NDRG3 Human

N-Myc Downstream Regulated 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles and Signaling Pathways

NDRG3 regulates diverse cellular processes through non-catalytic mechanisms:

Oncogenic and Anti-Apoptotic Functions

  • Hypoxia Responses: Mediates lactate-induced hypoxia signaling via the NDRG3-Raf-ERK pathway, promoting survival in prostate cancer .

  • Wnt/β-Catenin Regulation: Overexpression in hepatocellular carcinoma (HCC) enhances β-catenin nuclear accumulation, driving drug resistance and proliferation .

  • Imatinib Resistance: In chronic myeloid leukemia (CML), NDRG3 overexpression elevates IC50 for imatinib, linked to β-catenin stabilization and cell cycle progression .

Immune Cell Regulation

  • T-Cell Maturation: Critical for peripheral T-cell homeostasis, particularly CD8+ subsets. Deficiency reduces naïve T cells and skews toward effector memory phenotypes .

  • Phosphorylation-Dependent Signaling: S331 phosphorylation shifts lymphoid progenitor fate toward B cells (S331E mutant) or T cells (S331A mutant) .

Reproductive and Developmental Roles

  • Meiosis Regulation: Required for DNA double-strand break (DSB) repair during spermatogenesis. Haploinsufficiency causes subfertility in males .

  • Testicular Expression: Highest expression observed in germ cells, peaking during pachytene stages .

Therapeutic Targets

  • Imatinib Resistance: NDRG3 knockdown restores drug sensitivity in CML by reducing β-catenin levels .

  • Hypoxia-Induced Pathways: Targeting NDRG3-Raf-ERK axis may suppress tumor progression in hypoxic microenvironments .

Experimental Models

ModelObservationSource
Ndrg3 TKO MiceReduced CD8+ T-cell survival, skewed memory subsets
NDRG3-S331A Transgenic MiceAccelerated thymocyte maturation, T-cell predominance
K/G Imatinib-Resistant CellsNDRG3 knockdown + imatinib reduces tumor growth in xenografts

Product Specs

Introduction
N-Myc Downstream Regulated 3, also known as NDRG3, is a protein coding gene that belongs to the NDRG family. It is primarily expressed in the brain. NDRG2 is a significant paralog of NDRG3.
Description
Recombinant human NDRG3, produced in E. coli, is a single polypeptide chain consisting of 386 amino acids (residues 1-363) with a molecular weight of 42.4 kDa. The NDRG3 protein has a 23 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution that has been filtered for sterilization.
Formulation
The NDRG3 solution is provided at a concentration of 0.25mg/ml in a buffer containing Phosphate buffered saline (pH 7.4), 20% glycerol, and 1mM DTT.
Stability
The product can be stored at 4°C for 2-4 weeks if the entire vial will be used within that time frame. For longer-term storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid subjecting the product to multiple freeze-thaw cycles.
Purity
The purity of the NDRG3 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Protein NDRG3, N-myc downstream-regulated gene 3 protein, NDRG3, N-Myc Downstream Regulated 3.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMDELQDV QLTEIKPLLN DKEHDIETTH GVVHVTIRGL PKGNRPVILT YHDIGLNHKS CFNAFFNFED MQEITQHFAV CHVDAPGQQE GAPSFPTGYQ YPTMDELAEM LPPVLTHLSL KSIIGIGVGA GAYILSRFAL NHPELVEGLV LINVDPCAKG WIDWAASKLS GLTTNVVDII LAHHFGQEEL QANLDLIQTY RMHIAQDINQ DNLQLFLNSY NGRRDLEIER PILGQNDNKS KTLKCSTLLV VGDNSPAVEA VVECNSRLNP INTTLLKMAD CGGLPQVVQP GKLTEAFKYF LQGMGYIPSA SMTRLARSRT HSTSSSLGSG ESPFSRSVTS NQSDGTQESC ESPDVLDRHQ TMEVSC.

Q&A

What is NDRG3 and to which protein family does it belong?

NDRG3 (N-myc downstream-regulated gene 3) is a member of the NDRG family that consists of four paralogs: NDRG1, NDRG2, NDRG3, and NDRG4. These proteins are involved in various cellular processes including cell proliferation, differentiation, and development. The NDRG family has structural similarities but demonstrates distinct tissue expression patterns and biological functions . NDRG3 specifically shows enriched expression in testis and prostate tissues, suggesting tissue-specific functions that differ from other family members .

For researchers beginning to study NDRG3, it is advisable to first examine the evolutionary conservation of this protein across species and its structural relationship to other NDRG family members. Comparative sequence analysis can provide insights into functional domains that may guide hypothesis generation about NDRG3's biochemical activities.

What are the primary tissues where NDRG3 is expressed in humans?

NDRG3 demonstrates tissue-specific expression patterns in humans, with enrichment particularly in the testis and prostate. This has been demonstrated through gene expression data derived from massively parallel signature sequencing from 33 different human organs . Beyond these primary sites, NDRG3 is also expressed in other tissues including liver and breast tissue, albeit at different levels .

Methodologically, researchers can detect and quantify NDRG3 expression using several approaches:

  • qRT-PCR for mRNA expression analysis

  • Immunohistochemistry (IHC) for protein localization in tissues

  • Western blotting for protein expression quantification

  • RNA-seq for transcript variant identification and expression profiling

When designing expression studies, researchers should consider including multiple detection methods to validate findings and include appropriate positive control tissues (testis/prostate) to benchmark expression levels.

How is NDRG3 expression regulated in normal and pathological conditions?

NDRG3 expression appears to be regulated through multiple mechanisms. In prostate cancer cells, NDRG3 has been shown to be androgen-regulated . The regulation patterns of NDRG3 differ from other family members—for instance, while NDRG1 is significantly up-regulated by androgen in LNCaP cells, NDRG3 shows distinct regulatory patterns .

In pathological conditions like cancer, NDRG3 expression is frequently dysregulated. For example, NDRG3 expression was detected in 58.6% (41/70) of prostate cancer specimens compared to only 13.2% (5/38) of benign prostatic hyperplasia specimens . In hepatocellular carcinoma (HCC), NDRG3 shows significantly higher expression in tumor tissues compared to non-tumor tissues .

Experimental approaches to study NDRG3 regulation should include:

  • Promoter analysis to identify transcription factor binding sites

  • Chromatin immunoprecipitation (ChIP) to confirm transcription factor binding

  • Luciferase reporter assays to assess promoter activity under various conditions

  • Analysis of epigenetic modifications that may influence NDRG3 expression

What is the role of NDRG3 in cancer progression and metastasis?

NDRG3 appears to function as a tumor promoter in several cancer types. In prostate cancer, over-expression of NDRG3 in stably transfected PC-3 cells increased their growth rates and migration capabilities compared to parental or mock empty vector transfected PC-3 cells . Additionally, overexpression of NDRG3 promoted the growth of xenograft tumors in nude mice, supporting its role in cancer progression in vivo .

Mechanistically, NDRG3 overexpression up-regulates the expression of multiple angiogenic chemokines including CXCL1 (chemokine ligand 1), CXCL3 (chemokine ligand 3), and CXCL5 (chemokine ligand 5), which may enhance tumor angiogenesis . This suggests that NDRG3 may influence the tumor microenvironment to facilitate cancer progression.

To investigate NDRG3's role in cancer progression, researchers should consider:

  • Gene knockdown and overexpression studies in relevant cell lines

  • Migration and invasion assays to assess metastatic potential

  • Angiogenesis assays to evaluate effects on blood vessel formation

  • Analysis of downstream signaling pathways using phospho-specific antibodies

How does NDRG3 expression correlate with clinical outcomes in different cancer types?

NDRG3 expression has shown significant correlation with clinical outcomes across multiple cancer types:

In hepatocellular carcinoma (HCC):

In invasive breast cancer (IBC):

For clinical correlation studies, researchers should employ:

  • Tissue microarrays for high-throughput analysis

  • Multivariate Cox regression analysis to account for confounding variables

  • Kaplan-Meier survival analysis to visualize survival differences

  • Stratification by molecular subtypes to identify context-dependent effects

What molecular mechanisms underlie NDRG3's effects on tumor cell behavior?

The molecular mechanisms through which NDRG3 influences tumor cell behavior involve several signaling pathways:

  • Angiogenesis promotion: NDRG3 overexpression upregulates angiogenic chemokines (CXCL1, CXCL3, CXCL5), which may increase tumor angiogenesis and provide a favorable microenvironment for tumor growth .

  • Cell proliferation and migration: In prostate cancer cells, NDRG3 overexpression enhances growth rates and migration capabilities, suggesting involvement in cell cycle regulation and cytoskeletal organization .

  • Potential involvement in PI3K-Akt signaling: While not directly demonstrated for NDRG3, other NDRG family members like NDRG2 regulate PI3K-Akt signaling in T cells by binding to protein phosphatase 2A and promoting PTEN activity . NDRG1b has been shown to suppress overactivation of the PI3k-Akt pathway . Given their structural similarities, NDRG3 may also influence these pathways.

Recommended experimental approaches:

  • Phosphoproteomics to identify changes in signaling cascade components

  • Co-immunoprecipitation to identify protein interaction partners

  • RNA-seq analysis following NDRG3 modulation to identify transcriptional targets

  • CRISPR-Cas9 knockout studies to establish causality in observed phenotypes

What is the function of NDRG3 in T cell development and homeostasis?

Recent research has identified NDRG3 as a critical regulator of peripheral T cell maturation and homeostasis. In Ndrg3-deficient mice (Ndrg3TKO), significant changes were observed in peripheral T cell populations:

  • Reduced total numbers of CD4+ and CD8+ T cells in the spleen

  • Altered distribution of naïve (CD44lowCD62L+), effector memory (EM; CD44highCD62L−), and central memory (CM; CD44highCD62L+) T cell subsets

  • More pronounced effects on CD8+ T cells than CD4+ cells, suggesting differential sensitivity to Ndrg3 loss

  • Decreased absolute numbers of all T cell subsets (naïve, EM, and CM) with particularly strong reduction in naïve cells and relative increase in EM T cells

Ndrg3 appears to be dispensable for thymic development but crucial for peripheral T cell homeostasis. This suggests a role in regulating T cell survival, proliferation, or differentiation in secondary lymphoid organs.

To study NDRG3's immunological functions, researchers should consider:

  • Conditional knockout models with T cell-specific deletion

  • Adoptive transfer experiments to assess cell-intrinsic defects

  • Competitive bone marrow chimeras to evaluate fitness relative to wild-type cells

  • In vitro T cell activation and proliferation assays to assess functional responses

How does NDRG3 influence T cell signaling pathways?

NDRG3 appears to be involved in modulating key signaling pathways in T cells:

  • TCR signal integration: Ndrg3 may be required for naïve T cells to successfully integrate TCR and cytokine signals, especially when TCR avidity is low . This suggests a role in fine-tuning signal transduction downstream of the TCR complex.

  • Potential involvement in PI3K-Akt signaling: While direct evidence for NDRG3's role is limited, other NDRG family members regulate this pathway in T cells. Given their structural similarities, NDRG3 may function similarly in modulating PI3K-Akt signaling .

  • Possible role in cytokine responsiveness: The altered proportions of memory-phenotype cells in Ndrg3-deficient mice suggest potential involvement in cytokine-mediated homeostatic signals that maintain the peripheral T cell pool .

Recommended experimental approaches:

  • Phospho-flow cytometry to assess activation of signaling components

  • Calcium flux assays to evaluate early TCR signaling events

  • Analysis of transcription factor activation (e.g., NFAT, NF-κB, AP-1)

  • Measurement of cytokine production and responsiveness

What are the optimal methods for detecting and quantifying NDRG3 expression in human samples?

Multiple complementary approaches are recommended for comprehensive analysis of NDRG3 expression:

  • Transcriptomic analysis:

    • qRT-PCR: Allows specific quantification of NDRG3 mRNA levels with appropriate reference genes

    • RNA-seq: Provides comprehensive transcriptome data, enabling analysis of different transcript variants

    • Advantages: High sensitivity, ability to detect transcript variants

    • Limitations: May not correlate perfectly with protein expression

  • Protein detection:

    • Immunohistochemistry (IHC): Enables visualization of NDRG3 localization in tissue sections and semi-quantitative analysis

    • Western blotting: Allows quantification of total protein levels

    • Immunofluorescence: Provides subcellular localization information

    • Advantages: Direct detection of the functional protein

    • Limitations: Antibody specificity concerns, challenges in quantification

  • Validation strategies:

    • Include positive control tissues (testis/prostate) known to express NDRG3

    • Employ multiple antibodies targeting different epitopes

    • Use NDRG3 knockout/knockdown samples as negative controls

    • Compare results across multiple detection methods

  • Data analysis considerations:

    • For IHC, establish clear scoring criteria (e.g., H-score, proportion of positive cells)

    • For qRT-PCR, select stable reference genes appropriate for the tissue being analyzed

    • For RNA-seq, consider normalized metrics like FPKM or TPM for cross-sample comparisons

What are effective genetic manipulation strategies to study NDRG3 function in vitro and in vivo?

Several genetic manipulation strategies can be employed to investigate NDRG3 function:

  • In vitro approaches:

    • siRNA/shRNA: Transient or stable knockdown of NDRG3 expression

    • CRISPR-Cas9: Generation of NDRG3 knockout cell lines or specific mutations

    • Overexpression systems: Plasmid-based or viral vector-mediated NDRG3 overexpression

    • Advantages: Relatively quick, cost-effective, easily applicable across cell lines

    • Limitations: Potential off-target effects, incomplete knockdown, non-physiological expression levels

  • In vivo approaches:

    • Conditional knockout mice: Tissue-specific deletion using Cre-loxP system (as demonstrated with pLck-Cre for T cell-specific deletion)

    • Transgenic overexpression models: Tissue-specific NDRG3 overexpression

    • CRISPR-based in vivo editing: Direct mutation of NDRG3 in adult tissues

    • Advantages: Physiological context, allows study of systemic effects

    • Limitations: Time-consuming, expensive, potential developmental compensation

  • Validation strategies:

    • Rescue experiments to confirm specificity

    • Use of multiple targeting sequences to minimize off-target effects

    • Careful selection of control conditions (empty vector, non-targeting guide RNA)

  • Specialized approaches:

    • Structure-function studies using deletion mutants or point mutations

    • Domain-swapping with other NDRG family members to identify functional domains

    • Inducible expression systems to study temporal effects

How do the functions of NDRG3 compare with other NDRG family members, and what are the implications for targeted therapies?

The NDRG family shows both overlapping and distinct functions:

  • Comparative expression patterns:

    • NDRG1: Widely expressed, particularly in epithelial cells

    • NDRG2: Primarily in brain, heart, and skeletal muscle

    • NDRG3: Enriched in testis and prostate

    • NDRG4: Predominantly in brain and heart

  • Functional distinctions in cancer:

    • NDRG1: Generally considered a tumor suppressor in multiple cancers

    • NDRG2: Predominantly tumor suppressive

    • NDRG3: Appears to function as a tumor promoter in prostate cancer, HCC, and breast cancer

    • NDRG4: Varied roles depending on cancer type

  • Signaling pathway involvement:

    • NDRG family members modulate PI3K-Akt signaling, but potentially through different mechanisms

    • Differential regulation by upstream factors (e.g., androgens regulate NDRG1 and NDRG3 differently)

  • Therapeutic implications:

    • The tumor-promoting role of NDRG3 makes it a potential therapeutic target in certain cancers

    • Selective targeting would be necessary to avoid interfering with potentially tumor-suppressive functions of other family members

    • Understanding the structural and functional differences between NDRG proteins could guide the development of specific inhibitors

Research strategies should include:

  • Comparative expression analysis across family members in the same tissues

  • Rescue experiments to determine functional redundancy

  • Structural biology approaches to identify unique binding sites for selective targeting

What is the role of NDRG3 in the tumor microenvironment and how might this influence immunotherapy responses?

NDRG3's influence on the tumor microenvironment represents an emerging area of investigation:

  • Angiogenesis regulation:

    • NDRG3 overexpression upregulates angiogenic chemokines (CXCL1, CXCL3, CXCL5)

    • Enhanced angiogenesis could promote tumor growth and potentially drug resistance

  • Potential immune modulation:

    • Given NDRG3's role in T cell homeostasis , its expression in tumors might influence tumor-infiltrating lymphocyte function

    • The altered distribution of naïve and memory T cells in Ndrg3-deficient mice suggests potential impacts on anti-tumor immune responses

    • CXCL chemokines induced by NDRG3 can attract specific immune cell populations, potentially shaping the immune microenvironment

  • Immunotherapy implications:

    • NDRG3 expression in tumors might serve as a biomarker for immunotherapy response

    • Targeting NDRG3 could potentially enhance immunotherapy efficacy by modulating T cell function or altering the chemokine milieu

  • Dual-targeting strategies:

    • Combined inhibition of NDRG3 and immune checkpoint molecules might represent a synergistic approach

Research approaches should include:

  • Analysis of immune cell populations in NDRG3-high versus NDRG3-low tumors

  • Assessment of immunotherapy response correlation with NDRG3 expression

  • Co-culture systems to evaluate tumor-immune cell interactions influenced by NDRG3

  • In vivo models combining NDRG3 modulation with immunotherapy

What are the emerging technologies that could advance NDRG3 research and therapeutic development?

Several cutting-edge technologies hold promise for advancing NDRG3 research:

  • Single-cell technologies:

    • Single-cell RNA-seq to identify cell populations dependent on NDRG3

    • Single-cell proteomics to analyze NDRG3 protein interactions at the individual cell level

    • Spatial transcriptomics to map NDRG3 expression within tissue architecture

  • Advanced protein analysis:

    • Proximity labeling approaches (BioID, APEX) to identify NDRG3 interaction partners

    • Hydrogen-deuterium exchange mass spectrometry to study structural dynamics

    • AlphaFold or other AI-based protein structure prediction to model NDRG3 structure

  • High-throughput functional genomics:

    • CRISPR screens to identify synthetic lethal interactions with NDRG3

    • Pooled CRISPR library approaches to discover modulators of NDRG3 expression

    • CRISPR activation/inhibition screens to identify NDRG3-dependent genes

  • Therapeutic development platforms:

    • Structure-based drug design targeting NDRG3

    • Proteolysis-targeting chimeras (PROTACs) for NDRG3 degradation

    • RNA-based therapeutics (siRNA, antisense oligonucleotides) for specific NDRG3 inhibition

  • Advanced in vivo models:

    • Patient-derived xenografts to study NDRG3 in human tumor context

    • Humanized immune system models to study NDRG3's immunological effects

    • Organ-on-chip technologies for high-throughput drug screening

Implementation strategies should consider:

  • Interdisciplinary collaborations between molecular biologists, structural biologists, and computational scientists

  • Biobanking of patient samples with comprehensive clinical data for translational research

  • Integration of multi-omics data to develop comprehensive models of NDRG3 function

How can NDRG3 expression be utilized as a prognostic or predictive biomarker in clinical oncology?

NDRG3 shows significant potential as a clinically relevant biomarker:

Recommended validation approach:

  • Retrospective analysis in archived tumor samples with long-term follow-up data

  • Prospective biomarker studies in clinical trial settings

  • Development of companion diagnostics for potential NDRG3-targeted therapies

What considerations are important when designing clinical trials targeting NDRG3 or utilizing it as a biomarker?

Designing clinical trials involving NDRG3 requires careful consideration of several factors:

Trial design recommendations:

  • Basket trials enrolling patients with various cancer types based on NDRG3 expression

  • Adaptive designs allowing modification based on emerging biomarker data

  • Incorporation of quality-of-life assessments to understand the patient experience

Table 1. NDRG3 Expression Patterns Across Human Tissues and Cancer Types

Tissue/Cancer TypeNDRG3 ExpressionDetection MethodClinical CorrelationReference
Normal TestisHighMPSS*, qRT-PCRN/A
Normal ProstateHighMPSS*, qRT-PCRN/A
Prostate Cancer58.6% of casesIHCAssociated with malignancy
Benign Prostatic Hyperplasia13.2% of casesIHCN/A
Hepatocellular CarcinomaSignificantly higher than non-tumor tissuesqRT-PCR, IHCAssociated with tumor size, pathological grade, and poor prognosis
Invasive Breast Cancer14.5% of casesIHCAssociated with high grade, high Ki-67, ER/PR negativity, HER2 positivity, and poor survival

*MPSS: Massively Parallel Signature Sequencing

Table 2. Phenotypic Effects of NDRG3 Modulation in Experimental Models

Model SystemInterventionObserved EffectsProposed MechanismReference
PC-3 prostate cancer cellsNDRG3 overexpressionIncreased growth rates and migration capabilitiesUpregulation of angiogenic chemokines (CXCL1, CXCL3, CXCL5)
Nude mice with xenograftsNDRG3 overexpressionPromoted xenograft tumor growthEnhanced angiogenesis
Ndrg3TKO mice (T cell-specific knockout)NDRG3 deletionReduced peripheral T cell numbers, altered naïve/memory distributionImpaired integration of TCR and cytokine signals
HY-TCR transgenic miceNDRG3 deletionImpaired survival of T cells with low-avidity TCR stimulationPotentially altered PI3K-Akt signaling

Product Science Overview

Introduction

N-Myc Downstream Regulated 3 (NDRG3) is a member of the N-Myc Downstream Regulated Gene (NDRG) family, which is part of the larger α/β-hydrolase superfamily . The NDRG family includes four members: NDRG1, NDRG2, NDRG3, and NDRG4, each playing distinct roles in cellular processes such as proliferation, differentiation, and stress responses .

Gene and Protein Structure

The NDRG3 gene is located on chromosome 20q11.21 and encodes a protein that is approximately 41 kDa in size . The protein structure of NDRG3 adopts the α/β-hydrolase fold, which is characterized by eight canonical β-strands surrounded by α-helices . Despite its structural similarity to other hydrolases, NDRG3 lacks enzymatic activity due to the substitution of key catalytic residues .

Expression and Regulation

NDRG3 expression is regulated by the MYC oncogene, specifically N-Myc, which is known to repress the expression of NDRG family members . NDRG3 is expressed in various tissues, with notable expression in the prostate, liver, and brain . During embryonic development, NDRG3 expression is observed as early as embryonic day 9.5 (E9.5) .

Biological Functions

NDRG3 plays a crucial role in several physiological processes:

  • Cell Proliferation and Differentiation: NDRG3 is involved in the regulation of cell proliferation and differentiation, particularly in prostate cancer cells .
  • Hypoxia Response: NDRG3 has been implicated in hypoxia-induced cancer metabolism, where it responds to lactate levels and contributes to tumorigenesis .
  • Oncogenic Progression: The flexible loop region of NDRG3, which differs from other NDRG family members, is believed to play a distinct role in oncogenic progression .
Clinical Implications

Given its involvement in cancer metabolism and tumor progression, NDRG3 is a potential target for therapeutic interventions. Understanding the structural and functional characteristics of NDRG3 can aid in the development of targeted therapies for cancers where NDRG3 plays a significant role .

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.