Ribosome-inactivating protein cucurmosin Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

Ribosome-inactivating protein cucurmosin antibodies are polyclonal immunoglobulins raised against recombinant CUS protein (1–244 amino acids). They specifically recognize CUS and are validated for use in Western blot (WB) and enzyme-linked immunosorbent assay (ELISA) . These antibodies do not cross-react with other RIPs or unrelated proteins, ensuring target specificity.

Key Features

ParameterDetails
HostRabbit
ImmunogenRecombinant CUS (1–244AA)
ApplicationsELISA, WB
Conjugates AvailableHRP, FITC, Biotin, Magnetic Beads, Fluorophores (AF405, AF546, etc.)
DilutionWB: 1:1000–1:5000; ELISA: Varies by conjugate

Immunodetection and Validation

  • Western Blot: Used to confirm CUS expression in recombinant systems or detect CUS in experimental samples .

  • ELISA: Quantifies CUS levels in cell lysates or purified preparations .

  • Immunotoxin Studies: Antibodies may assist in validating CUS conjugation to monoclonal antibodies (e.g., durvalumab) in PD-L1-targeted immunotoxins .

Mechanistic Insights

CUS inhibits protein synthesis via N-glycosidase activity, cleaving adenine residues in 28S rRNA. Antibodies help track CUS’s intracellular localization or binding to target cells in studies of its cytotoxicity .

CUS Antibody in Immunotoxin Development

CUS-based immunotoxins (e.g., D-CUS245C) target PD-L1+ tumors, leveraging CUS’s cytotoxicity. While antibodies against CUS are not directly used in these constructs, they support quality control during toxin production .

Challenges in Antibody Utilization

  • Limited Cross-Reactivity: CUS antibodies show specificity for CUS but may not detect other RIPs (e.g., trichosanthin, ricin) .

  • Research-Only Use: Not approved for clinical diagnostics, restricting applications to preclinical studies .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
Ribosome-inactivating protein cucurmosin antibody; EC 3.2.2.22 antibody; rRNA N-glycosidase antibody
Uniprot No.

Target Background

Function
Exhibits cytotoxic activity against cancer cells, but not normal cells. Inhibits the growth of the human leukemia cell line K562, the murine melanoma cell line B16, and the lung adenocarcinoma cell line A549 with IC50 values of 88.1 nM, 63.4 nM, and 359.3 nM, respectively.
Protein Families
Ribosome-inactivating protein family, Type 1 RIP subfamily

Q&A

What is cucurmosin and how is it classified among ribosome-inactivating proteins?

Cucurmosin (CUS) is a novel type I ribosome-inactivating protein (RIP) isolated from Cucurbita moschata (pumpkin). As a type I RIP, it consists of a single polypeptide chain with N-glycosidase activity (EC 3.2.2.22) that can inactivate ribosomes by cleaving the N-glycosidic bond of adenine in rRNA . Unlike type II RIPs which contain a lectin-binding B chain, type I RIPs like cucurmosin lack this cell-binding domain, which affects their cellular uptake mechanism and potential cytotoxicity profile .

How does the structure of cucurmosin relate to its biological function?

The spatial structure of cucurmosin has been determined through crystallographic studies, revealing structural features that contribute to its ribosomal inactivation capacity. The protein contains crucial catalytic residues that participate in the depurination of the adenine residue in rRNA. Research has shown that understanding this structure-function relationship is essential for engineering modified versions of cucurmosin, such as CUS245C, which contains an engineered cysteine residue at position 245 that facilitates conjugation with antibodies for immunotoxin development .

What are the common antibodies available for detecting cucurmosin in research applications?

Several antibodies targeting cucurmosin are available for research applications, including:

  • Rabbit recombinant polyclonal antibodies conjugated with HRP

  • Non-conjugated polyclonal antibodies generated against recombinant Cucurbita moschata ribosome-inactivating protein cucurmosin

These antibodies have been validated for various applications including ELISA and Western blotting, with specific reactivity against Cucurbita moschata .

What are the main mechanisms through which cucurmosin exerts its anticancer effects?

Cucurmosin demonstrates anticancer effects through multiple cellular mechanisms:

  • Induction of apoptosis: Cucurmosin treatment leads to characteristic features of apoptosis including cell shrinkage, apoptotic bodies, and DNA fragmentation .

  • Mitochondrial pathway activation: Studies with K562 cells showed that cucurmosin causes:

    • Loss of mitochondrial membrane potential (MMP)

    • Increased cytochrome c release into the cytosol

    • Activation of caspase-3

    • Downregulation of anti-apoptotic Bcl-2 protein

    • Upregulation of pro-apoptotic Bax protein

    • Decrease in the Bcl-2/Bax ratio

  • Cell cycle arrest: Cucurmosin treatment results in cell cycle disruption, contributing to its cytotoxic effects .

  • Regulation of metabolic pathways: In pancreatic cancer models, high-dose cucurmosin positively regulates:

    • Pyruvate metabolism

    • Glycolysis/gluconeogenesis

    • Cysteine and methionine metabolism

  • Growth factor receptor signaling inhibition: Cucurmosin inactivates GFR signaling pathways, inducing tumor cell apoptosis .

How does cucurmosin compare to conventional chemotherapeutic agents in cancer treatment models?

In pancreatic cancer xenograft models, high-dose cucurmosin demonstrated comparable therapeutic efficacy to gemcitabine, which is the preferred single chemotherapeutic agent for pancreatic cancer. Importantly, cucurmosin showed a dose-dependent inhibition of pancreatic tumors. Metabolomic analysis revealed that both treatments positively regulated similar metabolic pathways, including pyruvate metabolism, glycolysis/gluconeogenesis, and cysteine and methionine metabolism .

What is the significance of engineering cucurmosin with a cysteine residue at position 245?

The introduction of a cysteine residue at the 245th amino acid position of recombinant cucurmosin (creating CUS245C) represents a significant engineering advancement that facilitates:

  • Enhanced production capabilities through improved folding and stability

  • Specific and controlled cross-linking with antibodies via disulfide bonds

  • Development of targeted immunotoxins with preserved cytotoxic activity

  • Improved conjugation efficiency compared to random conjugation methods

This strategic modification has been crucial for the development of cucurmosin-based immunotoxins such as D-CUS245C, where the engineered cysteine forms a disulfide bond with the antibody component, creating a stable yet conditionally cleavable linkage .

What are the optimal experimental conditions for evaluating cucurmosin's cytotoxicity in vitro?

For optimal evaluation of cucurmosin's cytotoxicity in vitro, researchers should consider the following methodological approaches:

  • Cell line selection:

    • Use appropriate PD-L1 expressing cell lines such as MDA-MB-231 or PD-L1/SPC-A-1

    • Include proper negative controls (e.g., NC/SPC-A-1 for PD-L1 negative control)

  • Incubation time:

    • Extended incubation periods (72-120 hours) show increasing potency of cucurmosin-based immunotoxins

    • Significant differences in IC50 values have been observed between 72h and 120h timepoints

  • Concentration range:

    • For native CUS245C: nanomolar range (150-500 nmol/L)

    • For conjugated immunotoxins (e.g., D-CUS245C): picomolar range (0.14-3.8 pmol/L)

  • Evaluation methods:

    • Cell viability assays (e.g., MTT or CCK-8)

    • Flow cytometry for apoptosis detection

    • Western blotting for protein expression analysis

    • Confocal microscopy for cellular localization studies

What are the critical factors to consider when designing cucurmosin-based immunotoxins?

When designing cucurmosin-based immunotoxins, researchers should consider several critical factors:

  • Target antigen selection:

    • Choose tumor-associated antigens with high expression in target tissues

    • Consider antigens like PD-L1 that have functional roles in tumor progression

    • Evaluate target internalization capacity as this affects immunotoxin efficacy

  • Antibody selection:

    • Use antibodies with high specificity and affinity for the target antigen

    • Consider antibody format (whole IgG vs. fragments)

    • For anti-PD-L1 immunotoxins, clinically validated antibodies like durvalumab have been successfully employed

  • Conjugation chemistry:

    • The disulfide linkage via SPDP (N-succinimidyl 3-(2-pyridyldithio)propionate) has proven effective

    • The engineered cysteine at position 245 in CUS245C provides a specific conjugation site

    • Maintain a 1:1 toxin-to-antibody ratio for consistent activity

  • Purification methods:

    • Size exclusion chromatography to separate conjugated immunotoxin from unconjugated components

    • Protein concentration determination using appropriate methods

    • Verification of conjugate integrity through SDS-PAGE and Western blotting

How should researchers design in vivo experiments to evaluate cucurmosin and cucurmosin-based immunotoxins?

For in vivo evaluation of cucurmosin and cucurmosin-based immunotoxins, researchers should implement the following design considerations:

  • Animal model selection:

    • Xenograft models using immunodeficient mice (e.g., nude mice)

    • Cell line-derived xenografts expressing the target antigen (e.g., PD-L1)

    • Consider subcutaneous vs. orthotopic models depending on the cancer type

  • Dosing strategy:

    • For cucurmosin alone: test multiple doses to establish dose-response relationship

    • For cucurmosin-based immunotoxins: start with low doses (0.4-0.8 mg/kg) based on previous successful studies

    • Establish appropriate dosing schedule (e.g., every 2-3 days)

  • Control groups:

    • Vehicle control

    • Unconjugated antibody control

    • Unconjugated toxin control

    • Positive control (established therapy, e.g., gemcitabine for pancreatic cancer models)

  • Evaluation parameters:

    • Tumor volume measurements

    • Tumor weight at study endpoint

    • Survival analysis

    • Toxicity assessments (body weight, organ weights, serum chemistry)

  • Pharmacokinetic considerations:

    • Half-life determination

    • Biodistribution studies

    • Immunogenicity assessment for repeated dosing

How can researchers optimize cucurmosin-based immunotoxins to overcome resistance mechanisms in cancer therapy?

To overcome resistance mechanisms to cucurmosin-based immunotoxins, researchers can explore several optimization strategies:

  • Combination therapy approaches:

    • Combining cucurmosin immunotoxins with immune checkpoint inhibitors

    • Using cucurmosin immunotoxins with conventional chemotherapeutics

    • Exploring synergy with radiation therapy or targeted small molecule inhibitors

  • Structural modifications:

    • Engineering cucurmosin variants with reduced immunogenicity

    • Creating fusion proteins with altered cellular trafficking properties

    • Developing multi-specific formats that target multiple antigens simultaneously

  • Alternative delivery systems:

    • Nanoparticle encapsulation for improved delivery to tumors

    • Stimuli-responsive release mechanisms

    • Cell-penetrating peptide conjugation for enhanced cellular uptake

  • Addressing immunogenicity:

    • Humanization of protein components

    • PEGylation strategies

    • Targeted delivery to reduce systemic exposure

  • Overcoming tumor heterogeneity:

    • Dual-targeting immunotoxins

    • Co-targeting tumor microenvironment components

    • Sequential administration strategies to address evolving resistance mechanisms

What are the comparative advantages and limitations of cucurmosin versus other ribosome-inactivating proteins in immunotoxin development?

Understanding the comparative profile of cucurmosin relative to other RIPs is essential for immunotoxin development:

Advantages of cucurmosin:

  • High potency when conjugated to targeting antibodies, with IC50 values in the picomolar range for D-CUS245C against PD-L1-positive cells

  • Significant therapeutic index (TI) with values ranging from 88,026 to over 1,100,000 in various cell lines and time points

  • Ability to induce apoptosis through multiple cellular pathways, potentially reducing resistance development

  • Engineered variants (CUS245C) with site-specific conjugation capabilities

  • Demonstrated efficacy in several cancer models, including pancreatic cancer, leukemia, and PD-L1-positive tumors

Limitations and challenges:

  • As with other RIPs, potential immunogenicity concerns for repeated administrations

  • Need for optimization of dose selection and surface modification according to the specific cancer type and stage

  • Requirement for further studies on long-term safety and efficacy profiles

  • Limited data on comparative efficacy versus other well-established RIPs like ricin, gelonin, or saporin

What methodological approaches can be used to study the detailed molecular mechanisms of cucurmosin-induced cell death?

For investigating the detailed molecular mechanisms of cucurmosin-induced cell death, researchers should consider the following methodological approaches:

  • Transcriptomic and proteomic analyses:

    • RNA-seq to identify differentially expressed genes following cucurmosin treatment

    • Proteomic profiling to identify protein level changes and post-translational modifications

    • Phosphoproteomics to analyze signaling pathway alterations

  • Metabolomic studies:

    • Nuclear magnetic resonance (NMR) spectroscopy to analyze serum metabolite profiles as demonstrated in pancreatic cancer models

    • Mass spectrometry-based metabolomics

    • Integrated pathway analysis of altered metabolites

  • Cellular pathway investigation techniques:

    • CRISPR-Cas9 screening to identify genes essential for cucurmosin sensitivity or resistance

    • Mitochondrial function assays (membrane potential, cytochrome c release)

    • Caspase activation assays

    • Bcl-2 family protein expression and interaction studies

  • Imaging approaches:

    • Live-cell imaging to track real-time cellular responses

    • Confocal microscopy for subcellular localization

    • Super-resolution microscopy for detailed structural analysis

  • Protein synthesis inhibition assays:

    • Polysome profiling

    • Measurement of protein synthesis rates using labeled amino acids

    • Ribosome depurination assays to directly measure RIP activity

What analytical methods should be employed to confirm successful conjugation and purity of cucurmosin-based immunotoxins?

To ensure the quality and integrity of cucurmosin-based immunotoxins, researchers should implement multiple analytical methods:

  • Size exclusion chromatography (SEC):

    • To separate conjugated immunotoxin from unconjugated components

    • To determine the molecular weight and homogeneity of the conjugate

  • SDS-PAGE analysis:

    • Under reducing and non-reducing conditions to verify conjugation

    • Followed by Coomassie or silver staining for protein visualization

  • Western blotting:

    • Using anti-CUS antibodies to detect the toxin component

    • Using anti-immunoglobulin antibodies to detect the antibody component

  • Mass spectrometry:

    • For precise molecular weight determination

    • For confirmation of the 1:1 toxin-to-antibody ratio

  • Binding assays:

    • Flow cytometry to verify binding to target-expressing cells

    • ELISA to confirm antigen recognition

    • Surface plasmon resonance to measure binding kinetics

  • Functional assays:

    • Protein synthesis inhibition assays

    • Cell viability/cytotoxicity assays using appropriate target-positive and target-negative cell lines

How can researchers quantitatively evaluate the specificity and potency of cucurmosin-based immunotoxins?

To quantitatively evaluate specificity and potency of cucurmosin-based immunotoxins, researchers should consider these methodological approaches:

  • Calculation of therapeutic index (TI):

    • Determine IC50 values on target-positive and target-negative cell lines

    • Calculate the ratio between IC50 on negative cells and IC50 on positive cells

    • Previous studies with D-CUS245C demonstrated TI values ranging from 88,026 to 1,102,143, indicating exceptional specificity

  • Competitive binding assays:

    • Perform displacement studies using unconjugated antibody

    • Measure reduction in cytotoxicity when target antigen is blocked

  • Dose-response curves:

    • Generate complete dose-response curves at multiple time points

    • Compare slopes and IC50 values between:

      • Unconjugated toxin

      • Immunotoxin on target-positive cells

      • Immunotoxin on target-negative cells

      • Non-targeting control immunotoxin

  • In vivo efficacy metrics:

    • Tumor growth inhibition (TGI) calculations

    • Comparison of tumor weights between treatment groups

    • Survival analysis with statistical evaluation

  • Binding specificity assessment:

    • Flow cytometry across a panel of cell lines with varying target expression levels

    • Correlation analysis between target expression level and cytotoxic potency

Cell LineCUS245C (nmol/L)D-CUS245C (pmol/L)Therapeutic Index
PD-L1/SPC-A-1 (72h)363.3 ± 10.683.8 ± 1.2788,026.3
PD-L1/SPC-A-1 (120h)365.7 ± 13.392.8 ± 0.081135,678.6
MDA-MB-231 (72h)503.7 ± 0.111.6 ± 0.11314,812.5
MDA-MB-231 (120h)154.3 ± 6.230.14 ± 0.00751,102,143
NC/SPC-A-1 (72h)994.7 ± 33.63>1000-
NC/SPC-A-1 (120h)375.6 ± 13.56>1000-

Table: IC50 values and therapeutic indices for CUS245C and D-CUS245C immunotoxin across different cell lines and timepoints .

What are promising approaches for developing next-generation cucurmosin-based therapeutics?

Several innovative approaches could advance the development of next-generation cucurmosin-based therapeutics:

  • Multi-specific immunotoxins:

    • Bispecific formats targeting both PD-L1 and another tumor-associated antigen

    • Trispecific constructs incorporating immune cell recruitment capabilities

    • Dual-toxin conjugates with synergistic cell-killing mechanisms

  • Engineered delivery systems:

    • Antibody-drug conjugate (ADC) technologies with cleavable linkers

    • Nanoparticle formulations for improved tumor penetration

    • Liposomal encapsulation to reduce immunogenicity and improve pharmacokinetics

  • Combination therapy optimization:

    • Rational combinations with checkpoint inhibitors

    • Integration with conventional chemotherapy regimens

    • Sequential treatment protocols based on tumor response dynamics

  • Structural engineering:

    • Identification and modification of immunogenic epitopes

    • Domain swapping with other RIPs to create hybrid toxins

    • Introduction of additional functional domains (e.g., cell-penetrating peptides)

  • Expanded target applications:

    • Beyond PD-L1 to other immune checkpoints

    • Targeting cancer stem cell markers

    • Applications in hematological malignancies and rare cancer types

What knowledge gaps remain in understanding the full potential of cucurmosin in cancer therapy?

Despite significant progress in cucurmosin research, several important knowledge gaps remain:

  • Long-term safety profile:

    • Comprehensive toxicology studies across multiple species

    • Evaluation of immunogenicity with repeated administrations

    • Assessment of potential off-target effects

  • Resistance mechanisms:

    • Molecular mechanisms of acquired resistance

    • Biomarkers predicting sensitivity or resistance

    • Strategies to overcome or prevent resistance development

  • Combination therapy rationale:

    • Optimal sequencing of cucurmosin immunotoxins with other therapies

    • Identification of synergistic drug combinations

    • Development of rational combination strategies based on mechanism of action

  • Expanded applications:

    • Efficacy in rare cancer types

    • Activity against cancer stem cells

    • Potential applications beyond oncology

  • Translational considerations:

    • Optimal dosing strategies for clinical translation

    • Pharmacokinetic/pharmacodynamic (PK/PD) relationship

    • Patient selection strategies for clinical trials

    • Manufacturing scalability and stability considerations

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.