. When replication forks stall due to topoisomerase inhibition or other stressors, CASP2 prevents the conversion of single-strand DNA regions to double-strand breaks . For investigating this function, researchers should employ the following methodology: (1) Comet assays to quantify DNA damage in wild-type versus CASP2-knockout cells following treatment with replication stress inducers; (2) Immunofluorescence for γH2AX foci formation to assess double-strand break accumulation; (3) Western blotting for phosphorylated ATM (Ser1981) using specific antibodies (clone 10H11) to measure DNA damage response activation . These approaches reveal that CASP2 deficiency leads to increased DNA damage accumulation and delayed exit from S-phase following replication stress.

How does CASP2 protect cells against ferroptosis?

CASP2 provides cellular protection against ferroptotic cell death, a form of regulated cell death characterized by iron-dependent lipid peroxidation. Methodologically, this protective function can be investigated using ferroptosis inducers such as erastin and RSL3 in both wild-type and CASP2-deficient cells. CRISPR/Cas9-generated CASP2 knockout cells demonstrate significantly increased susceptibility to ferroptosis compared to control cells . To quantify this effect, researchers should measure cell viability using MTT/MTS assays, assess colony formation in long-term clonogenic assays, and detect lipid peroxidation using C11-BODIPY dye. Following erastin treatment, CASP2-knockout cells form significantly fewer colonies than control cells, indicating compromised long-term survival . This protective role represents a novel function of CASP2 beyond its canonical role in apoptosis.

What is the role of CASP2 in protein quality control during cellular stress?

CASP2 functions as a condensate-mediated deubiquitinase (DUB) in protein quality control systems during cellular stress. Under stress conditions such as heat shock, oxidative stress, or osmotic stress, CASP2 accumulates in biomolecular condensates termed "ubstressomes," which contain ubiquitinated proteins and proteasomes . Within these condensates, CASP2 removes excessive ubiquitin chains from misfolded proteins through its protease activity. Methodologically, this function can be investigated using fluorescence microscopy to visualize CASP2 localization to stress-induced condensates, and through biochemical assays measuring deubiquitinating activity. CASP2 binds to poly-ubiquitinated conjugates via its allosteric ubiquitin-interacting motif-like region and reduces overloaded ubiquitin chains to promote substrate degradation . This represents a novel non-apoptotic function of CASP2 that is critical for maintaining cellular ubiquitin homeostasis.

What are the most effective approaches for developing CASP2-selective inhibitors?

Developing CASP2-selective inhibitors presents significant challenges due to the high sequence and structural homology among the twelve human caspases. Recent advances have employed the following methodological approaches:

  • Structure-based design of peptidomimetics derived from the VDVAD pentapeptide structure with non-natural modifications at the P2 position and an irreversible warhead .

  • Mass spectrometry-based chemoproteomics to identify highly reactive non-catalytic cysteines unique to CASP2 .

  • Gel-based activity-based protein profiling (ABPP) to assess labeling specificity, comparing general cysteine reactivity probes (IAA) with caspase-directed probes (Rho-DEVD-AOMK) .

Compounds such as LJ2, LJ2a, and LJ3a have demonstrated strong and irreversible inhibition of CASP2 with genuine selectivity. LJ2a inhibits human CASP2 with an extraordinarily high inactivation rate (k3/Ki ~5,500,000 M-1s-1), while LJ3a shows approximately 1000-fold higher selectivity for CASP2 compared to CASP3 . Structural analysis reveals that the spatial configuration of Cα at the P2 position is a critical determinant of inhibitor efficacy. These inhibitors have proven effective in preventing CASP2-mediated cleavage of site-1 protease (S1P) and sterol regulatory element-binding protein 2 (SREBP2) activation, suggesting therapeutic potential for nonalcoholic steatohepatitis (NASH) .

How can isoTOP-ABPP be applied to identify functional cysteine residues in CASP2?

Isotopic tandem orthogonal proteolysis activity-based protein profiling (isoTOP-ABPP) represents an advanced approach for identifying functional and ligandable cysteine residues in CASP2. The methodology involves:

  • Treatment of cell lysates (e.g., from Jurkat cells expressing elevated caspase levels) with different concentrations of iodoacetamide (IAA) (10μM vs. 100μM) .

  • Coupling the isoTOP-ABPP workflow with single-pot solid-phase-enhanced sample-preparation (SP3) and high field asymmetric ion mobility spectrometry (FAIMS) for high coverage reactivity datasets .

  • Analysis of cysteine residue functionality based on IAA-reactivity profiles.

This approach has successfully identified C370 as a highly reactive non-catalytic cysteine unique to CASP2, providing a target for selective inhibitor development. To validate findings, researchers should express recombinant pro-CASP2 harboring mutations (such as D333A and D347A to prevent activation) along with single and dual C370A and C320A mutations to assess cysteine specificity. Gel-based ABPP comparing IAA labeling with caspase-directed Rho-DEVD-AOMK probes allows for assessment of general cysteine reactivity versus caspase activity .

What are the optimal experimental conditions for assessing CASP2's deubiquitinating activity?

To effectively assess CASP2's deubiquitinating (DUB) activity, researchers should employ the following methodological approach:

  • Induce cellular stress using heat shock, oxidative stress agents, or proteasome inhibitors to promote CASP2 condensate formation.

  • Utilize advanced microscopy techniques including live-cell imaging and fluorescence recovery after photobleaching (FRAP) to visualize and characterize the dynamic "ubstressome" condensates containing CASP2, ubiquitinated proteins, and proteasomes .

  • Perform in vitro deubiquitination assays using purified CASP2 and poly-ubiquitinated substrates, followed by Western blot analysis to detect ubiquitin chain removal.

  • Compare wild-type CASP2 with catalytically inactive mutants to confirm that the deubiquitinating activity depends on CASP2's protease activity .

  • Use proximity ligation assays to confirm CASP2 interaction with ubiquitinated substrates via its allosteric ubiquitin-interacting motif-like region.

This approach will establish whether CASP2 directly removes ubiquitin chains from specific substrates, as demonstrated in studies showing that CASP2 deficiency in mice results in excessive accumulation of poly-ubiquitinated TAR DNA-binding protein 43, leading to motor defects .

How does CASP2 function in cancer cells with different genetic backgrounds?

CASP2 demonstrates context-dependent functions in cancer cells based on their genetic background. In acute myeloid leukemia (AML) cells, CASP2's role differs dramatically between NPM1 wild-type (NPM1wt) and NPM1 mutant (NPM1c+) cells. Research methodology to investigate these differences should include:

  • CRISPR/Cas9-mediated knockout of CASP2 in cell lines with different genetic backgrounds (e.g., comparing THP-1 cells (NPM1wt) with IMS-M2 cells (NPM1c+)) .

  • Proliferation assays, including growth curves and colony formation assays, to assess the impact of CASP2 deletion on cellular proliferation.

  • Cell viability measurements following CASP2 knockout compared to scramble controls.

These approaches have revealed that while CASP2 deletion increases proliferation in NPM1wt cells, it dramatically impairs growth and viability in NPM1c+ cells . This indicates that NPM1c+ cells have developed a dependency on CASP2 for survival and proliferation. Similar context-dependent effects are observed with RAIDD deletion, suggesting that the entire PIDDosome complex is essential for NPM1c+ cell viability. These findings highlight the importance of genetic background in determining CASP2 function and suggest potential therapeutic approaches targeting CASP2 in specific cancer subtypes.

What is the evidence for CASP2's role in neurodevelopmental disorders?

Recent genetic evidence strongly implicates CASP2 in neurodevelopmental disorders, particularly lissencephaly (LIS), a malformation of cortical development characterized by deficient neuronal migration and abnormal formation of cerebral convolutions. The methodological approach for establishing this connection includes:

  • Exome sequencing analysis of patients with neurodevelopmental phenotypes, revealing biallelic truncating variants in CASP2 (homozygous c.1156delT, c.1174 C>T, and compound heterozygous c.[130 C>T];[876+1 G>T]) .

  • RNA studies of splice site variants (like c.876+1 G>T) to demonstrate the usage of cryptic splice donor sites introducing premature stop codons .

  • Brain MRI analysis showing typical fronto-temporal LIS and pachygyria in patients with CASP2 variants, similar to findings in patients with CRADD and PIDD1-related disorders .

  • Clinical phenotyping revealing developmental delay, attention deficit hyperactivity disorder, hypotonia, seizures, poor social skills, and autistic traits in affected individuals .

These findings expand the genetic spectrum of LIS and support the critical role of each component of the PIDDosome complex (CASP2, CRADD, PIDD1) in normal development of the human cerebral cortex and brain function. For researchers studying CASP2 in neurodevelopment, animal models with these specific variants would be valuable tools for understanding the underlying mechanisms.

How can CASP2 inhibitors be evaluated for therapeutic potential in NASH and Alzheimer's disease?

CASP2 has emerged as a promising therapeutic target for nonalcoholic steatohepatitis (NASH) and Alzheimer's disease (AD). To evaluate CASP2 inhibitors for these conditions, researchers should employ the following methodology:

For NASH applications:

  • Transfect human cell lines to overexpress site-1 protease (S1P), sterol regulatory element-binding protein 2 (SREBP2), and CASP2 .

  • Treat with CASP2 inhibitors (such as LJ2a or LJ3a) and assess CASP2-mediated S1P cleavage and subsequent SREBP2 activation through Western blotting .

  • Perform dose-response studies to determine the minimum inhibitor concentration required to fully prevent CASP2-mediated S1P cleavage.

  • Validate findings in liver cell models and animal models of NASH, assessing lipid accumulation and liver damage markers.

For Alzheimer's disease applications:

  • Culture primary hippocampal neurons and treat with β-amyloid oligomers to induce synapse loss .

  • Apply CASP2 inhibitors at varying concentrations and assess their ability to prevent synapse loss through immunofluorescence for synaptic markers.

  • Quantify synapse density and analyze dose-dependent protection.

  • Evaluate cognitive outcomes in AD animal models following CASP2 inhibitor treatment.

Studies have shown that submicromolar concentrations of CASP2 inhibitors LJ2a and LJ3a prevent synapse loss in primary hippocampal neurons treated with β-amyloid oligomers, suggesting therapeutic potential for AD . These methodological approaches provide a framework for evaluating CASP2 inhibitors as potential therapeutics for these conditions.

What are the critical controls needed when studying CASP2 knockout phenotypes?

When studying CASP2 knockout phenotypes, implementing proper controls is essential for accurate interpretation of results:

  • Generate multiple independent knockout clones: To control for off-target effects of CRISPR/Cas9, use at least two independent guide RNAs targeting different regions of CASP2 and validate multiple clones .

  • Include rescue experiments: Reintroduce wild-type CASP2 and catalytically inactive CASP2 mutants to determine whether observed phenotypes are specifically due to CASP2 loss and which domains/activities are responsible .

  • Control for compensatory upregulation of other caspases: Assess expression levels of other caspase family members (particularly caspase-3, -8, and -9) that might compensate for CASP2 loss, as compensatory expression of closely related caspase homologues is a common confounding factor .

  • Include appropriate genetic background controls: When studying CASP2 in the context of specific genetic backgrounds (e.g., NPM1 mutations), include both wildtype and mutant background controls to discern context-specific functions .

  • Perform both acute and chronic CASP2 depletion experiments: Compare results from siRNA-mediated knockdown (acute) with CRISPR/Cas9 knockout (chronic) to distinguish between immediate versus adaptive responses to CASP2 loss .

These controls help ensure that observed phenotypes are specifically attributed to CASP2 loss rather than experimental artifacts or compensatory mechanisms.

What are the recommended approaches for detecting active CASP2 in human cells and tissues?

Detecting active CASP2 presents technical challenges due to antibody cross-reactivity and low endogenous expression levels. Recommended methodological approaches include:

  • Immunoblotting with CASP2-specific antibodies: Use validated antibodies such as clone 11B4 (Millipore) that specifically recognize the cleaved (active) form of CASP2 .

  • Activity-based probes: Employ caspase-directed activity-based probes like Rho-DEVD-AOMK that covalently bind to active caspases, followed by gel electrophoresis and detection .

  • Fluorogenic substrate assays: Use VDVAD-AFC or similar fluorogenic substrates with caution, recognizing potential cross-reactivity with other caspases. Always include specificity controls using selective CASP2 inhibitors like LJ2a or LJ3a .

  • Proximity ligation assays (PLA): Detect interaction between cleaved CASP2 fragments as an indicator of activation.

  • Immunoprecipitation of CASP2 complexes: Pull down PIDDosome components (PIDD, RAIDD) and assess co-precipitation of CASP2 to detect activation complexes .

  • CASP2 substrate cleavage: Monitor cleavage of validated CASP2 substrates such as MDM2, S1P, or BID as functional readouts of CASP2 activity .

When applying these techniques to human tissues, additional considerations include optimizing fixation methods for immunohistochemistry and employing laser capture microdissection to isolate specific cell populations for analysis.

How can researchers distinguish between CASP2's apoptotic and non-apoptotic functions experimentally?

Distinguishing between CASP2's apoptotic and non-apoptotic functions requires careful experimental design:

  • Use catalytic mutants: Compare the effects of wild-type CASP2 with catalytically inactive mutants (C320A) to determine whether specific functions require proteolytic activity .

  • Temporal analysis: Monitor CASP2 activity and cellular responses at early time points before apoptosis onset to identify non-apoptotic functions that precede cell death.

  • Co-treatment with pan-caspase inhibitors: Apply broad-spectrum caspase inhibitors (e.g., z-VAD-fmk) alongside specific CASP2 inhibitors to separate general apoptotic events from CASP2-specific functions .

  • Cell cycle synchronization: Synchronize cells at specific cell cycle phases to study CASP2's role in cell cycle regulation independently of apoptosis .

  • Sub-lethal stress conditions: Apply mild stress that activates CASP2 without triggering apoptosis to isolate non-apoptotic functions.

  • Subcellular localization studies: Track CASP2 localization during different cellular processes, as non-apoptotic functions may involve localization to specific compartments such as "ubstressomes" during protein quality control .

  • Assessment of specific non-apoptotic substrates: Monitor cleavage of substrates associated with non-apoptotic functions, such as S1P for metabolism regulation or deubiquitination activity for protein quality control .

These approaches have revealed CASP2's non-apoptotic roles in cell cycle regulation, DNA damage repair, ferroptosis protection, and protein quality control that operate independently of its role in cell death.

Product Science Overview

Introduction

Caspase-2, also known as CASP2, is a member of the cysteine-aspartic acid protease (caspase) family. Caspases are crucial for mediating cellular apoptosis through the proteolytic cleavage of specific protein substrates . Caspase-2 is unique among caspases due to its dual role in apoptosis and non-apoptotic cellular processes, including cell cycle regulation and tumor suppression .

Structure and Function

Caspase-2 is synthesized as an inactive zymogen that requires proteolytic cleavage to become active. The human recombinant form of Caspase-2 is produced in E. coli and consists of a single, non-glycosylated polypeptide chain containing 126 amino acids, with a molecular mass of 14.1 kDa . This recombinant protein is often fused to a His-tag for purification purposes .

Role in Apoptosis

Caspase-2 is classified as an initiator caspase, meaning it responds to apoptotic stimuli by initiating the apoptotic cascade . It is considered a pro-apoptotic caspase that can induce cell death through the cleavage of various substrates. However, its exact role in apoptosis has been a subject of debate due to conflicting evidence regarding its necessity for this process .

Non-Apoptotic Functions

Emerging evidence suggests that Caspase-2 also plays a role in non-apoptotic processes such as cell cycle regulation and protection from genomic instability . These functions are particularly relevant to its role as a tumor suppressor. Caspase-2 has been identified as a tumor suppressor in multiple tissue types, although the mechanisms underlying this function are not fully understood .

Clinical Relevance

Increased expression of Caspase-2 has been implicated in various neurodegenerative disorders, including Alzheimer’s disease, Huntington’s disease, and temporal lobe epilepsy . Its role in stress-induced cell death pathways and tumor suppression makes it a potential target for therapeutic interventions in cancer and neurodegenerative diseases .

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.