HRAS Human

V-Ha-ras Harvey Rat Sarcoma Viral Oncogene Homolog Human Recombinant
Shipped with Ice Packs
In Stock

Description

Genetic Structure and Functional Mechanisms

The HRAS gene spans 3,308 base pairs, encoding a 186-amino-acid protein with a C-terminal isoprenyl group enabling membrane anchoring . H-Ras cycles between GTP-bound (active) and GDP-bound (inactive) states, regulated by:

  • Guanine Nucleotide Exchange Factors (GEFs): Promote GTP binding (e.g., SOS1)

  • GTPase-Activating Proteins (GAPs): Accelerate GTP hydrolysis (e.g., RasGAP)

DomainFunctionKey Interactions
GTP-bindingSignal transduction activationc-Raf, PI3K, RalGEF
C-terminalMembrane localization via isoprenylationCaveolin-1, Farnesyltransferase
Switch regionsConformational changes for effector bindingKinase domains, adaptor proteins

H-Ras signaling is tightly regulated under normal conditions but becomes dysregulated in oncogenic mutations, leading to constitutive activation .

2.1. Costello Syndrome

HRAS mutations (e.g., G12S, G12V) cause Costello syndrome, characterized by:

  • Somatic overgrowth prenatally

  • Cardiovascular defects (e.g., hypertrophic cardiomyopathy)

  • Neurocognitive impairment

  • Tumor predisposition (rhabdomyosarcoma, neuroblastoma)

2.2. Cancer Involvement

HRAS mutations are implicated in diverse cancers, with distinct mutation patterns:

Cancer TypeMutation FrequencyCommon CodonsPrognostic ImpactSources
Bladder cancer1–2%G12V, G13RHigh-grade, advanced disease
Head and neck SCC3–4%G12SHPV-negative, poor OS
Thyroid carcinoma~10%G12V, G13CAggressive, poor prognosis
Gastric carcinoma5–10%G12V, G13DEnhanced metastasis, angiogenesis

3.1. Oncogenic Mutations

HRAS mutations typically occur at codons 12, 13, or 61, disrupting GTP hydrolysis:

CodonAmino Acid ChangePathogenic EffectAssociated Cancers
12Gly→Ser/Val/ArgConstitutive activation of MAPK/PI3KBladder, head/neck, thyroid
13Gly→Arg/CysImpaired GAP-mediated GTP hydrolysisGastric, colon
61Gln→Lys/ArgReduced GTPase activityRare in solid tumors

3.2. Comutation Patterns

HRAS-mutant cancers often co-occur with:

  • TERT promoter mutations (60% in HNSCC)

  • CASP8 inactivation (27% in HNSCC)

  • NOTCH1 mutations (30.5% in HNSCC)

  • CDKN2A loss (53% in HNSCC)

4.1. Head and Neck Squamous Cell Carcinoma (HNSCC)

  • Demographics: 64% male, median age 55–65 years

  • Treatment Outcomes:

    • Tipifarnib: Improved OS from 15 months (wild-type) to 25.5 months

    • Recurrence: 50–67% relapse within 6 months post-treatment

4.2. Gastric Carcinoma

  • HRAS Overexpression: Correlates with poor survival (HR: 2.1, P < 0.05)

  • Mechanisms:

    • VEGFA/PI3K/AKT activation: Promotes angiogenesis

    • Raf-1 signaling: Enhances proliferation and invasion

5.1. Small-Molecule Inhibitors

AgentTargetMechanismClinical Status
TipifarnibFarnesyltransferasePrevents H-Ras membrane localizationPhase II trials
DM6ACRISPRHRAS m6A modificationSuppresses translation elongationPreclinical

5.2. Epitranscriptic Regulation

HRAS mRNA undergoes N6-methyladenosine (m6A) modification, which:

  • Enhances translation: Via YTHDF1-mediated elongation

  • Therapeutic Potential:

    • FTO/YTHDF1 inhibitors: Reduce H-Ras expression in cancers

    • CRISPR-dCas9 systems: Target m6A sites to suppress tumor growth

Product Specs

Introduction
HRAS, a member of the Ras oncogene family, shares similarities with the transforming genes found in mammalian sarcoma retroviruses. These genes encode proteins involved in signal transduction pathways. These proteins exhibit the ability to bind both GTP and GDP and possess intrinsic GTPase activity. HRAS undergoes a continuous cycle of palmitoylation and depalmitoylation, facilitating its rapid movement between the plasma membrane and the Golgi apparatus. Mutations in HRAS are linked to Costello syndrome, a disorder characterized by excessive prenatal growth, postnatal growth retardation, increased susceptibility to tumor development, intellectual disabilities, skin and musculoskeletal abnormalities, distinctive facial features, and cardiovascular defects. Abnormalities in the HRAS gene are implicated in various cancers, including bladder cancer, follicular thyroid cancer, and oral squamous cell carcinoma.
Description
Recombinant Human HRAS, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 194 amino acids (residues 1-186). This protein has a molecular weight of 22 kDa. An 8-amino acid His-tag is fused to the C-terminus of the HRAS protein, which is purified using standard chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
A solution at a concentration of 0.5 mg/ml containing 20 mM Tris-HCl (pH 8), 0.1 M NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), keep refrigerated at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 90% as determined by SDS-PAGE analysis.
Synonyms
C-BAS/HAS, C-H-RAS, C-HA-RAS1, CTLO, H-RASIDX, HAMSV, HRAS1, K-RAS, N-RAS, RASH1.
Source
Escherichia Coli.
Amino Acid Sequence
MTEYKLVVVG AGGVGKSALT IQLIQNHFVD EYDPTIEDSY RKQVVIDGET CLLDILDTAG QEEYSAMRDQ YMRTGEGFLC VFAINNTKSF EDIHQYREQI KRVKDSDDVP MVLVGNKCDL AARTVESRQA QDLARSYGIP YIETSAKTRQ GVEDAFYTLV REIRQHKLRK LNPPDESGPG CMSCKCLEHH HHHH.

Q&A

What is the HRAS gene and what is its primary function in human cells?

HRAS (Harvey rat sarcoma viral oncogene homolog) is a member of the RAS family of oncogenes located on chromosome 11p15.5. It spans approximately 3,308 base pairs from position 532,242 to 535,550 on the plus strand . At the molecular level, HRAS functions as a GTPase that plays a crucial role in cell growth and differentiation by acting as a molecular switch in signal transduction pathways.

The HRAS protein participates in multiple cellular processes including:

  • MAPK cascade signaling

  • Cell surface receptor signaling pathways

  • Epidermal growth factor receptor signaling

  • Cell cycle arrest and mitotic cell cycle checkpoint functions

  • Endocytosis and chemotaxis

Research methodologies for studying HRAS function typically involve knockdown/knockout approaches using siRNA or CRISPR-Cas9, coupled with signaling pathway analyses using phospho-specific antibodies to detect activation of downstream effectors.

How does HRAS differ from other RAS family proteins, and why is this significant?

HRAS shares significant homology with other RAS family members (KRAS and NRAS), particularly in the G-domain, but contains distinct regions that influence its specific functions. While the three isoforms have similar biochemical properties, they exhibit different binding interactions that significantly impact their oncogenic potential.

Methodologically, understanding these differences requires:

  • Structural analysis through X-ray crystallography or molecular dynamics simulations

  • Protein-protein interaction studies using co-immunoprecipitation, proximity ligation assays, or FRET

  • Isoform-specific knockdown/knockout experiments to determine non-redundant functions

Recent molecular dynamics simulations have revealed that wildtype KRAS binds to HRAS with an average binding energy of -46.68 kcal/mol, demonstrating thermodynamically favorable interaction . This binding pattern is significant as it may serve as an intrinsic regulatory mechanism among RAS isoforms.

What experimental models are most appropriate for studying HRAS in human aging and cancer?

When selecting experimental models for HRAS research, consider both the research question and the translational relevance:

Model SystemAdvantagesLimitationsBest Applications
Human cell linesDirect relevance to human biologyLimited physiological contextMolecular mechanisms, signaling, drug screening
Patient-derived organoidsMaintains tissue architecture and heterogeneityShort lifespan, labor-intensivePersonalized medicine, tumor microenvironment
Mouse modelsIn vivo context, genetic manipulationSpecies differences, time-consumingTumorigenesis, aging, therapeutic testing
Computational modelsRapid hypothesis testing, structure predictionRequires validationDrug design, interaction prediction
C. elegansRapid aging studies, genetic tractabilityEvolutionary distance from humansBasic RAS pathway conservation studies

For aging research, C. elegans has provided valuable insights despite evolutionary distance, as the RAS pathway influences both development and aging in conjunction with the insulin/IGF1 signaling pathway . For cancer studies, patient-derived xenografts and genetically engineered mouse models expressing mutant HRAS provide the most translational relevance.

How do specific HRAS mutations alter protein structure and function in human cancers?

HRAS mutations in cancer predominantly occur at specific hotspots (particularly G12, G13, and Q61), creating constitutively active forms that promote uncontrolled cellular proliferation. These mutations impair intrinsic GTPase activity or prevent interaction with GTPase-activating proteins (GAPs).

Research approaches to characterize mutational effects include:

  • Structural analysis using molecular dynamics simulations to visualize conformational changes

  • Binding assays to measure altered interactions with effector proteins

  • Functional assays measuring downstream pathway activation (e.g., MAPK, PI3K)

  • Cellular transformation assays to quantify oncogenic potential

Molecular dynamics simulations have revealed significant differences in binding energies between wildtype HRAS and various KRAS mutants. For example, a G12D mutation in KRAS results in weaker binding to wildtype HRAS (with a standardized mean of 1.20 kcal/mol compared to wildtype), while a G13D mutation significantly strengthens this interaction (standardized mean of -22.04 kcal/mol) . These binding differences may explain variations in oncogenic potency among different RAS mutations.

What methodological approaches best capture the dynamic interactions between HRAS and other signaling proteins?

Studying dynamic HRAS interactions requires methodologies that can capture transient and regulated protein-protein interactions in living cells:

  • Live-cell imaging techniques:

    • FRET/BRET to measure real-time protein interactions

    • Optogenetic approaches to control RAS activation with spatial and temporal precision

    • Fluorescence correlation spectroscopy to measure diffusion dynamics

  • Biochemical approaches with temporal resolution:

    • Time-course immunoprecipitation following stimulation

    • Proximity labeling techniques (BioID, APEX) with controlled labeling windows

    • Hydrogen-deuterium exchange mass spectrometry to identify binding interfaces

  • Computational approaches:

    • Molecular dynamics simulations examining conformational changes over time

    • Root mean square fluctuation (RMSF) analysis to identify mobile regions involved in interactions

    • Binding energy calculations to quantify interaction strength

Studies have used these approaches to identify that HRAS interacts with multiple proteins relevant to aging and cancer, including TP53, PLAU, INSR, MAPK8, PIK3R1, and PIK3CA . These interaction partners provide insights into how HRAS integrates various signaling inputs to influence cellular outcomes.

How can molecular dynamics simulations advance our understanding of HRAS-isoform interactions?

Molecular dynamics (MD) simulations provide powerful insights into HRAS interactions that are difficult to capture experimentally:

Methodological workflow for effective MD simulations of HRAS:

Recent MD simulations revealed that wildtype KRAS binds to wildtype HRAS with remarkable stability (standard deviation of 3.85 kcal/mol), while mutant forms show variable binding patterns. The G13D KRAS mutant demonstrated the strongest binding to wildtype HRAS among all tested variants, suggesting potential therapeutic relevance for modulating these interactions .

What experimental challenges arise when studying HRAS post-translational modifications, and how can they be overcome?

Post-translational modifications (PTMs) of HRAS, including farnesylation, palmitoylation, and phosphorylation, critically regulate its localization and activity. Studying these modifications presents several challenges:

Challenge 1: Detecting specific modifications

  • Solution: Combine enrichment strategies (e.g., click chemistry for lipid modifications, phospho-specific antibodies) with high-resolution mass spectrometry

  • Method improvement: Use targeted multiple reaction monitoring (MRM) mass spectrometry for improved sensitivity

Challenge 2: Temporal dynamics of modifications

  • Solution: Use pulse-chase approaches with metabolic labeling

  • Method improvement: Develop biosensors that report specific PTM states in real-time

Challenge 3: Functional relevance of PTM patterns

  • Solution: Generate modification-specific mutants and assess activity

  • Method improvement: Combine with proximity labeling to identify PTM-specific interactors

Challenge 4: Spatial organization of modified HRAS

  • Solution: Use super-resolution microscopy to visualize nanoscale distribution

  • Method improvement: Correlative light and electron microscopy to link localization to membrane ultrastructure

When designing experiments, researchers should consider how PTMs may affect the results of interaction studies. For example, membrane-associated HRAS (with intact lipid modifications) may have different binding properties compared to recombinant proteins used in computational models or in vitro assays.

How do the kinetic properties of HRAS differ between normal and pathological states?

The kinetic properties of HRAS—including nucleotide binding, hydrolysis rates, and exchange kinetics—are fundamentally altered in pathological states such as cancer and certain developmental disorders:

Methodological approaches to measure HRAS kinetics:

  • Real-time nucleotide binding/hydrolysis assays:

    • Fluorescent nucleotide analogs (mant-GTP, BODIPY-GTP)

    • Stopped-flow spectroscopy for rapid kinetic measurements

    • Single-molecule approaches for heterogeneity analysis

  • Cellular activation dynamics:

    • FRET-based biosensors to measure activation in living cells

    • Optogenetic approaches to trigger activation with precise timing

    • Mathematical modeling to extract rate constants from cellular data

  • Comprehensive kinetic parameter determination:

    • Determine association (kon) and dissociation (koff) rate constants

    • Measure GTP hydrolysis rates (kcat)

    • Quantify nucleotide exchange rates with and without GEFs

Recent kinetic studies of RAS proteins, including HRAS, demonstrate that oncogenic mutations primarily affect the GTP-bound state stability and interaction with regulatory proteins rather than the intrinsic nucleotide binding properties. These alterations create a prolonged active state that drives downstream signaling .

What are the most reliable antibody-based methods for detecting HRAS in human samples?

Antibody-based detection of HRAS requires careful consideration of specificity, given the high homology between RAS family members:

Best practices for HRAS immunodetection:

  • Antibody selection considerations:

    • Verify isoform specificity using knockout/knockdown controls

    • Validate in multiple sample types (cell lines, tissues, etc.)

    • Test for cross-reactivity with KRAS and NRAS using recombinant proteins

  • Recommended detection methods:

    • Western blotting with isoform-specific antibodies

    • Immunoprecipitation followed by mass spectrometry for confirmation

    • Immunohistochemistry with validated antibodies and appropriate controls

  • Quantification approaches:

    • Use multiple antibodies targeting different epitopes

    • Include loading controls and standardization samples

    • Consider digital pathology tools for quantitative immunohistochemistry

How can researchers effectively design CRISPR-Cas9 experiments to study HRAS function?

CRISPR-Cas9 technology offers powerful approaches for manipulating HRAS in research models, but requires careful design:

CRISPR experimental design workflow for HRAS studies:

  • Guide RNA design:

    • Target unique regions that differ from KRAS/NRAS to ensure specificity

    • Design multiple guides with minimal off-target effects

    • Consider the functional domains being disrupted (e.g., GTP-binding regions)

  • Editing strategy selection:

    • Complete knockout via frameshift indels

    • Base editing for specific point mutations (e.g., G12V)

    • Knock-in of fluorescent tags for localization studies

    • Inducible CRISPR systems for temporal control

  • Validation requirements:

    • Sequence verification of edits

    • Western blotting to confirm protein changes

    • RNA-seq to rule out compensatory expression of other RAS isoforms

    • Functional testing of downstream pathways

  • Analytical considerations:

    • Develop clear phenotypic readouts relevant to HRAS function

    • Use clonal populations or single-cell analysis to address heterogeneity

    • Include rescue experiments with wildtype HRAS to confirm specificity

When introducing specific mutations, prime editing or base editing systems may offer advantages over traditional homology-directed repair, particularly for the common G→T transversions found in HRAS mutant cancers.

What bioinformatic tools and databases are most valuable for HRAS human research?

A comprehensive bioinformatic analysis of HRAS requires multiple specialized tools and databases:

CategoryTool/DatabasePrimary UseBest Applied To
Sequence AnalysisUniProtProtein sequence and annotationIdentifying domains and PTM sites
Structural AnalysisAlphaFold DBProtein structure predictionModeling HRAS mutants
GROMACSMolecular dynamics simulationsStudying dynamic interactions
PyMOLStructural visualizationAnalyzing binding interfaces
Mutation AnalysisCOSMICCancer mutation frequenciesIdentifying hotspots
cBioPortalCancer genomics dataCorrelating mutations with outcomes
Pathway AnalysisSTRINGProtein-protein interactionsIdentifying HRAS interactors
ReactomePathway mappingContextualizing HRAS signaling
Expression AnalysisGEPIAGene expression in cancersTumor vs. normal comparisons
GTExTissue-specific expressionUnderstanding normal expression patterns
Aging ResearchGenAgeAge-related gene databaseHRAS connections to aging phenotypes

When conducting computational analyses, researchers should be aware that the confidence intervals for HRAS interactions with other proteins vary. For example, the interaction between KRAS and HRAS has an experimental and biochemical data confidence interval of 0.893, while the interaction between HRAS and NRAS has a confidence interval of 0.877 . These confidence scores should inform interpretation of predicted interaction networks.

How can understanding HRAS-isoform interactions inform therapeutic strategies for RAS-driven cancers?

The complex interactions between HRAS and other RAS isoforms present novel therapeutic opportunities:

Recent research has revealed that wildtype HRAS can interact with mutant KRAS proteins, potentially modulating their oncogenic activity. Molecular dynamics simulations have identified specific binding interfaces and energetics that could be exploited therapeutically . This approach represents a paradigm shift from direct RAS inhibition to modulating inter-isoform interactions.

Therapeutic strategies based on HRAS interactions:

  • Peptide mimetics approach:

    • Design peptides mimicking the hypervariable region of HRAS that interact with KRAS

    • Optimize binding to mutant KRAS over wildtype forms

    • Engineer cell-penetrating capabilities and stability enhancements

  • Small molecule development:

    • Target specific residues identified in binding interfaces

    • Focus on compounds that stabilize HRAS-KRAS interactions

    • Leverage the different binding energies observed between wildtype and mutant KRAS

  • Biological approaches:

    • Modulate relative expression levels of RAS isoforms

    • Use gene therapy to increase wildtype HRAS expression in KRAS-mutant tumors

    • Develop transcriptional regulators of HRAS expression

Experimental data shows that wildtype KRAS binds to wildtype HRAS with a mean binding energy of -46.68 kcal/mol, while the G13D KRAS mutant shows significantly stronger binding (-22.04 kcal/mol standardized mean compared to wildtype) . These energy differences provide critical information for designing therapeutics that could mimic or enhance these interactions.

What methodological considerations are important when investigating HRAS in aging-related research?

HRAS has been implicated in aging processes across multiple model organisms, requiring specialized methodological approaches:

Key considerations for aging research involving HRAS:

  • Model selection for aging studies:

    • Short-lived organisms (C. elegans, Drosophila) for rapid aging assessment

    • Cell senescence models (replicative, stress-induced) for cellular aging

    • Progeria models to study accelerated aging

    • Longitudinal studies in longer-lived models for physiological relevance

  • Aging-specific endpoints:

    • Healthspan measures beyond simple lifespan

    • Molecular aging markers (telomere length, epigenetic clocks)

    • Senescence markers (SASP, SA-β-gal, p16INK4a levels)

    • Tissue-specific functional decline metrics

  • Crossover with cancer pathways:

    • Address the paradoxical roles of HRAS in senescence vs. oncogenesis

    • Consider the cell-type specificity of responses

    • Investigate context-dependent outcomes of HRAS activation

  • Integration with other aging pathways:

    • Connect HRAS signaling with established aging mechanisms

    • Study interactions with INS/IGF1 signaling

    • Explore interactions with mTOR and autophagy regulation

Research in C. elegans has demonstrated that the RAS pathway influences both development and aging in conjunction with the insulin/IGF1 signaling pathway . These findings illustrate the evolutionarily conserved role of HRAS in aging processes, suggesting similar mechanisms may be relevant in humans.

How should researchers interpret contradictory data regarding HRAS function across different experimental systems?

Conflicting results regarding HRAS function are common in the literature and require systematic approaches to reconciliation:

Framework for resolving contradictory HRAS data:

  • Identify sources of variation:

    • Cell/tissue type differences (context-dependency)

    • Experimental conditions (growth factors, stress levels)

    • Expression levels (physiological vs. overexpression)

    • Temporal aspects (acute vs. chronic activation)

    • Species differences (human vs. model organisms)

  • Technical validation strategies:

    • Replicate using multiple methodological approaches

    • Validate key findings in diverse cellular contexts

    • Use recombinant expression with controlled levels

    • Apply dose-response relationships rather than single conditions

  • Integrative analysis approaches:

    • Develop computational models that incorporate contextual variables

    • Use systems biology approaches to define network-level effects

    • Consider feedback mechanisms and pathway cross-talk

    • Analyze dynamic rather than steady-state responses

  • Reporting recommendations:

    • Clearly define experimental conditions and cellular context

    • Report cell densities, passage numbers, and culture conditions

    • Include all relevant controls (positive, negative, isotype)

    • Share raw data to enable re-analysis and meta-analysis

For example, in brain degeneration studies, RAS signaling appears detrimental in Drosophila models , while in certain cellular contexts HRAS activation promotes survival. These apparent contradictions likely reflect context-dependent effects and highlight the importance of specifying the precise experimental conditions when interpreting results.

What emerging technologies will advance our understanding of HRAS function in human biology?

Several cutting-edge technologies are poised to transform HRAS research:

  • Spatial multi-omics:

    • Spatial transcriptomics to map HRAS expression within tissues

    • Spatial proteomics to visualize HRAS protein localization and interactors

    • Integration with histopathology for clinicopathological correlations

  • Advanced protein engineering approaches:

    • Optogenetic HRAS variants for spatiotemporal control

    • Split protein complementation for visualization of specific interactions

    • Degron-based systems for rapid protein depletion

  • Single-cell technologies:

    • Single-cell RNA-seq to capture heterogeneity in HRAS expression

    • Single-cell proteomics to measure HRAS activity states

    • Live-cell tracking of HRAS signaling in individual cells over time

  • AI and machine learning applications:

    • Deep learning for structure prediction and dynamics

    • ML algorithms to predict mutation effects

    • Network inference to identify novel interactions

  • Refined computational simulation approaches:

    • Enhanced molecular dynamics simulations with longer timescales

    • Integration of experimental constraints into simulations

    • Quantum mechanics/molecular mechanics (QM/MM) for detailed reaction mechanisms

These technologies will be particularly valuable for understanding the context-dependent nature of HRAS function across different cell types and physiological states.

How can researchers design experiments to distinguish isoform-specific functions of HRAS vs KRAS and NRAS?

Despite high sequence homology, RAS isoforms have distinct functions that require specialized experimental approaches to differentiate:

Experimental design strategies for isoform specificity:

  • Genetic approaches:

    • Isoform-specific knockout/knockdown with rescue experiments

    • Chimeric proteins swapping domains between isoforms

    • Point mutations of isoform-specific residues

    • Domain swapping between isoforms

  • Interaction profiling:

    • BioID or APEX proximity labeling with each isoform as bait

    • Comparative interactomics across all three isoforms

    • Analysis of interaction dynamics following stimulation

    • Identification of isoform-specific adaptor proteins

  • Localization studies:

    • Super-resolution imaging of endogenous isoforms

    • Membrane microdomain analysis

    • Trafficking kinetics following synthesis

    • Response to membrane-targeting drugs

  • Computational approaches:

    • Comparative molecular dynamics simulations

    • Analysis of binding energy differences between isoforms

    • Identification of isoform-specific conformational states

Recent work has revealed that the orientation of wildtype KRAS when bound to wildtype HRAS versus wildtype NRAS differs by 180°, with binding sites that overlap with both the GDP/GTP active site and dimerization interface . These structural differences likely contribute to isoform-specific functions and provide targets for selective intervention.

What standardized reporting guidelines should researchers follow when publishing HRAS-related findings?

To enhance reproducibility and facilitate integration of findings, HRAS researchers should adhere to comprehensive reporting standards:

Recommended reporting elements for HRAS research:

  • Detailed methods documentation:

    • Complete sequence information for constructs used

    • Expression levels relative to endogenous proteins

    • Cell line authentication and mycoplasma testing results

    • Passage numbers and culture conditions

    • Antibody validation methods and catalog numbers

  • Results presentation:

    • Include all controls (positive, negative, loading)

    • Show representative images alongside quantification

    • Report biological and technical replicate numbers

    • Include statistical analysis methods and power calculations

    • Provide raw data in supplementary materials or repositories

  • Contextual information:

    • Cell density and confluency at experiment time

    • Growth factor conditions and serum percentages

    • Time points for all measurements

    • Drug concentrations with exposure durations

    • Consideration of cell cycle effects

  • Computational methods:

    • Software versions and parameters

    • Force fields used in simulations

    • Runtime parameters and convergence criteria

    • Validation metrics for structural predictions

    • Statistical approaches for energy calculations

These standardized reporting practices will not only improve reproducibility but also enable meta-analyses that can resolve apparent contradictions in the literature and accelerate progress in understanding HRAS function in human biology.

Product Science Overview

Discovery and Nomenclature

The HRAS gene was first identified in the early 1960s by J. J. Harvey, who discovered the Harvey strain of murine sarcoma virus (HaMSV) in rats . The viral oncogene, v-Ha-ras, was found to be homologous to the cellular proto-oncogene, c-Ha-ras, which is present in humans. The human homolog of this gene is located on the short arm of chromosome 11 at position 15.5 (11p15.5) .

Function and Mechanism

HRAS encodes a protein known as p21ras, which functions as a molecular switch in various signal transduction pathways. This protein is involved in regulating cell division in response to growth factor stimulation . The HRAS protein binds to guanosine triphosphate (GTP) in its active state and possesses intrinsic GTPase activity, which allows it to hydrolyze GTP to guanosine diphosphate (GDP), thereby turning itself off .

The activation and inactivation of HRAS are tightly regulated by accessory proteins such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). GAPs accelerate the hydrolysis of GTP to GDP, while GEFs facilitate the exchange of GDP for GTP, thus reactivating HRAS .

Role in Cancer

Mutations in the HRAS gene can lead to its constitutive activation, resulting in uncontrolled cell proliferation and tumor formation. Such mutations are implicated in various types of cancers, including bladder cancer, follicular thyroid cancer, and oral squamous cell carcinoma . HRAS is one of the three major Ras genes, along with KRAS and NRAS, that are frequently mutated in human cancers .

Clinical Significance

The HRAS gene is also associated with several genetic disorders. For instance, mutations in HRAS cause Costello syndrome, a condition characterized by distinctive facial features, developmental delays, and an increased risk of tumor formation . Understanding the function and regulation of HRAS is crucial for developing targeted therapies for cancers and other diseases associated with its mutations.

Research and Therapeutic Approaches

Ongoing research aims to develop inhibitors that specifically target the HRAS protein and its downstream signaling pathways. These therapeutic approaches hold promise for treating cancers driven by HRAS mutations. Additionally, recombinant forms of the HRAS protein are used in various experimental settings to study its function and interactions with other cellular proteins.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.