DBNDD2 Human

Dysbindin (Dystrobrevin Binding Protein 1) Domain Containing 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

DBNDD2 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 185 amino acids (1-161 a.a) and having a molecular mass of 20.1kDa (Molecular weight on SDS-PAGE will appear higher).
DBNDD2 is fused to a 24 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Dysbindin domain-containing protein 2 (DBNDD2), a member of the dysbindin family, regulates casein kinase-1 activity and inhibits CSNK1D autophosphorylation in vitro. It interacts with CSNK1D and CSNK1E and is found in the brain.
Description
Recombinant human DBNDD2, expressed in E. coli, is a non-glycosylated polypeptide chain containing 185 amino acids (residues 1-161) with a molecular mass of 20.1 kDa. Note: SDS-PAGE may show a higher molecular weight. The protein is fused to a 24 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
DBNDD2 protein is supplied in a solution at a concentration of 0.5 mg/ml. The solution contains 20mM Tris-HCl buffer (pH 8.0), 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Dysbindin domain-containing protein 2, Casein kinase-1 binding protein, CK1BP, HSMNP1, DBNDD2, C20orf35.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMDPNPR AALERQQLRL RERQKFFEDI LQPETEFVFP LSHLHLESQR PPIGSISSME VNVDTLEQVE LIDLGDPDAA DVFLPCEDPP PTPQSSGMDN HLEELSLPVP TSDRTTSRTS SSSSSDSSTN LHSPNPSDDG ADTPLAQSDE EEERGDGGAE PGACS.

Q&A

What is DBNDD2 and how is it related to the broader dysbindin protein family?

DBNDD2 belongs to the dysbindin family of proteins, which includes the better-characterized Dystrobrevin-binding protein 1 (DBNDD/Dysbindin). The dysbindin family proteins are components of the dystrophin-associated protein complex (DPC) and are expressed in neural tissue of the brain and skeletal muscle cells. DBNDD2 shares structural domains with its family members but has distinct expression patterns and potentially specialized functions .

Methodologically, researchers distinguish DBNDD2 from other dysbindin family members through:

  • Domain-specific antibodies targeting unique epitopes

  • Isoform-specific primers for RT-PCR expression analysis

  • Proteomic approaches coupled with mass spectrometry

  • Bioinformatic analysis of conserved structural domains

What brain regions and cell types show significant DBNDD2 expression?

DBNDD2, like other dysbindin family proteins, is prominently expressed in synaptic sites throughout the human brain. While dysbindin is known to be particularly abundant in the cerebellum and hippocampus, DBNDD2 shows a somewhat distinct distribution pattern .

Research approaches to map DBNDD2 expression include:

  • Immunohistochemistry with DBNDD2-specific antibodies

  • In situ hybridization for mRNA localization

  • Single-nucleus RNA sequencing for cell-type specific expression profiling

  • Quantitative proteomics from dissected brain regions

Current evidence suggests DBNDD2 expression in neurons and potentially in non-neuronal cells such as oligodendrocytes, though detailed cell-type specific expression maps are still being developed in ongoing research.

What are the established protocols for isolating and characterizing DBNDD2 from human samples?

Isolation and characterization of DBNDD2 from human samples requires specialized techniques:

Sample Preparation:

  • Flash-frozen post-mortem tissue is typically homogenized in buffer containing protease inhibitors

  • Subcellular fractionation may be performed to isolate synaptic versus non-synaptic fractions

  • Immunoprecipitation using DBNDD2-specific antibodies

Characterization Methods:

  • Western blotting with validated antibodies

  • Mass spectrometry for protein identification and post-translational modification analysis

  • Co-immunoprecipitation to identify binding partners

  • Proximity ligation assays for in situ protein interaction studies

Researchers should note that sample quality is critically dependent on post-mortem interval, with optimal results achieved from samples collected within 12 hours of death.

How can researchers experimentally distinguish between the functions of DBNDD2 and other dysbindin family proteins?

Distinguishing the specific functions of DBNDD2 from other dysbindin family proteins presents significant experimental challenges. Robust approaches include:

Genetic Manipulation Strategies:

  • CRISPR/Cas9-mediated knockout or knockin of DBNDD2-specific mutations

  • Isoform-specific RNA interference using validated siRNA or shRNA constructs

  • Rescue experiments with wild-type versus mutant DBNDD2 in knockout models

Functional Assays:

  • Electrophysiological recording following selective manipulation of DBNDD2

  • Live-cell imaging with fluorescently tagged DBNDD2 versus other family members

  • Proteomic comparison of interactomes using BioID or APEX proximity labeling

Comparative Analysis:

  • Cross-species comparison of DBNDD2-specific functions in model organisms

  • Computational modeling of structural differences affecting protein-protein interactions

The interpretation of results should account for potential compensatory mechanisms among family members, which may mask phenotypes in single-gene manipulation studies.

What methodological approaches best capture DBNDD2's potential role in neurological disorders?

Given the association between dysbindin family proteins and conditions like schizophrenia, methodological approaches to investigate DBNDD2's role in neurological disorders should be multi-faceted:

Genetic Association Studies:

  • Case-control studies examining DBNDD2 polymorphisms in disease populations

  • Analysis of rare variants through whole-exome or whole-genome sequencing

  • Expression quantitative trait loci (eQTL) studies linking genetic variation to expression changes

Functional Characterization:

  • Patient-derived induced pluripotent stem cells (iPSCs) differentiated into relevant neural cell types

  • Transcriptomic and proteomic profiling of DBNDD2 in disease versus control samples

  • Investigation of DBNDD2 in post-mortem brain tissue from patients with neurological disorders

Animal Models:

  • Creation and validation of DBNDD2 transgenic or knockout animals

  • Behavioral phenotyping relevant to human neurological symptoms

  • Electrophysiological studies to examine synaptic function

Research has suggested that, similar to dysbindin, altered DBNDD2 expression may contribute to cognitive impairments and memory deficits characteristic of several neurological disorders, though specific mechanisms remain under investigation .

How does DBNDD2 interact with the cellular machinery in synaptic function and stability?

Understanding DBNDD2's role in synaptic function requires sophisticated experimental approaches:

Interaction Studies:

  • Pull-down assays to identify synaptic binding partners

  • Microscopy techniques like FRET or FLIM to visualize protein-protein interactions in situ

  • Quantitative interaction proteomics using BioID or proximity labeling

Functional Synaptic Assays:

  • Patch-clamp electrophysiology following DBNDD2 manipulation

  • Optical techniques to measure synaptic vesicle cycling

  • Super-resolution microscopy to localize DBNDD2 at the synapse

Signal Transduction Analysis:

  • Phosphoproteomic analysis after DBNDD2 manipulation

  • Investigation of DBNDD2's role in receptor trafficking

  • Single-synapse calcium imaging following stimulation

What are the methodological considerations for studying DBNDD2 in the context of Alzheimer's disease and other neurodegenerative conditions?

Recent advances in single-cell and spatial genomics provide new avenues for investigating DBNDD2 in neurodegenerative diseases:

Integration with Cell Atlas Data:

  • Mapping DBNDD2 expression to cell types identified in brain cell atlases

  • Correlation with cell-type vulnerability patterns in neurodegenerative diseases

  • Integration with multi-modal data from initiatives like the BRAIN Initiative Cell Census Network

Temporal Analysis:

  • Longitudinal studies examining DBNDD2 expression across disease progression

  • Pseudo-time trajectory analysis in single-cell data to capture disease continuum

  • Correlation with continuous pseudo-progression scores derived from neuropathological measurements

Spatial Context:

  • Spatial transcriptomics to map DBNDD2 expression relative to pathological features

  • Multiplexed immunofluorescence to visualize DBNDD2 in relation to disease markers

  • Layer-specific analysis within affected cortical regions

Research suggests that proteins involved in synaptic function may show altered expression patterns during the early phase of Alzheimer's disease, characterized by inflammatory microglial and reactive astrocyte states, as well as selective neuronal vulnerability . The potential role of DBNDD2 in these processes warrants specific investigation.

How can computational methods enhance DBNDD2 research and drug discovery efforts?

Computational approaches offer powerful tools for advancing DBNDD2 research:

Structural Modeling:

  • Homology modeling based on related proteins with known structures

  • Molecular dynamics simulations to predict functional conformations

  • Protein-protein docking to identify potential interaction surfaces

Drug Discovery Applications:

  • Virtual screening for compounds that modulate DBNDD2 function

  • Structure-based drug design targeting specific DBNDD2 domains

  • Active learning approaches to optimize compound selection in screening campaigns

Network Analysis:

  • Integration of DBNDD2 into protein-protein interaction networks

  • Pathway enrichment analysis to identify functional contexts

  • Multi-omics data integration to predict functional consequences of DBNDD2 modulation

Human-in-the-loop machine learning approaches, which incorporate expert feedback into the iterative optimization of computational models, are particularly valuable for refining hypotheses about DBNDD2 function and identifying promising therapeutic strategies .

What are the optimal experimental controls for DBNDD2 research?

Antibody Validation:

  • Verification using DBNDD2 knockout or knockdown samples

  • Peptide competition assays to confirm specificity

  • Cross-validation with multiple antibodies targeting different epitopes

Expression Analysis Controls:

  • Multiple reference genes for qPCR normalization

  • Analysis of related family members to assess specificity

  • Inclusion of tissue/cell types with known DBNDD2 expression levels

Experimental Design Considerations:

  • Inclusion of age and sex-matched samples

  • Stratification by relevant genotypes (e.g., APOE status for Alzheimer's studies)

  • Power analysis to determine appropriate sample sizes

Rigorous control design is essential for distinguishing DBNDD2-specific effects from those of related family members and for controlling for the significant confounding factors present in neurological tissue samples.

How can discrepancies in DBNDD2 research findings be reconciled?

Resolving contradictory data on DBNDD2 function requires systematic approaches:

Meta-analysis Strategies:

  • Systematic review of methodological differences between studies

  • Statistical integration of findings with appropriate weighting for study quality

  • Assessment of publication bias in reported results

Standardization Efforts:

  • Development of reference materials and standard protocols

  • Cross-laboratory validation studies

  • Reporting standards for key experimental parameters

Contextual Factors:

  • Evaluation of cell type and tissue-specific effects

  • Consideration of developmental timing in observed phenotypes

  • Assessment of disease stage and severity in clinical studies

The apparent contradictions in research findings may reflect genuine biological complexity, with DBNDD2 potentially serving distinct functions depending on cellular context, developmental stage, and disease state.

What emerging technologies show promise for advancing DBNDD2 human research?

Several cutting-edge technologies hold significant potential for DBNDD2 research:

Advanced Imaging:

  • Cryo-electron microscopy for structural determination

  • Expansion microscopy for improved subcellular localization

  • Live-cell super-resolution microscopy for dynamic studies

Single-Cell Technologies:

  • Spatial transcriptomics to map expression in tissue context

  • Single-cell multi-omics for integrated analysis of genomic, transcriptomic, and proteomic data

  • Patch-seq for correlation of electrophysiological properties with gene expression

Functional Genomics:

  • High-throughput CRISPR screening for functional networks

  • RNA-targeting CRISPR systems for precise transcript modulation

  • Optogenetic and chemogenetic tools for temporal control of DBNDD2 function

These technologies, when applied to DBNDD2 research, promise to reveal new insights into its cellular functions and potential role in neurological disorders.

How might DBNDD2 research inform therapeutic development for neurological disorders?

Translating DBNDD2 research into therapeutic strategies requires:

Target Validation:

  • Genetic evidence linking DBNDD2 to disease phenotypes

  • Demonstration of disease-modifying effects following DBNDD2 modulation

  • Identification of accessible regulatory mechanisms

Therapeutic Modalities:

  • Small molecule modulators of DBNDD2 function or expression

  • Antisense oligonucleotides for selective transcript modulation

  • Gene therapy approaches for genetic forms of dysfunction

Biomarker Development:

  • DBNDD2 expression or post-translational modifications as diagnostic or prognostic markers

  • Identification of DBNDD2-dependent pathways as pharmacodynamic markers

  • Integration into multimodal biomarker panels for patient stratification

Given the association of dysbindin family proteins with conditions like schizophrenia and potentially neurodegenerative diseases, DBNDD2-focused research may yield valuable insights for developing targeted therapeutics for these difficult-to-treat conditions.

Product Science Overview

Introduction

Dysbindin, short for dystrobrevin-binding protein 1, is a protein that plays a crucial role in the dystrophin-associated protein complex (DPC) of skeletal muscle cells. It is also a part of the biogenesis of lysosome-related organelles complex 1 (BLOC-1). Dysbindin was discovered by Derek Blake’s research group through yeast two-hybrid screening for binding partners of α-dystrobrevin .

Structure and Function

Dysbindin is found in neural tissue of the brain, particularly in axon bundles and certain axon terminals, such as mossy fiber synaptic terminals in the cerebellum and hippocampus . It is involved in various cellular processes, including actin cytoskeleton reorganization, regulation of dopamine secretion, and neuron projection morphogenesis .

Dysbindin Domain Containing 2 (DBNDD2)

DBNDD2, or Dysbindin Domain Containing 2, is a protein-coding gene that shares significant homology with dysbindin. It is involved in the negative regulation of protein kinase activity and is predicted to be located in the cytoplasm . Diseases associated with DBNDD2 include Ectodermal Dysplasia 10B and Type 1 Diabetes Mellitus 7 .

Human Recombinant Dysbindin

Human recombinant dysbindin is produced using recombinant DNA technology, which involves inserting the gene encoding dysbindin into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the protein for research and therapeutic purposes.

Clinical Significance

Dysbindin has been implicated in several neurological and psychiatric disorders, including schizophrenia. Studies have shown that dysbindin is involved in synaptic function and plasticity, and its dysfunction can lead to cognitive deficits and other symptoms associated with these disorders .

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.