HN1 Human

Hematological And Neurological Expressed 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Protein Identity

HN1, also known as Jupiter microtubule associated homolog 1 (JPT1), ARM2, or HN1A, is encoded by a gene located on human chromosome 17q25.2 . The gene produces a small protein that is highly conserved in vertebrates, suggesting crucial biological functions . Interestingly, HN1 has no identified homolog protein other than HN1L, which shares only approximately 30% identity . Bioinformatic analysis predicts HN1 to be an intrinsically disordered protein lacking fixed tertiary structure, a characteristic often associated with proteins involved in signaling and regulation .

Expression Pattern

HN1 demonstrates ubiquitous expression across various human tissues, with particularly high levels in hematological and neurological tissues during development . This widespread expression pattern underscores its fundamental role in cellular processes. Gene Expression Profiling Interactive Analysis (GEPIA) data from The Cancer Genome Atlas (TCGA) has revealed differential expression of HN1 in numerous cancer types compared to normal tissues .

Cell Cycle-Dependent Regulation

A distinctive feature of HN1 is its fluctuating expression throughout the cell cycle. Research involving synchronized cell populations has demonstrated that HN1 levels are lowest in G1 phase, increase during the G1/S boundary, and peak during the late S-phase . Notably, HN1 expression decreases as cells progress through G2 and into mitosis . During mitosis, a unique phosphorylated form of HN1 appears, which is not observed in other cell cycle phases . This phosphorylation is regulated by specific kinases including GSK3β and Cdk1, as demonstrated through inhibitor studies .

Cell Cycle PhaseHN1 Expression LevelNotable Features
G0/G1 (serum-starved)LowestBase expression
G1/S boundaryIncreasedRising expression
Late S-phaseHighestPeak expression
Post-G2 (Nocodazole block)DecreasedHigher than G1
MitosisDecreased with phosphorylated formDistinct higher band on immunoblots

Cell Cycle Regulation

HN1 plays critical roles in cell cycle progression, particularly influencing the S-phase and mitotic exit. Experimental overexpression of HN1 before mitosis results in S-phase accumulation, while post-G2 overexpression causes accelerated mitotic exit . These findings establish HN1 as a novel cell cycle regulatory protein with potentially dual functions in cell cycle dynamics .

The relationship between HN1 and Cyclin B1 is particularly significant. Multiple studies have observed an inverse correlation between HN1 and Cyclin B1 expression—HN1 overexpression decreases Cyclin B1 levels, while HN1 depletion leads to Cyclin B1 accumulation . Mechanistically, HN1 facilitates Cyclin B1 degradation through increased ubiquitination, a process that can be reversed by proteasome inhibition .

Protein-Protein Interactions

HN1 engages in numerous protein-protein interactions that explain its diverse cellular functions:

  1. Interaction with Cdh1: HN1 associates with and stabilizes Cdh1, a co-factor of the Anaphase Promoting Complex/Cyclosome (APC/C). This stabilization enhances APC/C-Cdh1 activity, promoting ubiquitination and subsequent degradation of Cyclin B1 .

  2. Interaction with γ-tubulin: HN1 physically associates with γ-tubulin and regulates centrosome organization. Depletion of HN1 leads to increased γ-tubulin foci and disrupted microtubule spindle assembly .

  3. Interaction with APC/β-Catenin/GSK3β complex: HN1 physically interacts with this complex and is involved in regulating β-catenin cellular levels .

  4. Interaction with HMGB1: In hepatocellular carcinoma, HN1 prevents HMGB1 protein from ubiquitination and degradation via the autophagy-lysosome pathway, which involves interaction between HN1 and TRIM28 protein .

Signaling Pathway Modulation

HN1 influences several key signaling pathways:

  1. PI3K/AKT pathway: HN1 has inhibitory roles in PI3K/AKT signaling activities, particularly in prostate cancer cells .

  2. Androgen signaling: HN1 is regulated by androgens and EGF, and conversely modulates androgen signaling activities .

  3. c-Myc activation: In hepatocellular carcinoma, HN1 sustains stabilization and persistent activity of MYC via interaction with GSK3β, promoting cancer progression .

  4. Autophagy regulation: Through its interaction with HMGB1, HN1 regulates autophagy processes in cancer cells .

Expression in Cancer

HN1 overexpression has been documented in numerous cancer types compared to normal tissues:

Cancer TypeHN1 Expression PatternAssociated Clinical FeaturesReferences
Prostate CancerHighly expressedMalignant cell behaviors, poor prognosis
Hepatocellular CarcinomaUp-regulatedAssociated with age, stage, histologic grade, poor survival
Breast CancerElevatedPromotes invasion via MYC activity
NeuroblastomaHigher in undifferentiated cellsActs as dedifferentiation factor
Lung CancerOverexpressedPoor prognosis
MelanomaHighly expressedCancer progression
Malignant GliomasElevatedTumor development
Epithelial Ovarian CancerOverexpressedDisease progression

Diagnostic and Prognostic Value

Clinical studies have associated high HN1 expression with several adverse clinical features:

Cancer Promotion Mechanisms

HN1 contributes to cancer development through multiple mechanisms:

  1. Cell Proliferation: HN1 promotes cancer cell proliferation by regulating cell cycle progression, particularly influencing S-phase entry and mitotic exit .

  2. Migration and Invasion: Overexpression of HN1 enhances migration and invasion capabilities of cancer cells, demonstrated in both in vitro and in vivo models .

  3. Metastasis: HN1 facilitates metastatic processes, with experimental knockdown inhibiting tumor metastasis in animal models .

  4. Chemoresistance: In hepatocellular carcinoma, HN1 reduces sensitivity to oxaliplatin through its interaction with HMGB1. HN1 knockdown sensitizes cancer cells to chemotherapy .

  5. DNA Damage Response: Through its regulation of HMGB1, HN1 influences DNA damage and repair processes, affecting cancer cell survival following chemotherapy .

HN1 as a Therapeutic Target

The critical involvement of HN1 in cancer progression positions it as a promising therapeutic target. Several lines of evidence support this potential:

  1. Anti-proliferative effects: Depletion of HN1 leads to cell cycle arrest and apoptosis in cancer cells .

  2. Chemosensitization: HN1 knockdown sensitizes cancer cells to chemotherapeutic agents such as oxaliplatin in hepatocellular carcinoma .

  3. Tumor suppression: In animal models, HN1 knockdown inhibits tumor growth and metastasis, and enhances the efficacy of anticancer treatments .

  4. Pathway modulation: Targeting HN1 could simultaneously affect multiple oncogenic pathways, including MYC activation, β-catenin signaling, and autophagy regulation .

Challenges and Future Directions

Despite its promising potential, several challenges remain in developing HN1-targeted therapies:

  1. Target specificity: As HN1 is expressed in normal tissues, particularly in hematological and neurological systems, targeting strategies must be highly specific to cancer cells.

  2. Mechanistic complexity: HN1's involvement in multiple cellular pathways necessitates careful consideration of potential off-target effects.

  3. Delivery methods: Effective delivery of HN1 inhibitors to tumor sites represents another challenge for therapeutic development.

Future research directions include:

  • Development of small molecule inhibitors specific to HN1

  • Validation of efficacy and safety of HN1 inhibition in diverse cancer models

  • Development of diagnostic methods to determine HN1 expression levels in patients

  • Exploration of combination therapies involving HN1 inhibition and conventional treatments

Product Specs

Introduction
Hematological and neurological expressed 1 protein (HN1) is a highly conserved protein found across various species. It plays a role in neural tissue regeneration, as evidenced by its increased expression in damaged nerve cells. HN1 is also present during embryonic development and in areas of the adult brain associated with adaptability.
Description
This recombinant HN1 protein is produced in E. coli and consists of a single chain of 174 amino acids (specifically, amino acids 1 to 154 of the human HN1 sequence), resulting in a molecular weight of 18.1kDa. For purification and detection purposes, a 20 amino acid His-tag is attached to the protein's N-terminus. The protein is purified using specialized chromatographic methods.
Physical Appearance
Clear and sterile solution.
Formulation
The HN1 protein is provided at a concentration of 0.5mg/ml in a solution containing 20mM Tris-HCl buffer (pH 8.0), 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the protein at 4°C. For longer periods, freeze the protein at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
The purity of this protein is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
ARM2, HN1A, Hematological and neurological expressed 1 protein, Androgen-regulated protein 2, HN1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MTTTTTFKGV DPNSRNSSRV LRPPGGGSNF SLGFDEPTEQ PVRKNKMASN IFGTPEENQA SWAKSAGAKS SGGREDLESS GLQRRNSSEA SSGDFLDLKG EGDIHENVDT DLPGSLGQSE EKPVPAAPVP SPVAPAPVPS RRNPPGGKSS LVLG.

Q&A

What is HN1 and what are its primary biological functions?

HN1 (Hematological and Neurological expressed 1), also known as JPT1 (Jupiter Microtubule Associated Homolog 1), belongs to a small family of evolutionarily conserved genes found across vertebrate species . The protein plays a significant role in cell division processes, particularly through its interactions with centrosome-related structures. HN1 has been observed to co-localize with γ-tubulin at centrosomes during different phases of the cell cycle, demonstrating its involvement in microtubule spindle assembly and potentially centrosome clustering . This functionality appears critical for proper chromosomal segregation during mitosis, with alterations in HN1 expression linked to increased chromosomal aberrations and abnormal nuclei formation in prostate cancer cell models . The protein's expression levels vary significantly between normal human cells and cancerous cell lines, suggesting a regulatory role in cellular proliferation that becomes dysregulated during carcinogenesis.

How is HN1 expression regulated in normal versus cancerous human tissues?

Research indicates differential expression patterns of HN1 between normal and cancerous human tissues. In prostate tissue specifically, HN1 expression is significantly lower in normal RWPE-1 cells compared to cancerous cell lines such as androgen-responsive LNCaP and androgen-insensitive PC-3 cells . Experimental evidence suggests that HN1 expression levels correlate with the aggressiveness of cancer phenotypes, with higher expression observed in more advanced cancer stages. The regulatory mechanisms controlling HN1 expression appear complex and tissue-specific, involving both transcriptional and post-transcriptional processes. In experimental models, restoration of HN1 expression after silencing demonstrates the protein's potential role in centrosome clustering, suggesting feedback mechanisms that attempt to maintain appropriate levels for proper cell division . This differential regulation makes HN1 a potential biomarker for distinguishing between normal and cancerous states in multiple tissue types.

What is the relationship between HN1 and the HN-1 peptide in cancer research?

It's crucial for researchers to distinguish between HN1 (the gene/protein) and HN-1 (the peptide), as they represent different entities in cancer research despite their similar nomenclature. While HN1 functions primarily in cell division processes and has been implicated in prostate carcinogenesis , HN-1 peptide is a tumor-targeting molecule specifically designed to target head and neck squamous cell carcinoma (HNSCC) . The HN-1 peptide demonstrates remarkable tumor-selectivity and can translocate across cell membranes to penetrate solid tumors . Unlike the endogenous HN1 protein, the HN-1 peptide has been extensively studied as a delivery vehicle for cancer therapeutics and imaging agents, with applications expanded to breast, thyroid, cervical, lung, uterine, and colon cancers . Understanding this distinction is essential for proper experimental design and interpretation of research findings, as conflation of these entities could lead to significant methodological errors.

What are the optimal experimental models for studying HN1 function in human cancers?

When designing experiments to investigate HN1 function in human cancers, researchers should consider a multi-model approach that accounts for HN1's differential expression across cancer types. Cell line models should include both normal cells (e.g., RWPE-1 for prostate studies) and cancerous cell lines with varying characteristics (e.g., androgen-responsive LNCaP and androgen-insensitive PC-3 cells) . This comparative approach enables investigation of HN1's role across the spectrum of cancer progression. For mechanistic studies of HN1's role in centrosome function, immunofluorescence microscopy with co-localization analysis of HN1 and γ-tubulin provides valuable insights into physical interactions and spatial distribution . Complementary approaches include co-immunoprecipitation to validate protein-protein interactions and gene silencing/overexpression experiments to assess functional outcomes. For translational relevance, these in vitro findings should be validated in patient-derived xenograft models and clinical specimens, comparing HN1 expression and associated phenotypes across tumor grades and stages.

How can researchers effectively measure the impact of HN1 alterations on centrosome function?

Quantitative assessment of HN1's impact on centrosome function requires a multi-parametric approach combining advanced imaging techniques with molecular analyses. High-resolution confocal microscopy with immunofluorescent labeling of centrosomal markers (γ-tubulin, centrin) alongside HN1 enables visualization of structural alterations . For quantitative assessment, researchers should implement automated image analysis workflows to measure centrosome number, size, and clustering across large cell populations. Live-cell imaging using fluorescently tagged centrosome components allows temporal tracking of centrosome dynamics throughout the cell cycle in response to HN1 modulation. Complementary biochemical approaches should include analysis of centrosome-associated protein complexes via proximity ligation assays or mass spectrometry following HN1 immunoprecipitation. Functional consequences can be assessed through mitotic spindle formation analysis, chromosomal segregation tracking, and quantification of micronuclei formation as indicators of chromosomal instability . This multi-dimensional approach provides comprehensive insights into how HN1 alterations affect centrosome biology in both normal and pathological contexts.

What methodological approaches are most effective for studying HN1's interaction with γ-tubulin in centrosome regulation?

To rigorously study HN1's interaction with γ-tubulin in centrosome regulation, researchers should employ complementary techniques that validate physical associations and functional consequences. Co-immunoprecipitation experiments using antibodies against either HN1 or γ-tubulin, followed by Western blot analysis for the reciprocal protein, provide biochemical evidence of interaction . For spatial resolution, proximity ligation assays or fluorescence resonance energy transfer (FRET) microscopy offer visualization of protein-protein interactions at the nanometer scale within intact cells. Domain mapping through truncation mutants or site-directed mutagenesis of HN1 can identify specific regions required for γ-tubulin interaction. Functional consequences should be assessed through γ-tubulin recruitment assays following HN1 depletion or overexpression, with quantification of γ-tubulin foci number and intensity . Cell cycle synchronization techniques enable investigation of temporal dynamics of this interaction across mitotic phases. For mechanistic insights, in vitro reconstitution assays using purified components can determine whether the interaction is direct or requires additional factors, while structural biology approaches (X-ray crystallography, cryo-EM) may elucidate the molecular basis of the interaction.

How should researchers design experiments to investigate HN1's role in cell cycle progression?

Designing robust experiments to investigate HN1's role in cell cycle progression requires careful consideration of variables and controls. The independent variable would be HN1 expression levels (manipulated through overexpression, knockdown, or knockout approaches), while dependent variables should include cell cycle phase distribution, proliferation rates, and centrosome dynamics . Begin with a specific, testable hypothesis, such as "HN1 depletion alters G2/M progression through disruption of centrosome clustering." Cell synchronization techniques (double thymidine block or nocodazole treatment) allow precise examination of specific cell cycle phases. Flow cytometry with propidium iodide staining provides quantitative data on cell cycle distribution, while EdU incorporation assays measure S-phase progression. For mechanistic insights, combine these approaches with time-lapse microscopy of fluorescently labeled centrosome markers following HN1 modulation . Proper experimental controls should include scrambled siRNA/shRNA for knockdown studies and empty vector transfections for overexpression experiments. Multiple cell lines should be tested to determine whether HN1's effects on cell cycle are context-dependent or universally applicable across different cancer types.

What experimental approaches are appropriate for investigating the clinical significance of HN1 expression in human cancers?

To establish the clinical significance of HN1 expression in human cancers, researchers should implement a translational research framework combining retrospective tissue analyses with functional validation. The experimental design should include:

Research PhaseMethodologyKey MeasurementsStatistical Approach
DiscoveryImmunohistochemistry on tissue microarraysHN1 expression across cancer stages/gradesCorrelation with clinicopathological features
ValidationMining public genomic databases (TCGA, GEO)HN1 mRNA expression, mutation statusSurvival analysis, multivariate modeling
FunctionalRNAi or CRISPR in patient-derived modelsProliferation, invasion, drug responseComparative analysis with expression data
MechanisticProteomic analysis of HN1 interactomePathway enrichment, centrosome componentsNetwork analysis, enrichment testing

This multi-phase approach enables robust assessment of HN1's prognostic and predictive value. To control for confounding variables, tissue samples should be matched for demographic factors, treatment history, and molecular subtypes . Statistical analysis should include multivariate Cox regression to determine whether HN1 expression represents an independent prognostic factor when accounting for established clinical variables. Functional validation in patient-derived models provides causal evidence linking HN1 expression to clinically relevant phenotypes, strengthening translational relevance.

How can researchers effectively design experiments to study the dynamics of HN1 expression throughout the cell cycle?

Studying HN1 expression dynamics throughout the cell cycle requires experimental designs that capture temporal changes with high precision. Researchers should implement cell synchronization methods (serum starvation-release, thymidine block-release, or selective inhibitors) to obtain populations enriched in specific cell cycle phases . Following synchronization, time-course sampling should be performed at carefully determined intervals, with cell cycle phase verification using flow cytometry with propidium iodide staining or EdU incorporation. For protein-level analysis, Western blotting of HN1 alongside established cell cycle markers (cyclins A, B, D, E) provides correlation with specific phases . Immunofluorescence microscopy enables visualization of HN1 subcellular localization changes throughout the cycle, particularly in relation to centrosomal structures. For single-cell resolution, live-cell imaging using fluorescently tagged HN1 combined with cell cycle phase markers (PCNA, H2B) allows continuous monitoring of expression and localization dynamics. Quantitative image analysis should include measurement of HN1 intensity at centrosomes relative to total cellular levels across different phases. To determine regulation mechanisms, researchers should investigate how cyclin-dependent kinase inhibitors affect HN1 expression/localization and whether HN1 undergoes post-translational modifications during cell cycle progression.

How should researchers approach conflicting data regarding HN1's role in different cancer types?

When confronted with conflicting data regarding HN1's role across different cancer types, researchers should implement a systematic analytical framework rather than dismissing contradictions. Begin by evaluating methodological differences between studies, including cell line models, experimental techniques, and analytical approaches that might account for discrepancies. Context-dependent functions of HN1 should be considered, as its interactions with γ-tubulin and effects on centrosome dynamics may vary based on the molecular landscape of different cancer types . To resolve conflicts, design comparative experiments that directly test HN1 function across multiple cancer models under identical conditions, controlling for variables like proliferation rate, p53 status, and centrosome abnormality baseline. Meta-analysis of published data can identify patterns in HN1's effects across cancer types, potentially revealing tissue-specific cofactors or regulatory mechanisms. Computational approaches, including pathway enrichment analysis of HN1-associated genes in different cancers, may uncover context-specific interaction networks. Ultimately, these conflicts should be viewed as opportunities to discover nuanced biological mechanisms rather than experimental failures, potentially revealing how HN1's function adapts to different cellular environments.

What statistical approaches are most appropriate for analyzing HN1 expression data in relation to clinical outcomes?

The analysis of HN1 expression data in relation to clinical outcomes requires robust statistical approaches that account for the complexity of cancer progression and treatment response. For survival analysis, Kaplan-Meier curves with log-rank tests provide initial visualization of differences between high and low HN1 expression groups, but must be followed by multivariate Cox proportional hazards models to adjust for confounding clinical variables (age, stage, grade, treatment) . Rather than arbitrary cutoffs, researchers should determine HN1 expression thresholds through methods like minimal p-value approach, quartile distribution, or computational algorithms (X-tile, CUTP). For continuous analysis of HN1 expression, restricted cubic splines in Cox models can reveal non-linear relationships with outcomes. When analyzing multiple datasets, meta-analysis techniques should be applied to estimate pooled hazard ratios, with random-effects models preferred when significant heterogeneity exists between studies. For predictive biomarker assessment, interaction tests between HN1 expression and treatment in Cox models determine whether HN1 predicts differential treatment benefit. To validate prognostic models incorporating HN1, researchers should report discrimination (Harrell's C-index) and calibration metrics, with internal validation through bootstrapping and external validation in independent cohorts. Advanced machine learning approaches may reveal complex patterns in HN1's relationship with outcomes when combined with other molecular features.

How can researchers accurately interpret changes in centrosome number and structure in relation to HN1 alterations?

Accurate interpretation of centrosome abnormalities in relation to HN1 alterations requires careful quantitative analysis and contextual understanding. When assessing centrosome changes, researchers should implement standardized classification systems that distinguish between numerical abnormalities (amplification) and structural defects (enlargement, fragmentation) . Quantification should include multiple parameters:

  • Centrosome number per cell (using γ-tubulin as marker)

  • Centrosome size and intensity

  • Centrosome clustering patterns

  • Correlation with nuclear abnormalities

  • Distribution across cell cycle phases

Statistical analysis should consider the non-normal distribution of centrosome numbers, often requiring non-parametric tests. When comparing HN1-altered cells to controls, researchers should report both mean values and frequency distributions of centrosome abnormalities, as subtle shifts in distribution can have significant biological consequences . Importantly, researchers must distinguish between primary effects of HN1 on centrosomes and secondary consequences of altered cell cycle dynamics, potentially using cell cycle-arrested populations to isolate direct centrosomal effects. The functional consequences of observed centrosome changes should be assessed through mitotic spindle analysis, chromosome segregation tracking, and micronuclei quantification. Correlation analyses between HN1 expression levels and the degree of centrosome abnormality can establish dose-dependency of this relationship, strengthening causal interpretations.

What are the most promising approaches for leveraging HN1 knowledge in cancer therapeutic development?

Leveraging HN1 knowledge for cancer therapeutic development presents multiple promising avenues based on its role in centrosome regulation and cell division. Disrupting HN1's interaction with γ-tubulin using small molecule inhibitors or peptide mimetics represents a targeted approach, potentially exploiting cancer cells' heightened dependency on centrosome clustering for survival . High-throughput screening of compound libraries against the HN1-γ-tubulin interaction interface could identify lead candidates for development. For more selective targeting, researchers should investigate cancer-specific post-translational modifications of HN1 that might enable development of compounds with preferential activity in malignant cells. Synthetic lethality approaches present another opportunity, identifying genes whose inhibition is selectively lethal when combined with HN1 overexpression or depletion. The development of proteolysis-targeting chimeras (PROTACs) directed against HN1 could achieve more complete protein elimination than conventional inhibitors. For advanced delivery strategies, conjugating anti-cancer agents to HN-1 peptide could enhance tumor-specific delivery, leveraging its demonstrated ability to translocate across cell membranes and penetrate solid tumors . Combination therapies pairing HN1-targeted agents with microtubule-disrupting drugs might produce synergistic effects by simultaneously targeting interdependent aspects of mitotic machinery.

How might single-cell technologies advance our understanding of HN1's role in cancer heterogeneity?

Single-cell technologies offer transformative approaches for understanding HN1's contribution to cancer heterogeneity, moving beyond bulk tissue analyses that mask important subpopulation dynamics. Single-cell RNA sequencing can reveal how HN1 expression varies across individual cells within tumors, potentially identifying rare cell populations with extreme expression levels that drive aggressive phenotypes . This approach enables correlation of HN1 with broader transcriptional programs at single-cell resolution, revealing co-expression patterns that suggest functional relationships. Complementary single-cell proteomics through mass cytometry (CyTOF) or multiplexed immunofluorescence can map HN1 protein levels alongside post-translational modifications and interacting partners across thousands of individual cells. Spatial transcriptomics and imaging mass cytometry provide critical spatial context, determining whether HN1-expressing cells occupy specific tumor microenvironments or form distinct niches. For functional assessment, high-throughput CRISPR perturbation combined with single-cell readouts can systematically map genetic interactions with HN1 across heterogeneous populations. Computational integration of these multi-omic datasets can construct predictive models of how HN1 contributes to emergence of resistant cell populations during treatment, potentially identifying combination therapy strategies that prevent adaptive resistance driven by HN1-expressing subclones.

What interdisciplinary approaches might reveal new insights into HN1's evolutionary conservation and fundamental biological functions?

Understanding HN1's evolutionary conservation and fundamental functions requires interdisciplinary approaches spanning computational biology, structural biology, developmental biology, and systems biology. Comparative genomics analyses across species can trace HN1's evolutionary history, identifying conserved domains and regulatory elements that suggest fundamental functions maintained through natural selection . Structural biology approaches (X-ray crystallography, cryo-EM, NMR) can elucidate HN1's three-dimensional structure and conformational changes upon binding to partners like γ-tubulin, providing mechanistic insights into its centrosomal functions. Developmental biology models offer opportunities to study HN1's role in embryogenesis and tissue differentiation, potentially revealing functions beyond cancer contexts. Using gene editing to create conditional knockout models in model organisms allows tissue-specific and temporal control of HN1 expression, revealing its role in organismal development and tissue homeostasis. Systems biology approaches combining mathematical modeling with experimental validation can place HN1 within broader regulatory networks governing centrosome dynamics and cell division. These models could predict emergent properties and system-level responses to HN1 perturbation that aren't evident from reductionist approaches. Interdisciplinary collaboration between cancer biologists, evolutionary biologists, and structural biologists will be essential for developing a comprehensive understanding of HN1's fundamental biological significance beyond its cancer-associated functions.

Product Science Overview

Expression and Function

HN1 expression is upregulated in regenerating neural tissues, such as the axotomized adult rodent facial motor nerve and dedifferentiating retinal pigment epithelial cells of the Japanese newt . It is also expressed in numerous tissues during embryonic development and in regions of the adult brain that exhibit high plasticity .

Role in Tumorigenesis

HN1 is upregulated in many tumors, including breast cancer. It contributes to the migration, invasion, and tumorigenesis of breast cancer cells . However, the regulatory mechanisms of HN1 in cancer progression are not yet fully understood .

Recombinant Human HN1 Protein

Recombinant human HN1 protein is produced using Escherichia coli (E. coli) as the expression system. The recombinant protein is fused to a His-tag at the N-terminus and corresponds to the amino acids 1-154 of human HN1 . The protein is purified using conventional chromatography techniques and has a purity of over 90% as determined by SDS-PAGE .

Applications

Recombinant human HN1 protein is used in various research applications, including studies on neural regeneration, embryonic development, and cancer research. It is important to note that this product is for research use only and is not approved for use in humans or clinical diagnosis .

Storage and Handling

The recombinant human HN1 protein should be stored at 4°C for short-term use and at -20°C for long-term storage. It is recommended to avoid freeze-thaw cycles to maintain the protein’s stability .

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.