RAN Human

RAN, Member RAS Oncogene Family Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RAN Human

RAN Human (ras-related nuclear protein) is a small GTP-binding protein (25 kDa) encoded by the RAN gene (chromosome 12) in humans. It belongs to the Ras superfamily and plays critical roles in nuclear transport, mitosis, and DNA synthesis regulation . Recombinant RAN Human is produced in E. coli as a non-glycosylated polypeptide (236 amino acids, including a 20-amino-acid His-tag) with a molecular mass of 26.5 kDa . This protein is purified using chromatographic techniques and stored in a buffer containing Tris-HCl, DTT, and glycerol .

Biochemical Properties and Structure

PropertyDetailSource
Molecular Mass26.5 kDa
Amino Acids236 residues (1–216 aa + His-tag)
PurificationProprietary chromatographic techniques
Storage Conditions4°C (short-term), -20°C (long-term) with carrier proteins (HSA/BSA)
GTP/GDP BindingGDP-bound (cytoplasmic) and GTP-bound (nuclear) states regulate activity

RAN Human exists in two nucleotide-bound states: RanGDP (cytoplasmic) and RanGTP (nuclear). The Ran cycle involves nucleotide exchange (RCC1) and hydrolysis (RanGAP/RanBP), creating a RanGTP gradient critical for nuclear transport .

Nuclear Transport

RAN Human regulates the translocation of macromolecules (RNA, proteins) through nuclear pore complexes (NPCs) .

  • Import: RanGTP binds importins, releasing cargo (e.g., transcription factors) into the nucleus .

  • Export: RanGTP binds exportins, facilitating cargo release into the cytoplasm .

Mitosis

RAN Human drives mitotic spindle assembly and chromosome segregation:

  1. RanGTP Gradient: High RanGTP near chromatin activates spindle assembly factors (e.g., TPX2, NuMA) .

  2. Kinetochores: RanGAP/RanBP2 localize to kinetochores, stabilizing spindle-microtubule attachments .

Androgen Receptor Interaction

RAN Human coactivates the androgen receptor (AR), but polyglutamine expansions in AR (linked to Kennedy’s disease) reduce this interaction, causing partial androgen insensitivity .

Clinical Significance in Cancer

RAN Human is overexpressed in multiple cancers, correlating with poor prognosis .

Cancer TypeKey FindingsSource
BreastOverexpression promotes invasion, metastasis; knockdown reduces survival .
LungElevated RAN expression linked to shorter survival .
PancreaticSilencing RAN reduces liver metastases in preclinical models .
GastricHigh RAN levels in adenocarcinomas vs. normal tissue .

Mechanistic Insights:

  • Metastasis: RAN enhances adhesion, colony formation, and invasive potential via pathways like PI3K/MAPK .

  • Therapeutic Target: RAN knockdown sensitizes cells to drugs (e.g., gefitinib) and induces aneuploidy .

Research Applications and Production

Recombinant RAN Human is used in studies to:

  • Model Nuclear Transport: Analyze cargo release mechanisms .

  • Investigate Mitosis: Study spindle assembly defects in cancer .

  • Screen Therapeutics: Identify inhibitors of RanGTP-dependent processes .

RAN Translation in Neurodegenerative Diseases

While not directly linked to RAN Human, RAN translation (bidirectional translation of repeat expansions) produces toxic homopolymeric proteins implicated in ALS/FTD and spinocerebellar ataxia . This highlights RAN’s broader pathological relevance.

Product Specs

Introduction
RAN, a GTP-binding protein, plays a crucial role in nucleocytoplasmic transport by facilitating protein import into the nucleus and RNA export. It is also involved in chromatin condensation and cell cycle regulation. During mitosis, RAN interacts with BIRC5 to form a complex that acts as a scaffold, aiding in the delivery of the RAN effector molecule TPX2 to microtubules, thereby contributing to mitotic spindle formation. Furthermore, RUN enhances AR-mediated transactivation.
Description
Recombinant Human RAN, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 236 amino acids (with a sequence spanning from amino acid position 1 to 216) and possessing a molecular weight of 26.5 kDa. A 20 amino acid His-Tag is fused to the N-terminus of the RAN protein to facilitate purification, which is achieved through proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution that has been filtered for sterility.
Formulation
The provided solution of Human RAN is formulated in a buffer containing 20mM Tris-HCl at pH 8, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. It's important to avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of the protein is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
ARA24, Gsp1, TC4, GTP-binding nuclear protein Ran, GTPase Ran, Ras-related nuclear protein, Ras-like protein TC4, Androgen receptor-associated protein 24, Ras-like, RANP1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MAAQGEPQVQ FKLVLVGDGG TGKTTFVKRH LTGEFEKKYV ATLGVEVHPL VFHTNRGPIK FNVWDTAGQE KFGGLRDGYY IQAQCAIIMF DVTSRVTYKN VPNWHRDLVR VCENIPIVLC GNKVDIKDRK VKAKSIVFHR KKNLQYYDIS AKSNYNFEKP FLWLARKLIG DPNLEFVAMP ALAPPEVVMD PALAAQYEHD LEVAQTTALP DEDDDL.

Q&A

What is RAN protein and what is its primary function in human cells?

RAN (Ras-related Nuclear protein), also known as GTP-binding nuclear protein Ran, is a small 25 kDa protein encoded by the RAN gene in humans. It serves as a critical regulator of nucleocytoplasmic transport and plays essential roles during both interphase and mitosis. As a member of the Ras superfamily of small G proteins, RAN exists in two nucleotide-bound states—GDP-bound and GTP-bound—which are essential for its biological functions. The protein's primary role involves facilitating the translocation of RNA and proteins through the nuclear pore complex, effectively serving as a molecular switch that determines directional transport across the nuclear envelope .

RAN's functionality extends beyond nuclear transport to include involvement in DNA synthesis regulation and cell cycle progression. Mutations in the RAN gene have been found to disrupt DNA synthesis, highlighting its significance in maintaining cellular homeostasis. The protein maintains a concentration gradient between the nucleus and cytoplasm, with higher concentrations typically found within the nucleus compared to cytoplasmic regions .

How does the RAN cycle operate to regulate nuclear transport?

The RAN cycle operates through a series of precisely controlled molecular interactions that create and maintain nucleocytoplasmic gradients. This cycle involves the interconversion between RAN's GDP-bound and GTP-bound states through the activities of specific regulatory proteins:

  • RCC1 (Regulator of Chromosome Condensation 1), also known as RanGEF (Ran Guanine nucleotide Exchange Factor), converts RanGDP to RanGTP inside the nucleus.

  • RanGAP (Ran GTPase Activating Protein), facilitated by Ran-binding protein (RanBP), activates Ran's intrinsic GTPase activity, converting RanGTP back to RanGDP primarily in the cytoplasm.

  • NUTF2 (Nuclear Transport Factor 2) imports cytoplasmic RanGDP into the nucleus to continue the cycle .

This system generates a steep concentration gradient with high RanGTP levels inside the nucleus and low levels in the cytoplasm. For nuclear import, cargo proteins containing nuclear localization signals (NLS) bind to importins and enter the nucleus. Once inside, RanGTP binds to importin, causing it to release the cargo. Conversely, for nuclear export, cargo binds to exportin along with RanGTP in a ternary complex that exits the nucleus. Upon GTP hydrolysis in the cytoplasm, the complex dissociates, releasing the export cargo .

What is RAN translation and how does it differ from canonical translation?

RAN (Repeat-Associated Non-AUG) translation represents a non-canonical protein synthesis mechanism discovered in 2011 by the Ranum laboratory. Unlike conventional translation, which requires the canonical AUG start codon, RAN translation initiates protein synthesis at repeat expansions without this canonical start signal. This process produces unexpected toxic repetitive proteins that have been implicated in various neurodegenerative disorders .

The key differences between RAN translation and canonical translation include:

  • Initiation site: RAN translation occurs at repeat expansions rather than requiring an AUG start codon

  • Products: RAN translation generates repetitive proteins that often form toxic aggregates

  • Regulation: RAN translation appears to bypass traditional translational control mechanisms

  • Disease association: RAN translation has been linked to a growing number of repeat expansion disorders

RAN proteins have been detected in autopsy samples from patients with diseases like myotonic dystrophy type 2, C9-ALS/FTD, and more recently, Friedreich's ataxia (FA), where they form aggregates in the cerebellum and spinal cord .

How do researchers design experiments to study RAN protein dynamics?

Researchers employ multiple sophisticated experimental approaches to investigate RAN protein dynamics in cellular contexts. A comprehensive experimental design typically involves several key steps:

  • Defining Variables: Researchers must clearly identify independent variables (e.g., RAN protein concentration, mutation status, cellular conditions) and dependent variables (e.g., nuclear transport rates, mitotic progression, protein interactions) . For example, in studies examining RAN's role in nuclear transport, researchers might manipulate RanGTP/GDP ratios while measuring cargo movement across the nuclear envelope.

  • Hypothesis Formulation: Well-designed RAN experiments require clear null and alternative hypotheses. For instance, H₀: "Mutation X in RAN does not affect its interaction with importin-β" versus H₁: "Mutation X in RAN significantly reduces its binding affinity for importin-β" .

  • Treatment Design: Experimental treatments must systematically manipulate RAN-related variables. This might include creating concentration gradients of wild-type versus mutant RAN proteins, altering the levels of RAN regulatory factors (RCC1, RanGAP), or introducing specific inhibitors of the RAN cycle .

Live-cell imaging techniques using fluorescently tagged RAN constructs allow researchers to track RAN protein movement and interactions in real time. Biochemical approaches, including co-immunoprecipitation and pull-down assays, help identify RAN-interacting proteins. Additionally, structural studies employing X-ray crystallography and cryo-electron microscopy provide atomic-level insights into RAN conformational changes during nucleotide cycling.

What methodological challenges exist in studying RAN protein-mediated nuclear transport?

Investigating RAN-mediated nuclear transport presents several significant methodological challenges:

  • Maintaining physiological relevance: Creating experimental systems that accurately reflect the natural RanGTP/GDP gradient can be difficult. In vitro systems may not fully recapitulate the complex cellular environment.

  • Temporal resolution limitations: The rapid cycling of RAN between GDP and GTP-bound states occurs on timescales that challenge current imaging technologies.

  • Spatial resolution constraints: Distinguishing RAN activities at the nuclear pore complex versus other cellular locations requires specialized high-resolution microscopy techniques.

  • Distinguishing direct versus indirect effects: Manipulating RAN often affects multiple cellular processes simultaneously, making it difficult to isolate specific transport effects from broader cellular responses.

  • Quantitative measurements: Accurately quantifying RAN gradient steepness and transport rates requires sophisticated biophysical approaches and mathematical modeling.

Researchers overcome these challenges by combining multiple complementary techniques, including FRAP (Fluorescence Recovery After Photobleaching), single-molecule tracking, and computational modeling of transport kinetics. Developing cell-free systems that reconstitute nuclear transport with purified components also helps isolate specific mechanisms.

How does RAN protein contribute to mitotic spindle assembly?

RAN protein plays crucial roles during mitosis, particularly in mitotic spindle assembly and nuclear envelope dynamics. During prophase, as the nuclear envelope breaks down, the steep RanGTP-RanGDP gradient dissipates, but RanGTP concentration remains high around chromosomes because RCC1 (the nucleotide exchange factor) remains attached to chromatin .

The role of RAN in mitotic spindle assembly involves several mechanisms:

  • Releasing spindle assembly factors: RanGTP releases spindle assembly factors like NuMA and TPX2 from inhibitory complexes with importins, thereby activating them to promote spindle formation.

  • Regulating kinetochore-microtubule attachments: RanBP2 (Nup358) and RanGAP relocate to kinetochores during mitosis where they facilitate the proper attachment of spindle fibers to chromosomes.

  • Controlling microtubule nucleation: RanGTP promotes microtubule nucleation and stabilization around chromosomes, contributing to the formation of the spindle apparatus.

  • Nuclear envelope reassembly: During telophase, RanGTP hydrolysis and nucleotide exchange are required for vesicle fusion events that reform the nuclear envelopes of daughter nuclei .

Experimental approaches to study these processes include live-cell imaging of fluorescently tagged RAN proteins, immunofluorescence microscopy of mitotic cells, cell-free extracts that recapitulate spindle assembly, and targeted perturbations of RAN pathway components using RNAi or specific inhibitors.

What experimental evidence supports the role of RAN proteins in human disorders?

The involvement of RAN proteins in human disorders is supported by several lines of experimental evidence:

  • Detection in patient tissues: Using specific antibodies, researchers have identified RAN protein aggregates in autopsy samples from patients with various repeat expansion disorders. In Friedreich's ataxia (FA), RAN aggregates have been found in the cerebellum and spinal cord of patient samples (n>5) but not in control samples (n>4) .

  • Correlation with disease pathology: The distribution of RAN protein aggregates often correlates with regions showing neurodegeneration in repeat expansion disorders.

  • Cellular toxicity studies: In vitro experiments demonstrate that RAN proteins can be toxic to cells, disrupting cellular functions and potentially contributing to disease pathogenesis.

  • Animal models: Transgenic animal models expressing repeat expansions that produce RAN proteins often recapitulate disease features, supporting their pathogenic role.

The Ranum laboratory has developed antibodies that specifically recognize the repetitive sequences in RAN proteins, enabling the detection of these proteins in human tissues and experimental models. This technical advance has been crucial for establishing the presence of RAN proteins in multiple repeat expansion disorders .

How do researchers experimentally detect and analyze RAN proteins?

Detecting and analyzing RAN proteins in experimental settings requires specialized methodologies:

  • Specific antibodies: Researchers develop antibodies that recognize the repetitive amino acid sequences produced by RAN translation. These are critical tools for immunohistochemistry, immunofluorescence, and Western blot analyses.

  • Reporter assays: Experimental constructs containing repeat expansions fused to reporter genes (like luciferase or fluorescent proteins) help quantify RAN translation in cellular models.

  • Mass spectrometry: This technique identifies RAN proteins and characterizes their post-translational modifications in cellular or tissue samples.

  • Cellular models: Patient-derived cells (such as fibroblasts, induced pluripotent stem cells, or neuronal cultures) expressing repeat expansions serve as platforms to study RAN protein production and effects.

  • Pulse-chase labeling: Using radioactive or stable isotope-labeled amino acids helps track the synthesis and turnover rates of RAN proteins.

The Ranum laboratory has successfully developed methods to inhibit RAN translation, which opens opportunities for therapeutic interventions. This approach allows researchers to reduce RAN protein levels in disease models and determine whether this ameliorates cellular pathology .

What therapeutic strategies are being explored to target RAN translation in human disorders?

Research into therapeutic strategies targeting RAN translation represents an emerging frontier in treating repeat expansion disorders. Current approaches under investigation include:

  • Antisense oligonucleotides (ASOs): These target the repeat-containing RNA to prevent RAN translation without affecting canonical gene expression.

  • Small molecule inhibitors: Compounds that selectively inhibit the non-canonical translation machinery involved in RAN translation are being explored.

  • Genetic approaches: CRISPR-Cas9 and related technologies may be employed to edit or remove the repeat expansions responsible for RAN translation.

  • RNA-binding proteins: Manipulating the binding of regulatory proteins to repeat-containing RNAs could potentially modulate RAN translation.

  • Degradation of RAN proteins: Enhancing cellular mechanisms for clearing toxic RAN protein aggregates represents another potential therapeutic avenue.

The long-term goal of such research is to understand how RAN proteins contribute to diseases like Friedreich's ataxia and to develop targeted therapeutic strategies. The Ranum laboratory is exploring both genetic and pharmacological approaches to reduce RAN proteins in FA models to determine if lowering RAN protein levels improves cellular phenotypes associated with the disease .

How is RAN research integrated into large-scale human cell mapping initiatives?

RAN research is increasingly being integrated into comprehensive human cell mapping initiatives, particularly through projects like the Human Reference Atlas (HRA). The HRA represents a collaborative effort by experts from 18 consortia to map the approximately 37 trillion cells in the healthy human body . This atlas incorporates multi-omics data that can provide insights into RAN protein distribution and function across different cell types and tissues.

The HRA utilizes information from multiple sources:

  • Expert knowledge

  • Published literature

  • Experimental datasets

The HRA v1.4 release contains detailed information on 31 organs with 4,279 unique anatomical structures, 1,210 unique cell types, and 2,089 unique biomarkers, creating a framework where RAN protein expression and function can be contextualized within specific cellular and anatomical locations . This comprehensive mapping approach helps researchers understand tissue-specific variations in RAN function and potentially identify regions particularly susceptible to RAN-related pathologies.

What computational approaches are used to analyze RAN protein interactions?

Computational approaches play a crucial role in understanding RAN protein interactions and functions across different cellular contexts. Researchers employ various bioinformatic and modeling techniques:

  • Network analysis: Protein-protein interaction networks help identify RAN binding partners and characterize the broader impact of RAN on cellular signaling pathways.

  • Structural modeling: Molecular dynamics simulations predict how RAN protein conformational changes during GTP/GDP cycling affect its interactions with transport receptors and other binding partners.

  • Transcriptomic integration: Analysis of RNA-seq data across tissues helps identify genes co-regulated with RAN and potential downstream effectors.

  • Machine learning approaches: These methods predict RAN interaction sites on target proteins and identify potential regulatory mechanisms.

  • Systems biology modeling: Mathematical models of the RAN gradient and nuclear transport incorporate diffusion, binding kinetics, and compartmentalization to predict system behavior under different conditions.

The HRAlit database linking HRA digital objects to publications, experts, funded projects, and experimental datasets provides a valuable resource for these computational approaches. With 22 tables containing 20,939,937 records and 13,170,651 relationships, this database offers rich opportunities for mining RAN-related information .

What experimental design considerations are crucial for studying RAN translation in human tissues?

Studying RAN translation in human tissues presents unique experimental design challenges that researchers must address:

  • Tissue selection and procurement: Researchers must carefully select tissues known to be affected in repeat expansion disorders and ensure proper procurement and preservation to maintain RNA and protein integrity.

  • Controls: Appropriate controls from age-matched, non-disease individuals are essential for comparative analyses.

  • Antibody validation: Thorough validation of antibodies against RAN proteins is critical, as these repetitive proteins can present challenges for specific detection.

  • Quantification methods: Developing reliable quantification approaches for RAN proteins in complex tissue samples requires careful optimization.

  • Regional analysis: Since RAN protein aggregation may vary across anatomical regions, comprehensive mapping of their distribution is important.

  • Correlation with pathology: Experimental designs should include methods to correlate RAN protein presence with markers of cellular dysfunction or tissue pathology.

When designing these experiments, researchers should follow a structured approach that includes:

  • Clearly defining research questions and hypotheses

  • Identifying independent and dependent variables

  • Controlling for extraneous variables

  • Determining appropriate sample sizes

  • Planning for statistical analyses that can address potential heterogeneity in human samples

What emerging technologies are advancing the study of RAN protein dynamics?

Several cutting-edge technologies are revolutionizing how researchers study RAN protein dynamics:

  • Super-resolution microscopy: Techniques like STORM, PALM, and STED microscopy enable visualization of RAN protein localization and interactions at nanometer resolution, providing unprecedented insights into its spatial organization at nuclear pores and during mitosis.

  • Live-cell single-molecule tracking: These approaches allow researchers to follow individual RAN molecules in real-time, revealing the kinetics and trajectories of RAN movement between cellular compartments.

  • Optogenetic tools: Light-controllable versions of RAN pathway components enable precise spatiotemporal manipulation of RAN function in living cells.

  • CRISPR-based approaches: CRISPR interference and activation systems provide new ways to modulate RAN expression with high specificity, while CRISPR knock-in strategies facilitate endogenous tagging of RAN proteins.

  • Proximity labeling: Techniques like BioID and APEX2 identify proteins in close proximity to RAN in living cells, expanding our understanding of its interaction network.

  • Cryo-electron tomography: This method reveals the three-dimensional organization of RAN-containing complexes in near-native states within cells.

  • AI-assisted image analysis: Machine learning algorithms enhance the detection and quantification of RAN distribution patterns in complex cellular environments.

These technologies are helping researchers overcome traditional limitations in studying the rapidly cycling and dynamic RAN system, providing new mechanistic insights into its diverse cellular functions.

How might understanding RAN mechanisms contribute to therapeutic development?

Understanding RAN mechanisms—both the RAN protein cycle and RAN translation—holds significant promise for therapeutic development across multiple disease contexts:

  • Cancer therapeutics: Since RAN is often dysregulated in cancer and plays crucial roles in mitosis, targeting the RAN cycle could disrupt cancer cell division. Inhibitors of RAN-dependent nuclear transport might prevent cancer cells from importing transcription factors or exporting tumor suppressors.

  • Neurodegenerative disease treatments: For repeat expansion disorders involving toxic RAN proteins, therapeutic strategies include:

    • Antisense oligonucleotides to block RAN translation

    • Small molecules that inhibit the non-canonical translation machinery

    • Approaches to enhance clearance of toxic RAN protein aggregates

  • Anti-viral applications: Since many viruses depend on nuclear transport for replication, modulating the RAN cycle could disrupt viral life cycles.

  • Androgen receptor-related disorders: Given RAN's role as an androgen receptor coactivator (ARA24), understanding this interaction could inform treatments for conditions like spinal and bulbar muscular atrophy (Kennedy's disease) .

The Ranum laboratory has already made progress in developing methods to inhibit RAN translation, demonstrating that genetic and pharmacological approaches can reduce RAN proteins in disease models. Future work will determine if these interventions improve disease-related cellular phenotypes, potentially paving the way for clinical applications .

What knowledge gaps remain in our understanding of RAN functions in human biology?

Despite significant advances, several important knowledge gaps remain in our understanding of RAN functions:

  • Tissue-specific regulation: How RAN protein expression, localization, and function vary across different human tissues and cell types remains incompletely characterized. The Human Reference Atlas efforts may help address this gap .

  • Developmental dynamics: Changes in RAN function during human development and aging require further investigation, particularly in the context of tissue-specific differentiation programs.

  • Interaction with disease mechanisms: The precise mechanisms by which RAN protein dysfunction or RAN translation contribute to disease pathogenesis need further elucidation.

  • Regulatory mechanisms: The factors controlling RAN cycle kinetics and how these are modified under different cellular conditions (stress, differentiation, disease) remain to be fully characterized.

  • Non-canonical functions: Potential roles of RAN beyond nuclear transport and mitosis, such as in cytoplasmic organelle organization or RNA metabolism, warrant investigation.

  • Therapeutic targeting specificity: Developing approaches that selectively target disease-relevant aspects of RAN function without disrupting essential cellular processes remains challenging.

  • RAN translation initiation: The precise molecular mechanisms initiating RAN translation at repeat expansions are not completely understood, limiting our ability to selectively target this process.

Addressing these knowledge gaps will require interdisciplinary approaches combining structural biology, cell biology, genetics, computational biology, and clinical research. The development of more precise tools to manipulate RAN function in specific contexts will be essential for progress in this field.

Product Science Overview

Structure and Function

RAN is a 25 kDa protein encoded by the RAN gene. It exists in two nucleotide-bound forms: GDP-bound (RanGDP) and GTP-bound (RanGTP). The conversion between these forms is regulated by RCC1 (Ran Guanine nucleotide Exchange Factor) and RanGAP (Ran GTPase Activating Protein) . This conversion is essential for the directionality of nucleocytoplasmic transport, which is regulated by GTP hydrolysis .

Role in Cellular Processes
  1. Nucleocytoplasmic Transport: RAN is essential for the translocation of RNA and proteins through the nuclear pore complex. It shuttles between the nucleus and the cytoplasm, ensuring the proper transport of macromolecules .
  2. DNA Synthesis and Cell Cycle Progression: RAN is involved in the control of DNA synthesis and cell cycle progression. Mutations in RAN can disrupt DNA synthesis, highlighting its importance in maintaining cellular functions .
  3. Microtubule Organization: RAN regulates the formation and organization of the microtubule network independently of its role in nucleocytoplasmic transport. It is a key signaling molecule that regulates microtubule polymerization during mitosis .
Clinical Significance

RAN is an androgen receptor (AR) coactivator that binds differentially with different lengths of polyglutamine within the androgen receptor. Polyglutamine repeat expansion in the AR is linked to Kennedy’s disease (X-linked spinal and bulbar muscular atrophy). The coactivation of AR by RAN diminishes with polyglutamine expansion, potentially leading to partial androgen insensitivity during the development of Kennedy’s disease .

Recombinant RAN Protein

Recombinant RAN protein is used in various research applications to study its function and role in cellular processes. It is produced using recombinant DNA technology, which allows for the expression of the RAN protein in a controlled environment .

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.