KPNB1 Human

Karyopherin Beta 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Splice Variants

Two isoforms have been identified:

  1. 731 aa variant: Starts at Met146, missing the N-terminal IBB domain.

  2. 845 aa variant: Contains a 2 aa substitution (702–734) .

Nuclear Transport Mechanism

KPNB1 operates in two modes:

  1. Adaptor-dependent: Binds importin α, forming a heterodimer that recognizes nuclear localization signals (NLS) on cargo proteins (e.g., transcription factors, ribosomal proteins) .

  2. Adaptor-independent: Directly binds NLS-containing cargoes (e.g., ribosomal proteins RPL23A, RPS7, RPL5) .

Regulation of Cellular Pathways

PathwayKPNB1’s RoleImpact
Wnt SignalingTransports β-catenin and TCF/LEF into the nucleusModulates stem cell renewal
NF-κB SignalingImports NF-κB subunits, enabling pro-inflammatory gene expressionLinked to cancer progression
MitosisRegulates spindle assembly, kinetochore attachment, and nuclear envelop reformationCritical for cell division

Cancer

KPNB1 is upregulated in glioblastoma, ovarian, breast, and cervical cancers, acting as an oncogene:

  • Apoptosis Resistance: Overexpression suppresses pro-apoptotic proteins (BAX, BAK) and upregulates anti-apoptotic Mcl-1 .

  • Proteostasis Disruption: Inhibition induces unfolded protein response (UPR), leading to apoptosis via Puma/Noxa upregulation .

Neurodegenerative Disorders

Impaired KPNB1 function correlates with neurodegeneration:

  • Toxic Aggregate Formation: Cytosolic retention of cargoes promotes aggresome-like structures (ALIS), exacerbating neuronal stress .

Viral Infections

Viruses hijack KPNB1 for nuclear entry, highlighting its role in pathogen replication .

Inhibitors and Mechanisms

InhibitorMechanismOutcome
KPNB1 siRNADepletes KPNB1, causing mitotic arrestInduces apoptosis in glioblastoma
Small-Molecule InhibitorsBlocks cargo binding (e.g., ivermectin derivatives)Abrogates nuclear import of oncogenic proteins
Bcl-xL InhibitorsSynergizes with KPNB1 inhibition to enhance apoptosis in cancer cellsSensitizes tumors to therapy

Diagnostic and Therapeutic Applications

  • Biomarker: Elevated KPNB1 levels correlate with poor prognosis in ovarian cancer .

  • Drug Delivery: Exploited to shuttle therapeutic agents into nuclei .

Recombinant KPNB1 Human

PropertySpecificationUse Case
SourceE. coli (His-tagged)In vitro binding assays
Purity>85%Structural studies (e.g., X-ray crystallography)
ActivityBinds Ran-GTP and FxFG nucleoporinsNuclear transport assays

Product Specs

Introduction
KPNB1, a member of the importin beta family, plays a crucial role in nuclear protein import. It achieves this by either forming a complex with an adapter protein like importin-alpha (which binds to nuclear localization signals (NLS) on cargo substrates) or by acting independently as a nuclear transport receptor, facilitating the docking of the importin/substrate complex to the nuclear pore complex.
Description
Recombinant KPNB1 Human protein, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 899 amino acids (specifically, amino acids 1 to 876). With a molecular weight of 99.6 kDa, this protein features a 23 amino acid His-tag at its N-terminus. It undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized through filtration.
Formulation
This solution contains 0.25 mg/ml of KPNB1 protein in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, 30% glycerol, and 0.1M NaCl.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For longer storage, freeze the solution at -20°C. It is advisable to add a carrier protein (0.1% HSA or BSA) for extended storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity level exceeding 85%.
Synonyms
Importin subunit beta-1, Importin-90, Karyopherin subunit beta-1, Nuclear factor p97, Pore targeting complex 97kDa subunit, PTAC97, KPNB1, NTF97.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMELITIL EKTVSPDRLE LEAAQKFLER AAVENLPTFL VELSRVLANP GNSQVARVAA GLQIKNSLTS KDPDIKAQYQ QRWLAIDANA RREVKNYVLQ TLGTETYRPS SASQCVAGIA CAEIPVNQWP ELIPQLVANV TNPNSTEHMK ESTLEAIGYI CQDIDPEQLQ DKSNEILTAI IQGMRKEEPS NNVKLAATNA LLNSLEFTKA NFDKESERHF IMQVVCEATQ CPDTRVRVAA LQNLVKIMSL YYQYMETYMG PALFAITIEA MKSDIDEVAL QGIEFWSNVC DEEMDLAIEA SEAAEQGRPP EHTSKFYAKG ALQYLVPILT QTLTKQDEND DDDDWNPCKA AGVCLMLLAT CCEDDIVPHV LPFIKEHIKN PDWRYRDAAV MAFGCILEGP EPSQLKPLVI QAMPTLIELM KDPSVVVRDT AAWTVGRICE LLPEAAINDV YLAPLLQCLI EGLSAEPRVA SNVCWAFSSL AEAAYEAADV ADDQEEPATY CLSSSFELIV QKLLETTDRP DGHQNNLRSS AYESLMEIVK NSAKDCYPAV QKTTLVIMER LQQVLQMESH IQSTSDRIQF NDLQSLLCAT LQNVLRKVQH QDALQISDVV MASLLRMFQS TAGSGGVQED ALMAVSTLVE VLGGEFLKYM EAFKPFLGIG LKNYAEYQVC LAAVGLVGDL CRALQSNIIP FCDEVMQLLL ENLGNENVHR SVKPQILSVF GDIALAIGGE FKKYLEVVLN TLQQASQAQV DKSDYDMVDY LNELRESCLE AYTGIVQGLK GDQENVHPDV MLVQPRVEFI LSFIDHIAGD EDHTDGVVAC AAGLIGDLCT AFGKDVLKLV EARPMIHELL TEGRRSKTNK AKTLATWATK ELRKLKNQA.

Q&A

What is the core function of KPNB1 in human cellular transport systems?

KPNB1 (karyopherin subunit beta 1, also known as importin β) functions as a critical nuclear transport receptor that mediates the translocation of proteins from the cytoplasm to the nucleus. Methodologically, this function can be studied through:

  • Immunofluorescence microscopy to track subcellular localization patterns

  • Co-immunoprecipitation experiments to identify interaction partners

  • RNAi depletion to observe functional consequences

KPNB1 operates through two primary mechanisms:

  • Classical importin α-dependent pathway: KPNB1 mediates docking of nuclear localization signal (NLS)-containing cargo bound to importin α to the nuclear envelope

  • Direct cargo recognition: KPNB1 can directly recognize certain cargoes and facilitate nuclear transport independently of importin α

Research indicates that KPNB1 is located on human chromosome 17 (17q21) and contains 23 exons, making it a structurally complex protein with multiple functional domains .

How does KPNB1 regulate protein transport across the nuclear membrane?

KPNB1 regulates nuclear transport through a multi-step process:

  • Recognition of cargo proteins (either directly or via adaptor proteins like importin α)

  • Docking of the transport complex at nuclear pore complexes (NPCs)

  • Translocation through the nuclear pore

  • Release of cargo in the nucleus

  • Recycling of KPNB1 back to the cytoplasm

Experimental approaches to study this process include:

TechniqueApplicationKey Measurements
Live-cell imagingReal-time transport dynamicsTransport rates, cargo accumulation
FRAP (Fluorescence Recovery After Photobleaching)Transport kineticsRecovery half-time, mobile fraction
Proximity ligation assaysProtein-protein interactionsInteraction sites, temporal dynamics
Selective inhibitors (e.g., importazole)Functional perturbationTransport inhibition efficiency

After the target molecule is transported into the nucleus, KPNB1 binds to RanGTP, resulting in its dissociation from cargo. Free KPNB1 then returns to the cytoplasm for subsequent rounds of nuclear transport .

What is the mechanism by which KPNB1 regulates the circadian clock?

KPNB1 plays a crucial role in circadian rhythm regulation by mediating the nuclear translocation of PER/CRY repressor complexes, which is essential for the negative feedback loop of the circadian clock. Methodological approaches to investigate this relationship include:

  • Interaction studies: Co-immunoprecipitation experiments reveal that KPNB1 interacts more strongly with PER1 and PER2 proteins than with CRY1 or CRY2, suggesting that PER proteins play a leading role in PER/CRY nuclear translocation with KPNB1 .

  • Temporal dynamics analysis: KPNB1 exhibits a circadian pattern of nucleocytoplasmic localization with its nuclear abundance peaking at circadian time (CT) 14-18, coinciding with nuclear accumulation of PER2 and CRY1 .

  • Loss-of-function experiments: RNAi depletion of KPNB1 in U2 OS cells:

    • Elevates Per1 promoter activity

    • Reduces recruitment of PER1 and PER2 to CLOCK/BMAL1 responsive elements

    • Upregulates E-box dependent genes (PER1, CRY1, DBP, REV-ERBβ)

    • Severely disrupts circadian bioluminescence reporter activity

  • Model systems: Inducible inhibition of importin β in Drosophila lateral neurons abolishes behavioral rhythms, demonstrating evolutionary conservation of this mechanism .

How can researchers experimentally distinguish between KPNB1-dependent and independent nuclear transport of circadian proteins?

To differentiate between KPNB1-dependent and independent nuclear transport of circadian proteins, researchers can implement several strategic approaches:

  • Domain mutation studies: Create constructs with mutations in the NLS or KPNB1-binding domains of clock proteins to determine specificity of transport pathways.

  • Selective inhibition: Unlike broad nuclear transport inhibition, researchers should:

    • Use importazole (IPZ) which specifically inhibits KPNB1-mediated import

    • Compare results with inhibitors of other transport pathways

    • Conduct rescue experiments with KPNB1 mutants resistant to the inhibitor

  • Importin α dependency testing: Evidence shows KPNB1 functions independently of importin α in circadian regulation:

    • Deletion of the importin α binding domain of KPNB1 does not significantly affect transcriptional regulation

    • Knockdown of importin α1 (KPNA2) or importin α5 (KPNA1) does not affect circadian rhythmicity

    • KPNA2 or KPNA1 depletion does not alter PER2 nuclear localization, unlike KPNB1 depletion

  • Temporal resolution experiments: Monitor nuclear transport at different circadian time points to identify specific windows when KPNB1-dependency is highest.

What are the mechanisms underlying KPNB1's role in cancer progression?

KPNB1 is overexpressed in multiple cancer types and promotes cancer progression through several mechanisms that can be experimentally investigated:

  • Cell proliferation regulation:

    • Silencing KPNB1 in cancer cells (e.g., K562, K562R in CML) significantly inhibits clonal formation and reduces growth and proliferation

    • Methodological approach: Colony formation assays and proliferation assays using siRNA knockdown or inhibitors like importazole (IPZ)

  • Cell cycle control:

    • KPNB1 inhibition causes cell cycle arrest at G2/M phase

    • Research technique: Flow cytometry to analyze cell cycle distribution after KPNB1 inhibition

  • Apoptosis regulation:

    • KPNB1 inhibition promotes apoptosis through:

      • Upregulation of Puma and Noxa

      • Release of Mcl-1-sequestered Bax and Bak

      • Mitochondrial outer membrane permeabilization (MOMP)

    • Methodology: Annexin V staining and flow cytometry to quantify apoptosis rates

  • Oncogenic signaling:

    • KPNB1 mediates nuclear transport of transcription factors critical for cancer growth

    • In CML cells, KPNB1 mediates E2F1 nuclear transport, affecting downstream targets like c-Myc and KPNA2

    • Approach: Immunofluorescence to track subcellular localization of transcription factors

  • Proteostasis maintenance:

    • KPNB1 inhibition disrupts cellular proteostasis

    • Results in elevated polyubiquitination, formation of aggresome-like-induced structures, and unfolded protein response (UPR)

    • UPR activation leads to apoptosis through chronic activation of eIF2α/ATF4 cascade

    • Methodology: Western blotting to detect UPR markers, ubiquitination assays

Clinical data shows that EOC patients with higher expression levels of KPNB1 displayed earlier recurrence and worse prognosis than those with lower KPNB1 expression .

What are the most effective research models and methodologies for studying KPNB1 inhibition in cancer therapy?

For researchers investigating KPNB1 as a cancer therapeutic target, several models and methodologies have proven effective:

  • In vitro cancer cell line models:

    • Established cell lines with differential KPNB1 expression:

      • CML: K562, K562R (imatinib-resistant), KCL22

      • Glioblastoma: U2OS

      • Ovarian cancer: Various EOC cell lines

    • Methodological considerations:

      • Compare drug-sensitive and resistant lines (e.g., K562 vs K562R)

      • Include non-malignant controls (e.g., BaF3 vs Bcr-Abl+ BP210 cells)

  • Genetic manipulation approaches:

    • siRNA knockdown: Specific KPNB1 targeting sequences have been validated

    • CRISPR/Cas9 genome editing: For complete knockout studies

    • Overexpression systems: To study gain-of-function effects

    • Inducible systems: To study temporal dynamics of inhibition

  • Pharmacological inhibition:

    • Importazole (IPZ): A selective KPNB1 inhibitor that interferes with Ran-GTP interaction

    • Ivermectin: FDA-approved antiparasitic drug with KPNB1-inhibiting properties

    • Combination approaches: Test with standard chemotherapeutics (e.g., IPZ enhances CML cell sensitivity to imatinib)

  • In vivo models:

    • Human tumor xenografts in mice

    • Pooled shRNA library screening in vivo

    • CRISPR/Cas9 library screening

    • Measurement parameters: Tumor volume, survival time, metastasis formation

  • Clinical correlation studies:

    • Patient sample analysis: Western blotting for KPNB1 expression

    • Correlation with clinical outcomes: Disease progression, treatment response

    • Tissue microarray analysis: For large-scale protein expression studies

Model TypeAdvantagesLimitationsBest Applications
Cell linesEasy manipulation, homogeneousMay not reflect tumor heterogeneityMechanism studies, drug screening
Patient-derived xenograftsMaintain tumor heterogeneityMore resource intensivePreclinical validation
In vivo screensIdentifies targets in physiological contextComplex analysis, expensiveTarget discovery
Human samplesDirect clinical relevanceLimited manipulationBiomarker studies

How does KPNB1 inhibition disrupt cellular proteostasis and trigger the unfolded protein response?

KPNB1 inhibition disrupts cellular proteostasis through several interconnected mechanisms that can be experimentally investigated:

  • Cytosolic protein accumulation:

    • KPNB1 inhibition causes retention of its cargo proteins in the cytoplasm

    • This creates protein imbalance between nuclear and cytoplasmic compartments

    • Methodology: Subcellular fractionation followed by Western blotting to quantify protein distribution

  • Protein overload consequences:

    • Increased polyubiquitination of proteins

    • Formation of aggresome-like-induced structures (ALIS)

    • Activation of unfolded protein response (UPR)

    • Research techniques: Ubiquitination assays, fluorescence microscopy for aggresome detection, RT-PCR and Western blotting for UPR markers

  • UPR activation pathway:

    • PERK activation leads to eIF2α phosphorylation

    • Upregulation of ATF4 and downstream targets

    • Chronic activation of eIF2α/ATF4 cascade upregulates pro-apoptotic proteins Puma and Noxa

    • Methodology: Time-course analysis of UPR markers using Western blotting

  • Experimental validation approaches:

    • Reversal of effects: KPNB1 overexpression or protein synthesis inhibitors reduce ubiquitination elevation and UPR activation

    • Exacerbation of effects: Inhibitors of autophagy-lysosome or proteasome pathways aggravate proteostasis disruption

    • These findings indicate that rebalancing cytosolic/nuclear protein distribution alleviates protein overload

The connection between proteostasis disruption and cell death can be studied by manipulating specific UPR branches (PERK, IRE1, ATF6) to determine which is primarily responsible for apoptosis induction after KPNB1 inhibition.

What experimental approaches can distinguish between direct and indirect effects of KPNB1 on protein homeostasis?

To differentiate between direct and indirect effects of KPNB1 on protein homeostasis, researchers should employ these methodological strategies:

  • Temporal analysis of events:

    • Design time-course experiments with precise sampling points

    • Monitor chronological order of: nuclear transport inhibition → protein accumulation → UPR activation → cell death

    • Methodology: Combine live-cell imaging with fixed-time-point biochemical analyses

  • Cargo-specific manipulation:

    • Generate KPNB1 mutants that selectively transport specific cargo classes

    • Identify which cargo proteins' nuclear exclusion most strongly correlates with proteostasis disruption

    • Approach: Structure-based mutagenesis combined with interactome analysis

  • Proteomics-based approaches:

    • Quantitative proteomics to identify proteins most affected by KPNB1 inhibition

    • Pulse-chase experiments to distinguish effects on new vs. existing proteins

    • SILAC or TMT labeling for precise quantification of proteome changes

    • Methodology: Mass spectrometry following subcellular fractionation

  • Targeted rescue experiments:

    • Express individual KPNB1 cargo proteins with artificial nuclear localization methods

    • Determine if nuclear restoration of specific factors reverses proteostasis defects

    • Alternative nuclear import pathways (e.g., via other karyopherins) can be engineered

  • UPR branch-specific interventions:

    • Selective inhibition of PERK, IRE1, or ATF6 pathways

    • Determine which UPR branch is most critical for KPNB1 inhibition-induced effects

    • Approach: Genetic knockdown or pharmacological inhibitors of specific UPR components

How do importin α-dependent and independent KPNB1 transport mechanisms differentially impact cellular function?

KPNB1 can mediate nuclear import through both importin α-dependent and independent pathways, with distinct functional implications. Research methodologies to investigate these differences include:

  • Structural and functional domain analysis:

    • The importin α binding domain of KPNB1 can be deleted to specifically inhibit importin α-dependent transport

    • Research shows deletion of this domain does not significantly affect transcriptional regulation in circadian clock experiments

    • Methodology: Structure-function analyses using domain deletion or mutation constructs

  • Comparative knockout/knockdown experiments:

    • KPNB1 knockdown severely disrupts circadian rhythmicity and blocks PER2 nuclear localization

    • In contrast, depletion of importin α1 (KPNA2) or importin α5 (KPNA1) does not affect circadian rhythmicity or PER2 nuclear localization

    • This demonstrates differential requirements for these pathways in specific cellular processes

    • Approach: Parallel knockdown experiments with phenotypic analysis

  • Cargo-specific studies:

    • Different cargo proteins utilize distinct transport mechanisms:

    • PER/CRY complexes appear to utilize importin α-independent pathways

    • Other proteins (e.g., certain transcription factors) require the classical importin α-dependent pathway

    • Methodology: Cargo-specific binding assays with wild-type vs. mutant importins

  • Transport kinetics analysis:

    • Temporal dynamics of nuclear import may differ between pathways

    • KPNB1 exhibits circadian patterns of nucleocytoplasmic localization correlating with nuclear accumulation of specific cargoes

    • Research technique: Time-resolved imaging or biochemical fractionation studies

This dichotomy suggests specialized roles for different transport mechanisms, with importin α-independent transport being particularly important for circadian regulation.

What are the emerging techniques for studying KPNB1-mediated nuclear transport in living cells with minimal perturbation?

Advanced researchers studying KPNB1 transport dynamics are employing increasingly sophisticated techniques that minimize experimental artifacts:

  • Live-cell imaging with optogenetic tools:

    • Photoswitchable KPNB1 variants that can be activated in specific cellular regions

    • Light-inducible cargo-KPNB1 interactions to trigger transport on demand

    • Reversible inhibition of KPNB1 function using photocaged inhibitors

    • Methodology: Combine optogenetic constructs with high-resolution confocal or light-sheet microscopy

  • Single-molecule tracking:

    • Quantum dot or HaloTag labeling of individual KPNB1 molecules

    • Direct visualization of KPNB1 movement through nuclear pores

    • Measurement of dwell times, transport rates, and interaction kinetics

    • Technique: Super-resolution microscopy (PALM/STORM) with fast acquisition rates

  • Genome editing with minimal tags:

    • CRISPR-mediated insertion of small epitope tags or fluorescent proteins at endogenous loci

    • Maintains native expression levels and regulatory elements

    • Avoids overexpression artifacts common in traditional approaches

    • Methodology: Homology-directed repair with donor templates containing minimal tags

  • Proximity labeling techniques:

    • BioID or TurboID fused to KPNB1 to identify transient interaction partners

    • APEX2-based approaches for temporal control of labeling

    • Spatial mapping of KPNB1 interactome at the nuclear pore

    • Approach: Mass spectrometry following proximity labeling

  • Correlative light and electron microscopy (CLEM):

    • Combines functional imaging of KPNB1 transport with ultrastructural context

    • Allows visualization of transport events in relation to nuclear pore complex structure

    • Methodology: Fluorescence imaging followed by sample preparation for electron microscopy

These approaches provide unprecedented spatial and temporal resolution for studying KPNB1 dynamics while maintaining physiological conditions.

How does KPNB1 function differ between normal and disease states in human tissues?

KPNB1 exhibits significant functional differences between normal and disease states, particularly in cancer. These differences can be systematically investigated through:

  • Expression level analysis:

    • KPNB1 is overexpressed in multiple cancer types:

      • Chronic myeloid leukemia (CML)

      • Glioblastoma

      • Epithelial ovarian cancer (EOC)

      • Cervical cancer

    • Methodological approach: Western blotting, immunohistochemistry, and qRT-PCR comparing patient samples with normal controls

  • Subcellular localization patterns:

    • Altered distribution between nuclear and cytoplasmic compartments in cancer cells

    • Different cargo selection or transport dynamics

    • Technique: Subcellular fractionation followed by Western blotting or immunofluorescence microscopy

  • Interaction network changes:

    • Disease-specific protein-protein interactions

    • In CML, KPNB1 shows enhanced interaction with E2F1 and affects downstream targets like c-Myc

    • Research approach: Co-immunoprecipitation followed by mass spectrometry to map interactomes in normal vs. disease states

  • Dependency patterns:

    • Cancer cells show increased dependency on KPNB1 function

    • KPNB1 inhibition selectively affects cancer cells while sparing normal cells

    • Methodology: Comparative viability assays after KPNB1 knockdown or inhibition

  • Response to therapy:

    • KPNB1 inhibition can enhance sensitivity to existing therapies

    • IPZ enhances CML cell sensitivity to imatinib

    • Combination with Bcl-xL inhibitors enhances apoptosis in glioblastoma cells

    • Approach: Drug combination studies with dose-response matrices

These differences suggest that KPNB1 could be targeted therapeutically with potentially favorable therapeutic windows between normal and disease states.

What are the most reliable biomarkers for KPNB1 activity in human tissue samples?

For researchers working with clinical samples, reliable assessment of KPNB1 activity (not just expression) requires multi-parameter analysis:

  • Direct KPNB1 measurements:

    • Protein expression level: Immunohistochemistry, Western blotting

    • Phosphorylation status: Phospho-specific antibodies

    • Subcellular distribution: Nuclear/cytoplasmic ratio determination

    • Methodological considerations: Use validated antibodies with proper controls

  • Cargo localization patterns:

    • Nuclear/cytoplasmic distribution of known KPNB1 cargoes:

      • E2F1 (important in cancer contexts)

      • PER/CRY proteins (for circadian studies)

      • Other transcription factors (c-Myc, NF-κB)

    • Research technique: Multiplexed immunofluorescence or immunohistochemistry

  • Downstream transcriptional outputs:

    • Expression of genes regulated by KPNB1-transported transcription factors:

      • E-box-dependent genes (PER1, CRY1, DBP, REV-ERBβ) for circadian studies

      • E2F1 target genes for cancer studies

    • Methodology: RT-qPCR panels or RNA-seq analysis

  • UPR activation markers:

    • Phosphorylated PERK and eIF2α

    • ATF4 nuclear localization

    • CHOP expression

    • These markers indicate proteostasis disruption that may be linked to KPNB1 dysfunction

    • Approach: Multi-parameter immunohistochemistry or Western blotting

  • Correlation analysis framework:

    • Create tissue microarrays with multiple samples

    • Perform parallel staining for all markers

    • Conduct statistical correlation analyses

    • Establish a "KPNB1 activity signature" from combined parameters

Biomarker TypeExamplesDetection MethodSignificance
Direct KPNB1KPNB1 protein, phospho-KPNB1IHC, WBPrimary indicator
Cargo localizationNuclear E2F1, PER2IF, IHCFunctional output
TranscriptionalE2F1 target genesRT-qPCR, RNA-seqDownstream effects
UPR markersp-PERK, p-eIF2α, ATF4IHC, WBAssociated pathway
Clinical correlationSurvival, therapy responseStatistical analysisClinical relevance

What are the emerging technologies that will advance our understanding of KPNB1 biology?

Several cutting-edge technologies are poised to revolutionize KPNB1 research:

  • Cryo-electron tomography:

    • Visualization of KPNB1-cargo complexes in their native cellular environment

    • Resolution of structural intermediates during nuclear pore translocation

    • Methodological approach: Sample vitrification followed by tomographic imaging

  • Single-cell multi-omics:

    • Integrated analysis of KPNB1-associated transcriptome, proteome, and epigenome

    • Revelation of cell-to-cell variability in KPNB1 function

    • Identification of rare cell populations with altered KPNB1 activity

    • Technique: Combined single-cell RNA-seq, proteomics, and ATAC-seq

  • Protein phase separation studies:

    • Investigation of KPNB1's potential role in liquid-liquid phase separation

    • Exploration of how this affects nuclear transport regulation

    • Connection to stress granule formation and proteostasis

    • Methodology: Fluorescence microscopy with optogenetic tools to induce phase separation

  • CRISPR-based genetic screens:

    • Genome-wide identification of KPNB1 modifiers

    • Synthetic lethal interactions with KPNB1 inhibition

    • Cargo-specific transport pathways

    • Approach: CRISPR-Cas9 libraries with single-cell readouts

  • Targeted protein degradation approaches:

    • Development of KPNB1-specific PROTAC degraders

    • More complete protein elimination compared to inhibitors

    • Potential therapeutic applications

    • Research technique: Design of bifunctional molecules targeting KPNB1 for ubiquitin-mediated degradation

These technologies will help resolve current contradictions in the field and open new avenues for therapeutic intervention.

What are the critical unresolved questions about KPNB1 that require further investigation?

Despite significant progress in KPNB1 research, several fundamental questions remain unresolved:

  • Cargo specificity mechanisms:

    • How does KPNB1 select its cargo proteins with such specificity?

    • What determines importin α-dependent versus independent transport?

    • Why are certain cellular processes (e.g., circadian rhythm) particularly sensitive to KPNB1 dysfunction?

    • Research approach: Systematic mutagenesis combined with transport assays

  • Regulatory mechanisms:

    • How is KPNB1 expression and activity regulated under different physiological conditions?

    • What post-translational modifications affect KPNB1 function?

    • Is there circadian regulation of KPNB1 itself, or just its cargo distribution?

    • Methodology: Temporal proteomics and phosphoproteomics analysis

  • Evolutionary considerations:

    • Why is the KPNB1-interacting domain of RTEL1 divergent between mouse and human?

    • Does this suggest recently acquired, species-specific functions?

    • How conserved are KPNB1-mediated processes across species?

    • Approach: Comparative genomics and functional conservation studies

  • Therapeutic targeting challenges:

    • How can we achieve selective inhibition of disease-specific KPNB1 functions?

    • Is it possible to target specific cargo transport without affecting others?

    • What determines the therapeutic window between normal and cancer cells?

    • Research technique: Structure-based drug design of next-generation KPNB1 inhibitors

  • Integration with other cellular systems:

    • How does KPNB1 function coordinate with other nuclear transport pathways?

    • What is the relationship between KPNB1 and the nuclear pore complex during stress?

    • How does KPNB1 contribute to phase-dependent cellular processes beyond the cell cycle?

    • Methodology: Systems biology approaches combining multiple 'omics datasets

Product Science Overview

Structure and Function

KPNB1 contains an importin N-terminal domain and 19 HEAT repeats, which are structural motifs involved in protein-protein interactions . The primary function of KPNB1 is to mediate the transport of proteins with a nuclear localization signal (NLS) from the cytoplasm into the nucleus. This process is essential for various cellular functions, including gene expression, DNA replication, and cell cycle regulation.

Mechanism of Action

The transport mechanism involves the recognition of cargo proteins by KPNB1 in the cytoplasm. KPNB1 forms a complex with importin alpha, which directly binds to the NLS of the cargo protein. This complex is then transported through the nuclear pore complex (NPC) into the nucleus. Once inside the nucleus, the complex dissociates, releasing the cargo protein to perform its nuclear functions .

Evolutionary Significance

The Karyopherin-β family, to which KPNB1 belongs, has ancient origins and has continued to specialize throughout eukaryotic evolution. The family is highly conserved across different eukaryotic species, indicating its fundamental role in cellular processes. Studies have shown that at least fifteen Karyopherin-β subfamilies were established early in eukaryote evolution, highlighting the importance of nucleocytoplasmic transport mechanisms .

Recombinant KPNB1

Recombinant human KPNB1 is produced using recombinant DNA technology, which involves inserting the gene encoding KPNB1 into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of KPNB1 for research and therapeutic purposes. Recombinant KPNB1 is typically supplied in a solution containing Tris-HCl buffer, DTT, glycerol, and NaCl to maintain its stability and activity .

Applications

Recombinant KPNB1 is widely used in research to study nucleocytoplasmic transport mechanisms, protein-protein interactions, and the regulation of nuclear import. It is also used in drug discovery and development to identify potential therapeutic targets for diseases related to nuclear transport dysfunction.

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.