Recombinant Human Bcl-2/adenovirus E1B 19 kDa-interacting protein 2-like protein (BNIPL)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, but this can be adjusted to meet customer needs.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
Tag type is determined during production. Please specify your required tag type for preferential development.
Synonyms
BNIPL; Bcl-2/adenovirus E1B 19 kDa-interacting protein 2-like protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-357
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
BNIPL
Target Protein Sequence
MGTIQEAGKK TDVGVREIAE APELGAALRH GELELKEEWQ DEEFPRLLPE EAGTSEDPED PKGDSQAAAG TPSTLALCGQ RPMRKRLSAP ELRLSLTKGP GNDGASPTQS APSSPDGSSD LEIDELETPS DSEQLDSGHE FEWEDELPRA EGLGTSETAE RLGRGCMWDV TGEDGHHWRV FRMGPREQRV DMTVIEPYKK VLSHGGYHGD GLNAVILFAS CYLPRSSIPN YTYVMEHLFR YMVGTLELLV AENYLLVHLS GGTSRAQVPP LSWIRQCYRT LDRRLRKNLR ALVVVHATWY VKAFLALLRP FISSKFTRKI RFLDSLGELA QLISLDQVHI PEAVRQLDRD LHGSGGT
Uniprot No.

Target Background

Function

May act as a bridging molecule between BCL2 and ARHGAP1/CDC42 in the promotion of cell death.

Gene References Into Functions
  1. Two BNIP-S splicing variants function as cellular regulators; the BCH domain of BNIP-Sα exhibits a novel apoptotic function. PMID: 11741952
  2. Interaction with cell proliferation-related proteins MIF and GFER. PMID: 12681488
  3. BNIPL-2, a BNIP-2 homologue, interacts with Bcl-2 and Cdc42GAP in apoptosis. PMID: 12901880
  4. Bcl-2/adenovirus E1B 19 ku interacting protein 2-like (BNIPL-2) may regulate apoptosis by altering the expression of genes involved in apoptosis, growth inhibition, and cell proliferation. PMID: 15112343
  5. BNIPL-1 upregulates p16INK4, IL-12, TRAIL, and lymphotoxin beta (genes associated with growth suppression or apoptosis) and downregulates PTEN, suggesting growth inhibition via cell cycle arrest and/or apoptotic pathways. PMID: 16143817
Database Links

HGNC: 16976

OMIM: 611275

KEGG: hsa:149428

STRING: 9606.ENSP00000357927

UniGene: Hs.591473

Tissue Specificity
Isoform 2 is expressed in placenta and lung.

Q&A

What is the structural characterization of recombinant human BNIPL?

Recombinant human BNIPL belongs to the Bcl-2/adenovirus E1B 19 kDa-interacting protein family and contains key structural elements that determine its function. The protein contains a motif similar to the BH3 domain which is conserved across Bcl-2 family proteins, as well as a membrane-anchoring domain essential for subcellular localization . Immunofluorescence microscopy studies have confirmed that BNIPL localizes to the mitochondria when the membrane-anchoring domain is present . This localization is critical for its pro-apoptotic functions.

The protein's structure enables interaction with anti-apoptotic proteins like Bcl-2 and Bcl-xL, which is consistent with its role in promoting apoptosis . To effectively study recombinant BNIPL structure, researchers typically employ a combination of techniques including X-ray crystallography, nuclear magnetic resonance spectroscopy, and computational modeling to develop a comprehensive understanding of the protein's three-dimensional configuration and functional domains.

How does recombinant BNIPL induce apoptosis in cellular models?

Recombinant BNIPL induces apoptosis through several well-characterized mechanisms. When expressed in cell lines such as Rat-1 and HeLa cells, BNIPL triggers programmed cell death through mitochondrial pathways . The specific mechanism involves the interaction of BNIPL's BH3 domain with anti-apoptotic proteins like Bcl-2 and Bcl-xL, effectively neutralizing their protective function . Mutational analysis has conclusively demonstrated that both the BH3 domain and the membrane-anchoring domain are required for BNIPL to induce cell death .

At the mitochondrial level, recombinant BNIPL contributes to apoptosis by inducing membrane potential loss and cytochrome c release, both of which are prerequisites for apoptotic cell death . This mechanism has been validated through in vitro experiments with isolated mitochondria, where the addition of recombinant BNIPL directly induced these apoptotic events . These findings strongly support the classification of BNIPL as a BH3-containing pro-apoptotic protein that specifically targets mitochondria when inducing apoptosis.

What are the optimal expression systems for producing recombinant human BNIPL?

The selection of an appropriate expression system is critical for producing functional recombinant human BNIPL for research purposes. Based on current methodological approaches in protein expression, several systems have demonstrated efficacy for BNIPL production:

Expression SystemAdvantagesLimitationsYieldPurification Method
E. coliCost-effective, high yield, rapid growthPotential for inclusion bodies, lack of post-translational modifications10-15 mg/L cultureNi-NTA affinity chromatography followed by size exclusion
Mammalian cells (HEK293, CHO)Native-like post-translational modifications, proper foldingHigher cost, lower yield, longer production time2-5 mg/L cultureImmunoaffinity chromatography
Baculovirus-infected insect cellsIntermediate yield, proper folding, some post-translational modificationsMore complex than bacterial systems5-8 mg/L cultureFLAG-tag or His-tag affinity purification

For structural and functional studies of BNIPL, the expression system should be selected based on experimental requirements. Bacterial systems are suitable for basic structural studies and large-scale production, while mammalian expression systems are preferable for functional studies where post-translational modifications are critical for activity.

How can differential gene expression analysis be optimized when studying BNIPL-regulated pathways?

Differential gene expression analysis has proven valuable for understanding the broader impact of BNIPL on cellular pathways. As demonstrated in studies with human hepatocellular carcinoma cells, cDNA expression microarray technology can effectively analyze the differentially expressed genes regulated by BNIPL and related family members . When designing such experiments, researchers should consider several methodological optimizations:

First, appropriate control and experimental groups must be established, typically comparing cells with induced or overexpressed BNIPL against those with normal or knocked-down BNIPL expression. Time-course experiments can provide valuable insights into immediate versus delayed transcriptional responses. RNA extraction quality is critical, with RIN (RNA Integrity Number) values above 8 recommended for reliable results.

For data analysis, researchers should employ robust normalization methods to account for technical variation, and statistical approaches that control for multiple testing (such as FDR correction). Validation of microarray or RNA-seq findings through qRT-PCR for selected genes is essential for confirming expression changes. Pathway analysis tools such as GSEA, KEGG pathway mapping, or Ingenuity Pathway Analysis can help contextualize gene expression changes within biological processes, particularly focusing on apoptotic and mitochondrial pathways known to be influenced by BNIPL.

What experimental design considerations are crucial when studying BNIPL interactions with Bcl-2 family proteins?

When investigating BNIPL interactions with Bcl-2 family proteins, experimental design is critical for obtaining reliable and interpretable results. The experimental approach should follow the five key steps of experimental design outlined in scientific methodology :

  • Clearly define variables: The independent variable (typically BNIPL concentration or mutation status) and dependent variables (measures of interaction with Bcl-2 family proteins or downstream effects) must be precisely defined .

  • Formulate specific, testable hypotheses about BNIPL interactions based on its structural domains, particularly focusing on the BH3 domain that facilitates binding to anti-apoptotic proteins like Bcl-2 and Bcl-xL .

  • Design treatments to manipulate BNIPL expression, including wild-type and domain-specific mutants to determine the contribution of specific regions to protein-protein interactions .

  • Use appropriate subject assignment (between-subjects or within-subjects design) when comparing different experimental conditions .

  • Establish precise measurements for quantifying interaction strength, including techniques such as co-immunoprecipitation, surface plasmon resonance, or FRET-based approaches .

To minimize experimental bias and ensure reproducibility, controls should include both positive controls (known interacting pairs) and negative controls (proteins that should not interact with BNIPL). Additionally, researchers should consider the potential impact of fusion tags on protein function and interaction, ideally validating key findings using multiple detection methods.

How can researchers effectively measure BNIPL-induced mitochondrial membrane potential changes?

BNIPL has been shown to induce mitochondrial membrane potential loss, a critical event in the apoptotic cascade . To effectively measure these changes, researchers should consider several methodological approaches:

Fluorescent probe-based methods represent the gold standard for membrane potential assessment. JC-1 (5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide) is particularly valuable as it exhibits potential-dependent accumulation in mitochondria, indicated by a fluorescence emission shift from green (~529 nm) to red (~590 nm). Tetramethylrhodamine ethyl ester (TMRE) and MitoTracker Red CMXRos provide alternative options, with quantification possible through flow cytometry or confocal microscopy.

For real-time monitoring of membrane potential changes in response to recombinant BNIPL addition, researchers can employ time-lapse imaging using isolated mitochondria preparations or permeabilized cells. This approach allows for the calculation of kinetic parameters and dose-response relationships. Oxygen consumption rate (OCR) measurements using platforms like Seahorse XF analyzers can complement membrane potential studies by assessing the functional impact of BNIPL on mitochondrial respiration.

When designing these experiments, critical controls should include known depolarizing agents (e.g., CCCP or FCCP) as positive controls and mutant BNIPL lacking the BH3 or membrane-anchoring domains as negative controls . Researchers should also consider the potential impact of different cell types on the kinetics and magnitude of BNIPL-induced membrane potential changes.

What are the common pitfalls when investigating BNIPL interactions with other proteins?

Investigating protein-protein interactions involving BNIPL presents several technical challenges that researchers should anticipate and address. One common pitfall is non-specific binding during co-immunoprecipitation experiments, which can lead to false-positive results. To mitigate this, researchers should optimize salt and detergent concentrations in wash buffers and include appropriate negative controls, such as IgG or irrelevant antibodies.

Another challenge arises from the potential impact of fusion tags on BNIPL's interaction capabilities. While tags facilitate purification and detection, they may sterically hinder protein-protein interactions or induce conformational changes. Researchers should consider using small tags (e.g., FLAG or His) and validating key findings with differently tagged constructs or tag-free proteins when possible. The orientation of the tag (N- or C-terminal) should also be considered, especially given BNIPL's membrane-anchoring domain location.

Expression level disparities can also skew interaction results. Overexpression systems may force interactions that do not occur at physiological concentrations, while low expression may fall below detection thresholds. To address this, researchers should consider inducible expression systems for controlled protein levels and complement overexpression studies with endogenous protein interaction analysis when feasible.

How can researchers address the challenge of BNIPL solubility in functional studies?

The membrane-anchoring domain of BNIPL presents significant challenges for protein solubility during recombinant expression and purification . These challenges can compromise functional studies if not properly addressed. Several methodological approaches can help overcome these limitations:

First, researchers can employ detergent screening to identify optimal solubilization conditions. A systematic evaluation of detergents (ranging from harsh ionic detergents like SDS to milder non-ionic detergents like Triton X-100, CHAPS, or digitonin) can identify conditions that maintain protein solubility without compromising functional integrity. Alternatively, creating truncated constructs that lack the membrane-anchoring domain while preserving the functional BH3 domain can improve solubility, though researchers must verify that such modifications do not alter core functions.

Fusion partners known to enhance solubility (such as MBP, GST, or SUMO) can be employed, with subsequent removal using specific proteases if necessary. For functional studies requiring the full-length protein, liposome reconstitution represents an elegant solution, allowing the membrane-anchoring domain to insert into a lipid bilayer mimicking the mitochondrial membrane environment.

The table below summarizes effective approaches for addressing BNIPL solubility challenges:

ApproachMethodologyAdvantagesLimitations
Detergent solubilizationScreen multiple detergents; optimize concentrationMaintains full-length proteinPotential impact on protein-protein interactions
Truncated constructsDesign constructs lacking membrane domainImproved solubilityMay affect some functional aspects
Solubility-enhancing tagsMBP, GST, or SUMO fusionSignificantly improved expression and solubilityLarge tags may interfere with function
Liposome reconstitutionIncorporate protein into artificial membraneNative-like environment for functional studiesTechnical complexity and lower yield

What strategies can overcome the challenge of BNIPL instability during purification and storage?

Protein instability represents a significant challenge when working with recombinant BNIPL. The pro-apoptotic nature and specific structural features of BNIPL can lead to aggregation, degradation, or loss of function during purification and storage. Several evidence-based strategies can address these challenges:

Buffer optimization is crucial for maintaining BNIPL stability. Researchers should systematically test different pH conditions (typically in the range of 6.5-8.0), salt concentrations (150-500 mM NaCl), and stabilizing additives such as glycerol (10-20%), reducing agents (DTT or β-mercaptoethanol), and protease inhibitors. For membrane-associated forms of BNIPL, the inclusion of appropriate detergents above their critical micelle concentration is essential.

Chromatography strategies should minimize protein exposure to harsh conditions. Fast protein liquid chromatography (FPLC) with refrigerated systems helps maintain protein integrity during purification. Sequential purification steps should be carefully planned to minimize handling time, with immediate buffer exchange into stabilizing conditions following elution from affinity columns.

Proper storage conditions are critical for maintaining long-term stability. Flash freezing aliquots in liquid nitrogen and storage at -80°C with cryoprotectants like glycerol or sucrose can preserve activity. For working stocks, researchers should validate protein stability at 4°C over time and establish appropriate usage windows. Avoid repeated freeze-thaw cycles, which significantly accelerate protein degradation and aggregation.

How can BNIPL research contribute to understanding cancer cell apoptosis resistance mechanisms?

BNIPL research offers significant potential for elucidating cancer cell apoptosis resistance mechanisms. As a pro-apoptotic protein that interacts with anti-apoptotic Bcl-2 family members and induces mitochondrial membrane potential loss, BNIPL represents a valuable model for understanding how cancer cells evade programmed cell death . Studies with human hepatocellular carcinoma Hep3B cells have demonstrated that BNIPL and related family members can induce significant gene expression changes related to apoptosis pathways .

To leverage BNIPL research for understanding cancer cell apoptosis resistance, researchers can employ several methodological approaches. Comparative expression analysis between normal and cancer tissues can reveal whether BNIPL downregulation contributes to apoptosis resistance in specific cancer types. Structure-function studies focusing on the BH3 domain of BNIPL can provide insights into how cancer cells might develop resistance through mutations that disrupt pro-apoptotic protein interactions.

Additionally, researchers can investigate the regulatory mechanisms controlling BNIPL expression and activity in cancer cells, including transcriptional regulation, post-translational modifications, and protein stability. High-throughput screening approaches using cancer cell lines with varying degrees of apoptosis resistance may identify compounds that restore sensitivity by modulating BNIPL function or mimicking its pro-apoptotic activity. These investigations could ultimately inform the development of novel therapeutic strategies that overcome apoptosis resistance in cancer.

What methodological approaches can determine BNIPL's role in mitochondrial dynamics beyond apoptosis?

While BNIPL is well-established as a pro-apoptotic protein targeting mitochondria, emerging research suggests broader roles in mitochondrial dynamics. To investigate these potential functions, researchers should consider several methodological approaches that extend beyond traditional apoptosis assays.

Live-cell imaging using fluorescently tagged BNIPL combined with mitochondrial markers can reveal dynamic interactions and effects on mitochondrial morphology, fusion/fission events, and trafficking. Super-resolution microscopy techniques such as STED or STORM can provide nanoscale resolution of BNIPL's precise localization within mitochondrial compartments. Complementary biochemical approaches such as proximity labeling (BioID or APEX2) can identify the mitochondrial interactome of BNIPL beyond known Bcl-2 family proteins.

Functional impacts on mitochondrial processes can be assessed through comprehensive mitochondrial function analysis. This includes measuring oxygen consumption rates, ATP production, reactive oxygen species generation, and calcium handling in response to BNIPL expression or addition. CRISPR-Cas9 gene editing to create BNIPL knockout cell lines provides a valuable tool for assessing mitochondrial parameters in the absence of BNIPL, revealing potential roles in maintaining mitochondrial homeostasis under normal conditions.

A particularly promising approach involves using inducible expression systems to study the temporal effects of BNIPL on mitochondrial dynamics, distinguishing between immediate responses that may be linked to apoptosis initiation and delayed effects that might indicate roles in mitochondrial quality control or bioenergetics regulation.

How might structural modifications of recombinant BNIPL be utilized to develop targeted therapeutic approaches?

The structural and functional characterization of BNIPL provides a foundation for developing therapeutic approaches that either mimic or modulate its pro-apoptotic activity. Several methodological strategies can be employed to translate BNIPL research into therapeutic applications:

BH3 domain peptide mimetics represent a promising approach. By isolating the specific BH3 domain sequence from BNIPL that interacts with anti-apoptotic proteins like Bcl-2 and Bcl-xL, researchers can develop peptide-based therapeutics that mimic BNIPL's pro-apoptotic function . These peptides can be chemically modified to enhance stability, cell penetration, and mitochondrial targeting. Structure-based design using crystallographic data of BNIPL-Bcl-2 interactions can guide the development of small molecule compounds that mimic the BH3 domain binding interface.

For targeting approaches, researchers can exploit BNIPL's membrane-anchoring domain as a delivery vehicle for directing therapeutic cargo to mitochondria . This approach could enhance the efficacy of existing cancer therapeutics by improving their mitochondrial localization. Additionally, conditionally active BNIPL variants could be designed that become activated in response to cancer-specific conditions (pH, protease activity, or redox status), providing tumor selectivity.

BNIPL-inspired therapeutics would require extensive validation, beginning with in vitro testing in cancer cell lines with defined Bcl-2 family expression profiles, followed by animal models to assess efficacy, delivery, and potential off-target effects. The ultimate goal would be to develop therapeutic strategies that selectively induce apoptosis in cancer cells while sparing normal tissues.

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.