NPM1 Human

Nucleophosmin Human Recombinant
Shipped with Ice Packs
In Stock

Description

NPM1 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 314 amino acids (1-294 a.a) and having a molecular mass of 34.7kDa.
NPM1 is fused to a 20 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Nucleophosmin (NPM1) is a phosphoprotein that moves between the nucleus and the cytoplasm of a cell. It is believed to be involved in several cellular processes, including the regulation of the ARF/p53 pathway. NPM1 is known to interact with other genes, forming fusion partners, notably with the anaplastic lymphoma kinase gene found on chromosome 2. Mutations in the NPM1 gene are associated with acute myeloid leukemia.
Description
Recombinant human NPM1, produced in E. coli bacteria, is a single polypeptide chain that lacks glycosylation. It consists of 314 amino acids (specifically, amino acids 1 to 294), resulting in a molecular weight of 34.7 kDa. The NPM1 protein has a 20 amino acid His-tag attached to its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear and sterile solution.
Formulation
The NPM1 protein is provided in a solution with a concentration of 0.5 mg/ml. The solution contains 20 mM Tris-HCl buffer at a pH of 8.0, 10% glycerol, 0.15 M NaCl (sodium chloride), and 1 mM DTT (dithiothreitol).
Stability
For short-term storage (up to 2-4 weeks), the NPM1 protein solution should be kept at 4°C. For longer storage, it is recommended to freeze the solution at -20°C. To further enhance long-term stability, consider adding a carrier protein such as HSA or BSA (0.1% concentration). It is important to avoid repeated freezing and thawing of the protein solution.
Purity
The purity of the NPM1 protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
Nucleophosmin, NPM, Nucleolar phosphoprotein B23, Nucleolar protein NO38, Numatrin, NPM1, NPM, B23.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MEDSMDMDMS PLRPQNYLFG CELKADKDYH FKVDNDENEH QLSLRTVSLG AGAKDELHIV EAEAMNYEGS PIKVTLATLK MSVQPTVSLG GFEITPPVVL RLKCGSGPVH ISGQHLVAVE EDAESEDEEE EDVKLLSISG KRSAPGGGSK VPQKKVKLAA
DEDDDDDDEE DDDEDDDDDD FDDEEAEEKA PVKKSIRDTP AKNAQKSNQN GKDSKPSSTP RSKGQESFKK QEKTPKTPKG PSSVEDIKAK MQASIEKGGS LPKVEAKFIN YVKNCFRMTD QEAIQDLWQW RKSL.

Q&A

What is NPM1 and what are its primary cellular functions?

NPM1 (nucleophosmin) is a multifunctional protein with prominent nucleolar localization that shuttles between the nucleus and cytoplasm. It plays essential roles in several cellular processes, including:

  • Ribosome biogenesis and transport

  • Centrosome duplication regulation

  • Protein chaperoning

  • DNA repair mechanisms

  • Apoptotic response regulation

  • Transcriptional regulation through interaction with chromatin

The protein exhibits dynamic subcellular trafficking, with its proper nucleolar-nuclear-cytoplasmic distribution being critical for normal cellular function. NPM1 functions as an oligomer (pentamer/decamer) in physiological conditions, which is essential for proper nucleolus formation and maintenance of genomic stability .

What is the structural organization of the NPM1 protein?

The NPM1 protein is organized into distinct functional domains:

DomainLocationPrimary Function
N-terminal Oligomerization DomainResidues 1-120Mediates pentamer/decamer formation
Central Acidic RegionResidues 120-186Histone binding and chaperone activity
C-terminal Nucleic Acid Binding DomainResidues 187-294RNA binding and nucleolar localization
Nuclear Export Signal (NES)Located within proteinMediates cytoplasmic export
Nuclear Localization Signal (NLS)Located within proteinDirects nuclear import
Nucleolar Localization Signal (NoLS)C-terminal regionDetermines nucleolar retention

The C-terminal domain contains tryptophan residues critical for nucleolar localization. Mutations that disrupt these tryptophan residues or introduce additional nuclear export signals lead to aberrant cytoplasmic localization of the protein, which is a hallmark of NPM1-mutated acute myeloid leukemia .

How does NPM1 interact with other cellular proteins?

NPM1 functions as a hub protein, interacting with numerous binding partners to coordinate various cellular processes. Key interactions include:

  • Transcriptional regulators: BRD4, PU.1, CTCF, MIZ1, and others

  • Tumor suppressors: ARF, FBW7

  • Chromatin modifiers: MLL1-menin complex

  • Nuclear transport receptors: XPO1/CRM1

  • DNA repair proteins: APE1

  • Apoptotic regulators: Caspases 6 and 8, HEXIM1

These interactions are often regulated by post-translational modifications of NPM1, including phosphorylation, SUMOylation, and ubiquitination. In research contexts, co-immunoprecipitation followed by mass spectrometry is the methodological approach of choice to identify novel NPM1 interacting partners in different cellular contexts .

What types of NPM1 mutations are observed in acute myeloid leukemia?

NPM1 mutations in AML typically involve small insertions in exon 12 that result in a frameshift altering the C-terminal region of the protein. The most common mutations are:

Mutation TypeFrequencyMolecular Consequence
Type A (TCTG insertion)~75-80% of casesFrameshift creating NES motif
Type B (CATG insertion)~10% of casesFrameshift creating NES motif
Type D (CCTG insertion)~5% of casesFrameshift creating NES motif
Other variants~5-10% of casesVarious frameshifts, all disrupting NoLS

All these mutations lead to the common outcome of creating an additional nuclear export signal (NES) while disrupting the nucleolar localization signal (NoLS), resulting in aberrant cytoplasmic localization of the mutant protein. This cytoplasmic mislocalization is considered the pathogenic mechanism driving leukemogenesis .

How do NPM1 mutations contribute to leukemic transformation?

NPM1 mutations contribute to leukemic transformation through multiple mechanisms:

  • HOX gene dysregulation: NPM1 mutants maintain aberrant expression of HOXA and HOXB gene clusters, which are normally silenced during myeloid differentiation. This persistent HOX expression promotes self-renewal and blocks differentiation of hematopoietic progenitors .

  • Disruption of tumor suppressor pathways: Cytoplasmic delocalization of NPM1 results in mislocalization of key tumor suppressors including ARF, leading to impaired p53 responses .

  • Altered transcriptional regulation: NPM1 mutants disrupt normal functions of transcription factors like PU.1 and CTCF by mislocalizing them to the cytoplasm .

  • Deregulation of BRD4-dependent transcription: Mutant NPM1 alters the inhibitory effect of wild-type NPM1 on BRD4, leading to increased transcription of oncogenes like BCL2 and MYC .

  • Perturbation of the hematopoietic niche: Expression of CXCR4/CXCL12-related genes is significantly suppressed in NPM1-mutant cells, potentially disrupting normal hematopoietic stem cell interactions with the bone marrow microenvironment .

Experimental evidence suggests that NPM1 mutation alone is insufficient for leukemogenesis and requires cooperation with other genetic alterations to induce full-blown AML .

What is the significance of NPM1 haploinsufficiency in leukemia development?

The significance of NPM1 haploinsufficiency in leukemia development is a complex research question:

NPM1 mutations are always heterozygous, with one wild-type allele retained. Complete loss of NPM1 is embryonically lethal, indicating that at least one functional copy is essential for cell survival. While NPM1 heterozygous knockout mice develop myelodysplastic-like disorders due to centrosome duplication abnormalities and aneuploidy, this mechanism appears distinct from human NPM1-mutated AML .

In human NPM1-mutated AML, haploinsufficiency of wild-type NPM1 may contribute to leukemogenesis through:

  • Reduced nucleolar stress sensing capacity

  • Altered ribosome biogenesis

  • Compromised DNA damage response

Research methodology to study haploinsufficiency effects typically involves comparison of heterozygous knockout models with NPM1-mutant knock-in models to distinguish between loss-of-function and gain-of-function effects.

What are the recommended methodologies for detecting NPM1 mutations in research and clinical settings?

Several complementary methodologies are available for detecting NPM1 mutations with varying sensitivity, specificity, and applications:

MethodSensitivityAdvantagesLimitationsApplications
ImmunohistochemistryModerate- Rapid screening
- Detection of cytoplasmic NPM1
- Applicable to fixed tissue
- Cannot identify specific mutation
- False negatives possible
Initial screening
PCR and fragment analysisHigh (1-5%)- Identification of common mutations
- Quantitative assessment possible
- Limited to known mutations
- Primer design critical
Diagnosis and MRD
Sanger sequencingModerate (15-20%)- Detection of all mutation types
- Gold standard for mutation confirmation
- Limited sensitivity
- Labor intensive
Mutation confirmation
Next-generation sequencingVery high (0.1-1%)- Comprehensive mutation profiling
- Detection of co-occurring mutations
- Cost
- Complex data analysis
Research and comprehensive diagnostics
Digital PCRExtremely high (0.01%)- Ultrasensitive detection
- Absolute quantification
- Limited to known mutations
- Specialized equipment needed
MRD monitoring
RNA-based assaysHigh- Detection of variant transcripts
- Functional validation
- RNA quality dependent
- Post-transcriptional modifications
Research

For research purposes, the methodology selection should be guided by the specific research question. When studying clonal evolution or minimal residual disease, highly sensitive methods like digital PCR or targeted NGS are recommended. For functional characterization, RNA-based methods combined with protein localization studies provide comprehensive insights .

How can researchers effectively model NPM1 mutations in experimental systems?

Researchers have developed several experimental systems to model NPM1 mutations, each with specific advantages and limitations:

  • Cell line models:

    • Retroviral/lentiviral transduction of NPM1 mutant into hematopoietic cell lines

    • CRISPR/Cas9 knock-in of NPM1 mutations in appropriate cell backgrounds

    • Inducible expression systems allowing temporal control of mutant expression

    Methodological consideration: Cell lines often harbor additional mutations that may influence experimental outcomes. Researchers should validate findings across multiple cell backgrounds.

  • Primary cell models:

    • Ex vivo transduction of primary hematopoietic stem/progenitor cells

    • Patient-derived xenografts (PDX) from NPM1-mutated AML cases

    Methodological consideration: Primary cells have limited proliferative capacity in vitro. Co-culturing with stromal cells or supplementation with specific cytokines may be required to maintain viability.

  • Mouse models:

    • Conventional knock-in models (e.g., insertion of TCTG after nucleotide c.857)

    • Conditional knock-in models (allowing tissue-specific or temporal control)

    • Humanized mouse models (with human NPM1 sequence)

    Methodological consideration: Homozygous NPM1 mutant mice show embryonic lethality, while heterozygous mice develop myeloproliferative disease but not frank leukemia, suggesting that additional genetic events are required for leukemogenesis .

  • iPSC models:

    • CRISPR/Cas9 engineering of NPM1 mutations in induced pluripotent stem cells

    • Directed differentiation toward hematopoietic lineages

    Methodological consideration: iPSC models allow developmental studies but may not fully recapitulate the adult hematopoietic microenvironment.

When selecting a model system, researchers should consider the compatibility with their specific research question. Combination of multiple model systems often provides the most robust evidence for functional hypotheses .

What techniques are recommended for studying NPM1 protein localization and trafficking?

The study of NPM1 protein localization and trafficking is critical for understanding its function in normal and pathological states. Recommended techniques include:

  • Immunofluorescence microscopy:

    • Fixed-cell immunofluorescence with NPM1-specific antibodies

    • Co-staining with nucleolar markers (fibrillarin, nucleolin)

    • Super-resolution microscopy for detailed subnuclear localization

    Methodological consideration: Antibody selection is critical; use antibodies that can distinguish between wild-type and mutant NPM1.

  • Live-cell imaging:

    • Fluorescent protein tagging (GFP/RFP-NPM1 fusions)

    • Photobleaching techniques (FRAP, FLIP) to measure shuttling dynamics

    • Optogenetic approaches for controlled relocalization

    Methodological consideration: Tags may interfere with NPM1 localization; verify that fusion proteins maintain expected localization patterns.

  • Biochemical fractionation:

    • Subcellular fractionation followed by Western blotting

    • Sequential extraction protocols to isolate nucleolar, nucleoplasmic, and cytoplasmic fractions

    Methodological consideration: Maintain sample integrity during fractionation; cross-validate with microscopy techniques.

  • Proximity labeling:

    • BioID or APEX2 fusion proteins to identify proximal interactors in different compartments

    • Spatially-resolved interactome mapping

    Methodological consideration: Expression levels of fusion proteins should be near endogenous to avoid artifacts.

  • Transport assays:

    • In vitro nuclear import/export assays with recombinant proteins

    • Microinjection of labeled proteins to track transport kinetics

    • Inhibitor studies targeting specific transport pathways (e.g., XPO1/CRM1 inhibition)

    Methodological consideration: Transport dynamics may differ significantly between in vitro systems and intact cells.

For comprehensive analysis, researchers should combine multiple techniques and validate findings across different cell types and conditions .

How does NPM1 contribute to normal hematopoiesis?

NPM1 plays critical roles in normal hematopoiesis through multiple mechanisms:

  • Regulation of hematopoietic stem cell (HSC) maintenance:

    • NPM1 regulates the nucleolar stress response, which is essential for HSC quiescence

    • It contributes to genomic stability through proper centrosome duplication control

    • NPM1 is involved in ribosome biogenesis, which is tightly regulated in HSCs

  • Control of differentiation programs:

    • NPM1 interacts with key transcription factors involved in myeloid differentiation, including PU.1

    • It regulates HOX gene expression patterns, which are dynamically modulated during hematopoietic differentiation

    • NPM1 affects chromatin organization through interaction with histones and chromatin modifiers

  • Regulation of hematopoietic niche interactions:

    • NPM1 influences the expression of CXCR4/CXCL12-related genes, which are crucial for HSC homing and retention in the bone marrow niche

    • It affects cellular responses to cytokines and growth factors that regulate hematopoiesis

Research methodologies to study NPM1 in normal hematopoiesis include:

  • Colony-forming assays to assess progenitor function

  • Competitive transplantation experiments to evaluate HSC function

  • Flow cytometry analysis of hematopoietic stem and progenitor cell populations

  • Cobblestone area-forming cell (CAFC) assays to assess interaction with the hematopoietic microenvironment

Studies in NPM1 heterozygous knockout mice have demonstrated that reduced NPM1 levels lead to myelodysplastic-like features, indicating that proper NPM1 dosage is essential for normal hematopoiesis .

What cooperative genetic events interact with NPM1 mutations in leukemia progression?

NPM1 mutations rarely exist in isolation in AML and frequently co-occur with other genetic alterations that cooperate in leukemia progression:

Cooperating MutationFrequency with NPM1Prognostic ImpactMechanistic Interaction
DNMT3A mutations40-60%AdverseEpigenetic dysregulation; affects DNA methylation patterns
FLT3-ITD35-40%AdverseConstitutive signaling activation; synergizes with NPM1 to promote proliferation
IDH1/2 mutations20-30%VariableMetabolic rewiring; production of oncometabolite 2-HG; epigenetic alterations
NRAS/KRAS mutations10-15%NeutralRAS pathway activation; promotes cell proliferation
TET2 mutations10-15%VariableImpaired DNA hydroxymethylation; epigenetic dysregulation
PTPN11 mutations5-10%NeutralEnhanced MAPK signaling; promotes proliferation
WT1 mutations5-10%AdverseTranscriptional dysregulation; impaired differentiation
Cohesin complex mutations5-10%VariableAltered chromatin architecture; dysregulated gene expression

These cooperative events can be studied through several methodological approaches:

  • Comprehensive genomic profiling:

    • Targeted sequencing panels for recurrently mutated genes

    • Whole exome/genome sequencing to identify novel cooperating mutations

    • RNA-sequencing to identify fusion genes and expression patterns

  • Functional validation:

    • Serial introduction of mutations in cellular models

    • Combinatorial CRISPR/Cas9 genome editing

    • Compound transgenic/knock-in mouse models

  • Clonal evolution analysis:

    • Single-cell DNA sequencing to track mutation acquisition

    • Variant allele frequency analysis to infer clonal architecture

    • Longitudinal sampling to track clonal dynamics during disease progression

Understanding these cooperative interactions is essential for designing rational therapeutic strategies targeting both NPM1 mutations and their cooperating events .

How does NPM1 mutation status influence leukemic stem cell biology?

NPM1 mutation significantly influences leukemic stem cell (LSC) biology through several mechanisms:

  • Altered stem cell phenotype:

    • NPM1-mutated AML LSCs often lack the typical CD34+ phenotype of many other AML subtypes

    • LSC activity in NPM1-mutated AML may reside in more differentiated compartments (CD34-/CD38+)

    • The immunophenotypic profile of NPM1-mutated LSCs can be heterogeneous and evolve during disease progression

  • HOX-dependent self-renewal:

    • NPM1 mutations maintain aberrant expression of HOXA and HOXB clusters

    • This persistent HOX expression provides self-renewal capacity to leukemic progenitors

    • The HOX program is maintained by the MLL1-menin complex and directly depends on NPM1 mutants

  • Altered niche interactions:

    • NPM1-mutated LSCs show significant downregulation of CXCR4/CXCL12-related genes

    • This leads to abnormal homing and retention in the bone marrow microenvironment

    • Compromised cobblestone area formation in experimental models suggests pathology in the hematopoietic niche

  • Epigenetic reprogramming:

    • NPM1 mutations are associated with distinctive DNA methylation and histone modification patterns

    • These epigenetic changes contribute to the block in differentiation and enhanced self-renewal

    • Co-occurring mutations in epigenetic regulators (DNMT3A, IDH1/2, TET2) further modulate the epigenetic landscape

Research methodologies to study NPM1-mutated LSCs include:

  • Xenotransplantation assays using limiting dilution approaches

  • Flow cytometry sorting of distinct cellular compartments followed by functional assays

  • In vitro long-term culture-initiating cell (LTC-IC) assays

  • Single-cell RNA sequencing to identify LSC-specific gene expression signatures

  • Epigenetic profiling (ATAC-seq, ChIP-seq, DNA methylation arrays)

Understanding the unique biology of NPM1-mutated LSCs has important implications for developing targeted therapeutic approaches and monitoring minimal residual disease.

How can NPM1 mutation be targeted therapeutically?

Several therapeutic approaches targeting NPM1 mutations are being investigated:

  • Targeting nuclear export:

    • XPO1/CRM1 inhibitors (selinexor, KPT-8602) block nuclear export of NPM1 mutants

    • This approach restores nuclear localization of NPM1 mutants and their binding partners

    • Preclinical studies show differentiation and growth arrest of NPM1-mutated cells

    Methodological consideration: XPO1 inhibition affects multiple cellular proteins beyond NPM1; specific attribution of effects requires careful experimental design.

  • Targeting HOX-dependent pathways:

    • Menin-MLL inhibitors (MI-463, MI-503, VTP-50469) disrupt the menin-MLL interaction

    • This leads to downregulation of HOXA/HOXB genes and differentiation of NPM1-mutated cells

    • Combination with standard chemotherapy shows synergistic effects in preclinical models

    Methodological consideration: HOX dependency may vary between patients; biomarkers predicting response should be investigated.

  • Nucleolar stress induction:

    • Actinomycin D and other inhibitors of RNA polymerase I induce nucleolar stress

    • NPM1-mutated cells with nucleolar depletion of wild-type NPM1 may be particularly vulnerable

    • Low-dose regimens that selectively affect nucleolar function while minimizing general cytotoxicity are being investigated

    Methodological consideration: Therapeutic window may be narrow; dose-finding studies are critical.

  • Immunotherapeutic approaches:

    • The unique C-terminal sequences of NPM1 mutants can be recognized by the immune system

    • Peptide vaccines or adoptive T-cell therapies targeting these neoantigens are in development

    • Engineered T cells with T-cell receptors specific for NPM1 mutant peptides show promise in preclinical studies

    Methodological consideration: HLA restriction limits applicability; strategies to overcome HLA barriers are needed.

  • Combination approaches:

    • Targeting cooperating mutations (FLT3, IDH1/2) alongside NPM1

    • Epigenetic modifiers to reverse aberrant gene expression programs

    • Bcl-2 inhibitors (venetoclax) show particular efficacy in NPM1-mutated AML

    Methodological consideration: Rational combinations should be based on mechanistic understanding of pathway interactions .

What is the clinical significance of NPM1 mutation as a biomarker for minimal residual disease monitoring?

NPM1 mutation serves as an excellent biomarker for minimal residual disease (MRD) monitoring:

  • Stability and specificity:

    • NPM1 mutations are stable during disease course, with rare exceptions

    • They are highly specific for the leukemic clone

    • NPM1 mutations occur early in leukemogenesis and are present in virtually all leukemic cells

  • Sensitivity of detection methods:

    • Quantitative PCR (qPCR): Can detect 1 mutated cell among 10^4-10^5 normal cells

    • Digital PCR (dPCR): Can reach sensitivity of 1 in 10^6

    • Next-generation sequencing (NGS): Variable sensitivity depending on depth (typically 1 in 10^3-10^4)

  • Prognostic value:

    • MRD positivity after induction therapy predicts increased relapse risk

    • MRD positivity before allogeneic stem cell transplantation predicts worse outcomes

    • Rising MRD levels during follow-up can predict morphological relapse by 2-3 months

  • Methodological considerations for research and clinical implementation:

    • Standardization of sample collection, processing, and analysis is critical

    • Multiple timepoints should be assessed to capture MRD dynamics

    • Peripheral blood may be used for frequent monitoring, though bone marrow provides higher sensitivity

    • Concurrent assessment of common co-mutations (FLT3-ITD, DNMT3A) provides context for interpretation

  • Limitations and challenges:

    • Technical variations between laboratories necessitate standardization

    • Different mutation types require type-specific assays

    • Potential for false negatives due to sampling issues

    • Interpreting low-level positivity requires clinical context

Research applications of NPM1-based MRD include:

  • Evaluation of novel therapeutic agents on the molecular level

  • Investigation of clonal evolution and therapy resistance mechanisms

  • Development of intervention strategies based on molecular rather than morphological relapse .

How can researchers design experimental interventions targeting NPM1-dependent pathways?

Designing experimental interventions targeting NPM1-dependent pathways requires systematic approaches:

  • Target identification strategies:

    • Synthetic lethality screens to identify vulnerabilities specific to NPM1-mutated cells

    • CRISPR/Cas9 library screening (genome-wide or focused libraries)

    • Protein-protein interaction mapping to identify critical nodes in NPM1 mutant networks

    • Computational approaches integrating multi-omics data to predict therapeutic targets

  • Validation approaches:

    • Genetic validation: CRISPR/RNAi-mediated knockdown/knockout of candidate targets

    • Pharmacological validation: Testing available inhibitors of candidate pathways

    • Rescue experiments: Restoring wild-type NPM1 function to confirm specificity

    • Ex vivo drug sensitivity testing using primary patient samples

  • Candidate pathway targeting:

    • HOX-dependent pathways:

      • Develop or repurpose inhibitors of the HOX transcriptional machinery

      • Target HOX cofactors (PBX, MEIS) or downstream effectors

      • Exploit vulnerabilities created by HOX overexpression

    • Nucleolar stress pathways:

      • Screen compounds inducing selective nucleolar stress

      • Target ribosome biogenesis steps that may be particularly vulnerable in NPM1-mutated cells

      • Exploit the nucleolar stress surveillance pathway

    • Nuclear transport machinery:

      • Develop selective inhibitors of nuclear export that preferentially affect NPM1 mutants

      • Target nuclear import pathways to restore normal protein distribution

      • Design peptide inhibitors competing with abnormal nuclear export signals

  • Combinatorial approaches:

    • Design rational combinations based on pathway analysis

    • Test synergy with standard chemotherapeutic agents

    • Explore synthetic lethal interactions with common co-occurring mutations

  • Model systems for intervention testing:

    • Primary patient samples in ex vivo culture systems

    • PDX models to assess efficacy and toxicity in vivo

    • Genetically engineered mouse models with human-relevant mutations

    • Isogenic cell line pairs differing only in NPM1 mutation status

For comprehensive evaluation, interventions should be tested across multiple model systems with appropriate controls to distinguish NPM1-specific effects from general antileukemic activity .

How do single-nucleotide polymorphisms in NPM1 affect protein function and disease risk?

Recent research has identified several single-nucleotide polymorphisms (SNPs) in the NPM1 gene that may impact protein function and disease susceptibility:

  • Functional impact of NPM1 SNPs:
    Bioinformatic analysis has identified several potentially damaging missense SNPs in NPM1:

    SNPProtein ChangePredicted EffectAssociated Structural Change
    K54NLysine to AsparagineDisruption of oligomerizationAltered N-terminal domain stability
    I59TIsoleucine to ThreonineAffects hydrophobic coreDisruption of oligomerization interface
    L79SLeucine to SerineDisruption of protein foldingIntroduction of polar residue in hydrophobic core
    P152AProline to AlanineAltered protein flexibilityChanged structural rigidity
    K193R/NLysine to Arginine/AsparagineAltered nucleic acid bindingModified C-terminal domain function
    A283GAlanine to GlycineIncreased backbone flexibilityDestabilization of C-terminal domain
    I284FIsoleucine to PhenylalanineAltered aromatic interactionsDisruption of nucleolar localization signal

    These SNPs can affect critical functions including oligomerization, nucleic acid binding, subcellular localization, and protein-protein interactions .

  • Methodological approaches to study NPM1 SNPs:

    • Structure-function analysis using recombinant proteins

    • Cell-based assays to assess localization, oligomerization, and interaction patterns

    • In silico molecular dynamics simulations to predict structural consequences

    • Population-based association studies to identify disease correlations

    • Knock-in mouse models with specific SNPs to assess organismal effects

  • Disease associations:

    • Germline SNPs in NPM1 may contribute to susceptibility to hematologic malignancies

    • Certain SNPs might modify the penetrance or expressivity of somatic NPM1 mutations

    • NPM1 polymorphisms could influence response to therapy or disease progression

  • Research challenges:

    • Distinguishing causal SNPs from passenger variants requires robust functional validation

    • Low-frequency SNPs may have significant functional impact but require large sample sizes for detection

    • Contextual effects of genetic background can modify SNP impact

Emerging research suggests that NPM1 SNPs may represent an underexplored source of functional variation relevant to hematopoietic development and malignancy .

What are the implications of NPM1 mutations for clonal hematopoiesis and leukemia prevention?

NPM1 mutations have important implications for understanding clonal hematopoiesis and developing leukemia prevention strategies:

  • NPM1 in clonal hematopoiesis:

    • Unlike mutations in DNMT3A, TET2, and ASXL1, NPM1 mutations are rarely found in age-related clonal hematopoiesis of indeterminate potential (CHIP)

    • When NPM1 mutations are detected in apparently healthy individuals, they often represent early subclinical AML rather than benign CHIP

    • This suggests that NPM1 mutations may have stronger leukemogenic potential than common CHIP-associated mutations

  • Preleukemic clonal architecture:

    • In NPM1-mutated AML, mutations in epigenetic regulators (DNMT3A, TET2, IDH1/2) often precede NPM1 mutations

    • These pre-NPM1 mutations can persist after therapy, creating a reservoir for potential relapse

    • The persistence of preleukemic clones may predispose to second AML with the same or different driver mutations

  • Monitoring and intervention opportunities:

    • Early detection of NPM1 mutations in individuals with CHIP may identify those at imminent risk for AML

    • Serial molecular monitoring of individuals with high-risk CHIP could allow early intervention

    • The relatively defined genetic progression from CHIP to NPM1-mutated AML provides a window for preventive strategies

  • Research directions for prevention:

    • Identify factors that promote acquisition of NPM1 mutations in preleukemic clones

    • Develop interventions targeting preleukemic clones before NPM1 mutation acquisition

    • Investigate whether eradicating NPM1-mutated subclones before frank AML development can prevent leukemia

  • Methodological considerations:

    • Highly sensitive detection methods are required for early identification of NPM1-mutated clones

    • Longitudinal studies of individuals with CHIP are needed to understand progression kinetics

    • Animal models mimicking clonal progression may help test preventive interventions

Understanding the role of NPM1 mutations in the continuum from normal hematopoiesis to preleukemia to AML offers unique opportunities for early intervention and potentially prevention of leukemia development .

What novel technological approaches are advancing NPM1 research?

Several cutting-edge technologies are transforming NPM1 research and offering new insights into its function in normal and malignant hematopoiesis:

  • Single-cell multi-omics approaches:

    • Single-cell RNA sequencing reveals heterogeneity within NPM1-mutated leukemias

    • Single-cell ATAC-seq identifies altered chromatin accessibility patterns

    • Integrated single-cell multi-omics (RNA + DNA + protein) enables linkage of genetic lesions to their functional consequences

    • Spatial transcriptomics maps NPM1-mutated cell interactions within the bone marrow microenvironment

    Methodological consideration: Computational integration of multiple data layers requires sophisticated bioinformatic approaches.

  • Advanced genome editing technologies:

    • Base editing allows precise introduction of specific NPM1 mutations without DNA breaks

    • Prime editing enables scarless introduction of insertions mimicking clinical mutations

    • CRISPR activation/inhibition systems for modulating NPM1 and related pathways

    • CRISPR screens (genome-wide, focused libraries) to identify synthetic lethal interactions

    Methodological consideration: Off-target effects must be carefully controlled and verified.

  • Protein structure and interaction technologies:

    • Cryo-EM determination of NPM1 complexes at near-atomic resolution

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for mapping structural dynamics

    • Proximity labeling methods (BioID, APEX) to map compartment-specific interactomes

    • AlphaFold2 and other AI-based structural prediction tools to model mutation effects

    Methodological consideration: Validation of structural predictions with experimental data is essential.

  • Advanced imaging techniques:

    • Live-cell super-resolution microscopy for tracking NPM1 dynamics

    • Correlative light and electron microscopy (CLEM) for ultrastructural analysis

    • Lattice light-sheet microscopy for long-term 3D imaging with minimal phototoxicity

    • Expansion microscopy for visualization of subnuclear structures

    Methodological consideration: Maintaining physiological conditions during imaging is critical for relevant observations.

  • Organoid and advanced culture systems:

    • Patient-derived organoids recapitulating bone marrow architecture

    • Microfluidic "bone marrow-on-a-chip" systems

    • 3D bioprinted bone marrow models with defined cellular components

    • Co-culture systems mimicking the hematopoietic niche

    Methodological consideration: Validation of physiological relevance through comparison with in vivo systems.

These technological advances are enabling researchers to address previously intractable questions about NPM1 biology and provide new avenues for therapeutic development .

What are the most significant unresolved questions in NPM1 research?

Despite significant progress in understanding NPM1 biology and its role in leukemogenesis, several critical questions remain unresolved:

  • Mechanistic links between NPM1 mutation and HOX gene expression:

    • The precise molecular mechanisms by which cytoplasmic NPM1 maintains aberrant HOX expression remain unclear

    • Whether this involves direct chromatin interactions, regulation of transcription factors, or epigenetic modifications requires further investigation

  • Cooperative mechanisms with co-occurring mutations:

    • How NPM1 mutations functionally interact with common co-mutations (DNMT3A, FLT3-ITD, IDH1/2) at the molecular level

    • Whether specific cooperative interactions can be therapeutically targeted

  • Determinants of disease progression:

    • Why some individuals with NPM1 mutations develop overt AML while others may have more indolent disease

    • The role of the immune system in controlling NPM1-mutated clones

  • Therapeutic resistance mechanisms:

    • Molecular basis of relapse in NPM1-mutated AML

    • Whether NPM1-mutated subclones develop specific resistance mechanisms distinct from other AML subtypes

  • Normal functions of NPM1 in hematopoiesis:

    • Comprehensive understanding of NPM1's role in normal hematopoietic development

    • Cell type-specific functions of NPM1 across the hematopoietic hierarchy

  • Clinical translation:

    • Development of NPM1 mutation-specific therapies beyond general leukemia treatments

    • Optimal strategies for MRD-guided therapeutic decision-making

  • Long-term effects of persistent preleukemic clones:

    • Consequences of persistent DNMT3A or TET2 mutated clones after eradication of NPM1-mutated cells

    • Strategies to eliminate both leukemic and preleukemic clones

  • Biomarkers of response to targeted therapies:

    • Identification of predictive biomarkers for response to emerging therapies like menin-MLL inhibitors

    • Development of companion diagnostics for precision medicine approaches

Addressing these questions will require interdisciplinary approaches combining genomics, proteomics, structural biology, and functional studies in relevant model systems and clinical samples .

How can researchers effectively translate NPM1 research findings to clinical applications?

Translating NPM1 research findings to clinical applications requires strategic approaches to bridge laboratory discoveries and patient care:

  • Biomarker development and validation:

    • Standardization of NPM1 mutation detection methods for diagnosis

    • Validation of NPM1-based MRD assays in prospective clinical trials

    • Development of companion diagnostics for targeted therapies

    Methodological approach: Multi-center validation studies with harmonized protocols and quality control.

  • Preclinical therapeutic evaluation:

    • Testing candidate therapies across diverse NPM1-mutated models

    • Evaluating combination approaches based on mechanistic rationales

    • Investigating biomarkers of sensitivity and resistance

    Methodological approach: Use of clinically relevant endpoints and patient-derived models; evaluation of drug combinations using appropriate interaction models.

  • Clinical trial design:

    • Biomarker-driven patient selection strategies

    • Incorporation of MRD assessment as intermediate endpoint

    • Adaptive designs that allow rapid evaluation of promising agents

    • Inclusion of correlative studies to understand mechanisms of response and resistance

    Methodological approach: Collaboration between basic scientists, translational researchers, and clinicians in trial design; pre-planned translational endpoints.

  • Integration of real-world data:

    • Collection of outcomes data beyond clinical trials

    • Analysis of patterns of care and implementation of guideline recommendations

    • Health economics assessments to support access to novel therapies

    Methodological approach: Development of structured data collection systems; collaboration with health services researchers.

  • Knowledge dissemination and education:

    • Development of educational resources for clinicians

    • Integration of NPM1 testing into clinical practice guidelines

    • Patient education about the significance of NPM1 mutation status

    Methodological approach: Multidisciplinary consensus building; patient and advocate involvement in educational material development.

Product Science Overview

Gene Organization and Evolutionary History

The human NPM1 gene is located on chromosome 5q35 and is composed of 12 exons . It encodes at least two isoforms: NPM1.1 (or B23.1), which is the full-length transcript resulting in a 294 amino acids protein, and NPM1.3 (or B23.2), which lacks the last 35 amino acids of the NPM1 C-terminus . These isoforms have different expression levels and localization within the cell .

Functions of Nucleophosmin

Nucleophosmin is involved in several critical cellular functions:

  1. Ribosome Biogenesis: NPM1 is essential for the assembly and transport of ribosomes .
  2. mRNA Processing: It plays a role in the processing and transport of mRNA within the cell .
  3. Chromatin Remodeling: NPM1 interacts with chromatin, binding to histones and other chromatin remodeling proteins .
  4. Embryogenesis: It is involved in the early stages of embryonic development .
  5. DNA Repair and Apoptosis: NPM1 is crucial for maintaining genomic stability by participating in various DNA repair pathways and regulating apoptosis .
Role in Disease

NPM1 is frequently overexpressed, mutated, rearranged, and deleted in human cancers . It can function both as a proto-oncogene and a tumor suppressor . Mutations in the NPM1 gene, particularly those leading to the expression of a cytoplasmic mutant protein (NPMc+), are common in acute myeloid leukemia (AML) . These mutations are associated with a normal karyotype, involvement of different hematopoietic lineages, a specific gene-expression profile, and a favorable prognosis .

Recombinant Human Nucleophosmin

Recombinant human Nucleophosmin is typically expressed in E. coli cells using an N-terminal His tag . This recombinant protein retains the functional properties of the native protein and is used in various research applications to study its role in cellular processes and disease mechanisms .

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.