DCTN2 (1-403) Human

Dynactin 2 (1-403 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions of DCTN2

DCTN2 is a 50 kDa subunit of the dynactin complex, which regulates dynein motor activity for intracellular cargo transport. Key roles include:

  • Microtubule Anchoring: Stabilizes microtubules at centrosomes during mitosis .

  • Dynactin Assembly: Forms 4–5 copies per dynactin molecule, binding directly to the p150 subunit (DCTN1) to stabilize the complex .

  • Mitotic Regulation: Facilitates prometaphase chromosome alignment and spindle organization .

  • Synapse Formation: Critical for neuronal development and synaptic connectivity .

Table 1: Key Interactions of DCTN2 in the Dynactin Complex

Interacting ProteinRoleFunctional ImpactSources
DCTN1 (p150)Binds directly to stabilize dynactinEnhances dynein-mediated organelle transport
ACTR1A (Arp1)Forms the dynactin shoulder domainDetermines filament length and structural integrity
LC8 Dynein Light ChainLinks dynactin to dynein motorsEnables ultra-processive microtubule transport

Table 2: Clinical Implications of DCTN2 Dysregulation

ConditionDCTN2 RoleMechanismSources
Hepatocellular CarcinomaOverexpression drives proliferationActivates AKT pathway; knockdown reduces tumor growth
Pan-Cancer AnalysisHigh expression in 15+ tumor typesCorrelates with poor prognosis and metastasis
Charcot-Marie-Tooth DiseaseMutations disrupt dynein-dynactin functionImpairs axonal transport in neurons

Applications of Recombinant DCTN2 (1-403)

  • Mechanistic Studies: Used to investigate dynactin’s role in mitosis, neuronal transport, and cancer .

  • Antibody Development: Serves as an antigen for generating anti-DCTN2 antibodies (e.g., MAB6850) .

  • Drug Discovery: Target for inhibiting AKT-driven tumor progression .

Product Specs

Introduction
DCTN2, a 50kDa subunit of the dynactin complex, plays a crucial role in various cellular functions. This complex, comprising 10-11 subunits ranging from 22 to 150 kDa, interacts with both microtubules and cytoplasmic dynein. Dynactin is involved in processes such as ER-to-Golgi transport, lysosome and endosome movement, spindle formation, chromosome movement, nuclear positioning, and axonogenesis. DCTN2, present in 4-5 copies per dynactin molecule, consists of three short alpha-helical coiled-coil domains that facilitate interactions with other dynactin subunits, particularly the largest subunit, p150. This interaction is vital for stabilizing p150 within the complex. DCTN2 also regulates cytoplasmic dynein's binding to organelles and participates in chromosome alignment and spindle organization during mitosis. Furthermore, DCTN2 contributes to microtubule anchoring at centrosomes and plays a role in synapse formation during brain development.
Description
Recombinant Human Dynactin 2, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 426 amino acids, including the 403 amino acids of DCTN2 (1-403) and a 23 amino acid His-tag at the N-terminus. The protein has a molecular weight of 46.9kDa and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile filtered.
Formulation
The DCTN2 protein solution is provided at a concentration of 0.5mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 10% glycerol, 0.15M NaCl, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the protein can be stored at 4°C. For long-term storage, it is recommended to freeze the protein at -20°C. To ensure stability during long-term storage, adding a carrier protein like HSA or BSA (0.1%) is advisable. Avoid repeated freezing and thawing cycles.
Purity
The purity of the protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Dynactin 2 (P50), 50 KDa Dynein-Associated Polypeptide, Dynactin Complex 50 KDa Subunit, P50 Dynamitin, DCTN50, 50 KD Dynein-Associated Polypeptide, Epididymis Secretory Protein Li 77, Dynactin Complex 50 KD Subunit, DYNAMITIN, HEL-S-77, DCTN-50, RBP50.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMADPKYA DLPGIARNEP DVYETSDLPE DDQAEFDAEL EELTSTSVEH IIVNPNAAYD KFKDKRVGTK GLDFSDRIGK TKRTGYESGE YEMLGEGLGV KETPQQKYQR LLHEVQELTT EVEKIKTTVK ESATEEKLTP VLLAKQLAAL KQQLVASHLE KLLGPDAAIN LTDPDGALAK RLLLQLEATK NSKGGSGGKT TGTPPDSSLV TYELHSRPEQ DKFSQAAKVA ELEKRLTELE TAVRCDQDAQ NPLSAGLQGA CLMETVELLQ AKVSALDLAV LDQVEARLQS VLGKVNEIAK HKASVEDADT QSKVHQLYET IQRWSPIAST LPELVQRLVT IKQLHEQAMQ FGQLLTHLDT TQQMIANSLK DNTTLLTQVQ TTMRENLATV EGNFASIDER MKKLGK

Q&A

What is DCTN2 and what functional domains are present in the 1-403 amino acid region?

DCTN2 is a subunit of the dynactin complex that plays critical roles in cellular transport and division. The 1-403 amino acid region encompasses the primary functional domains responsible for its interactions with microtubules and other dynactin complex components. This region is particularly important for the protein's ability to participate in intracellular trafficking and mitotic processes. Current research indicates that DCTN2 is highly expressed in many tumor types compared to adjacent non-tumor tissues, suggesting its involvement in oncogenic pathways . The 1-403 region contains domains essential for its participation in cell cycle regulation, particularly the G1/S phase transition that becomes dysregulated in cancer cells .

How is DCTN2 expression measured in experimental contexts?

DCTN2 expression can be measured through multiple complementary techniques. Researchers typically employ quantitative PCR (qPCR) to measure mRNA expression levels from tissue samples. For protein detection, western blotting and immunohistochemistry are standard approaches. Large-scale studies often utilize RNA-sequencing data from databases like The Cancer Genome Atlas (TCGA), which was used to analyze DCTN2 expression across 371 HCC tissues and 50 adjacent non-tumor liver tissues . Additionally, microarray data from databases like Oncomine provide further validation, establishing a threshold value typically defined as a twofold change, a cutoff of P < 0.05, and a top 10% gene ranking . For robust statistical analysis, tools such as unpaired t-tests are employed to analyze differential expression between cancer and normal tissues .

What expression patterns of DCTN2 have been observed across normal tissues and cancer types?

DCTN2 exhibits variable expression patterns across tissues. In cancerous contexts, research through the Tumour Immune Estimation Resource (TIMER) database shows that DCTN2 expression is significantly elevated in multiple cancer types, including hepatocellular carcinoma (HCC), glioblastoma (GBM), and pancreatic cancer, while showing decreased expression in breast cancer, colorectal cancer, and ovarian cancer . This tissue-specific expression pattern suggests context-dependent functions of DCTN2 in different malignancies.

Analysis using data from the GTEX database confirms these findings, and paired analysis further reveals significantly higher expression of DCTN2 in many tumor tissues compared to their normal counterparts . The fold change values for DCTN2 in liver cancer were reported as 1.681 (P=2.15E-11) in the Roessler liver dataset and 1.588 (P=4.96E-04) in the Wurmbach liver dataset, indicating consistent upregulation in HCC .

How does DCTN2 contribute to hepatocellular carcinoma progression?

DCTN2 promotes HCC progression through multiple mechanisms. Experimental evidence demonstrates that DCTN2 acts as an oncogene in HCC, primarily by promoting cell cycle progression through the G1/S phase transition . Mechanistically, DCTN2 exerts tumor-promoting effects by modulating the AKT signaling pathway. Knockdown experiments show that silencing DCTN2 in HCC cells inhibits AKT phosphorylation and its downstream targets .

DCTN2 silencing dramatically suppresses both the proliferative and metastatic capacities of tumor cells in vitro . The anti-tumor effects of DCTN2 knockdown can be partially reversed upon AKT pathway activation, confirming this mechanistic relationship . Gene Set Enrichment Analysis (GSEA) further reveals that DCTN2 expression positively correlates with oncogenic pathways, including cell cycle and tumor metastasis-related pathways, while negatively correlating with anti-tumor immune signaling pathways, such as INF-γ response .

What prognostic value does DCTN2 expression have in cancer patients?

Specific hazard ratios demonstrate this relationship:

DCTN subunitsHR95% CIP-value
DCTN11.261.07–1.910.283
DCTN21.571.45–2.360.008 ∗
DCTN31.341.22–2.060.170
DCTN41.350.98–2.320.378
DCTN51.621.54–2.520.017 ∗
DCTN61.150.89–1.710.488

Both univariate and multivariate analyses confirmed DCTN2 as an independent prognostic factor:

DCTN subunitsUnivariate analysisMultivariate analysis
HR (95% CI)P-valueHR (95% CI)
DCTN11.365 (0.924–2.016)0.119
DCTN21.748 (1.190–2.568)0.004 ∗
DCTN31.098 (0.827–1.457)0.519
DCTN40.719 (0.465–1.111)0.137
DCTN51.441 (1.043–1.990)0.027 ∗
DCTN61.077 (0.810–1.431)0.611

These statistical analyses indicate that DCTN2 is a potential prognostic marker for HCC and potentially other cancers .

How does DCTN2 compare to other dynactin subunits as a cancer biomarker?

Among the dynactin family members, DCTN2 stands out as having exceptional performance as a biomarker. Receiver Operating Characteristic (ROC) curve analysis reveals that DCTN2 has the highest area under the curve (AUC) value among all dynactin subunits:

DCTN subunitsAUC95% CIP-value
DCTN10.7260.665–0.786<0.001
DCTN20.9320.901–0.962<0.001
DCTN30.5750.511–0.6390.084
DCTN40.7280.673–0.784<0.001
DCTN50.6390.577–0.7010.001
DCTN60.2020.161–0.244<0.001

When compared with the traditional HCC biomarker alpha-fetoprotein (AFP), DCTN2 still maintains superior performance:

DCTN subunitsAUC95% CIP-value
DCTN10.6890.614–0.763<0.001
DCTN20.9280.891–0.964<0.001
DCTN30.6250.548–0.7020.008
DCTN40.7520.686–0.819<0.001
DCTN50.5930.514–0.6720.047
DCTN60.2460.184–0.308<0.001

These data clearly indicate that DCTN2 has exceptional discriminatory power as a diagnostic biomarker for HCC, outperforming other dynactin subunits and even traditional markers like AFP .

What are recommended experimental designs for studying DCTN2 function in cancer models?

When designing experiments to study DCTN2 function in cancer, researchers should implement a systematic approach that addresses both expression and functional aspects. A comprehensive experimental design should include:

  • Expression analysis: Compare DCTN2 levels between cancer and adjacent normal tissues using both transcriptomic (qPCR, RNA-seq) and proteomic (western blot, immunohistochemistry) approaches.

  • Loss-of-function studies: Implement siRNA or shRNA knockdown of DCTN2 in appropriate cancer cell lines to assess effects on proliferation, migration, invasion, and cell cycle progression.

  • Mechanism investigation: Analyze downstream signaling pathways, particularly AKT signaling, through western blot for phosphorylated proteins and their targets.

  • Rescue experiments: Perform pathway activation (e.g., using constitutively active AKT) following DCTN2 knockdown to confirm specificity of the observed effects.

  • In vivo validation: Establish xenograft models using DCTN2-knockdown cells to confirm in vitro findings in a more physiologically relevant context .

Importantly, when designing experimental controls, remember that a good experimental design requires a strong understanding of the system being studied, including consideration of variables and their relationships, formulation of specific testable hypotheses, careful design of treatments, appropriate assignment of subjects to groups, and precise measurement planning for dependent variables .

How can researchers analyze the relationship between DCTN2 expression and clinical parameters?

To analyze relationships between DCTN2 expression and clinical parameters, researchers should employ a structured analytical approach:

  • Patient stratification: Categorize patients based on DCTN2 expression levels (high vs. low) using appropriate cutoff values derived from statistical methods like ROC curve analysis or median expression.

  • Statistical analyses: Use chi-square tests to evaluate correlations between DCTN2 levels and clinical features such as cancer stage or tumor grade . For continuous variables, employ Spearman correlation tests to detect associations between DCTN2 and other molecular markers, as was done for cycle-related proteins .

  • Survival analyses: Perform Kaplan-Meier analyses with log-rank tests to compare survival outcomes between high and low DCTN2 expression groups. Follow with Cox proportional hazards regression for both univariate and multivariate analyses to determine if DCTN2 is an independent prognostic factor .

  • Subgroup analyses: Examine DCTN2's prognostic significance across different clinical subgroups based on parameters like tumor stage, grade, or molecular subtypes.

  • Integration with public databases: Utilize tools like UALCAN to stratify patients according to individual cancer stages and tumor grades for more refined analyses .

What methodological approaches can be used to investigate DCTN2's role in cell cycle regulation?

To investigate DCTN2's role in cell cycle regulation, researchers should implement multiple complementary methodological approaches:

  • Flow cytometry: Perform cell cycle analysis using propidium iodide or other DNA intercalating agents to quantify the distribution of cells in G0/G1, S, and G2/M phases following DCTN2 manipulation (overexpression or knockdown).

  • BrdU incorporation assays: Measure the rate of DNA synthesis, particularly to assess S-phase entry and progression, which has been linked to DCTN2 function in G1/S phase transition .

  • Expression analysis of cell cycle regulators: Examine expression levels of cyclins, cyclin-dependent kinases (CDKs), and cell cycle inhibitors through western blotting or qPCR following DCTN2 modulation.

  • Immunofluorescence microscopy: Visualize subcellular localization of DCTN2 throughout different cell cycle phases and co-localization with known cell cycle regulators.

  • Chromatin immunoprecipitation (ChIP): Investigate if DCTN2 interacts with promoter regions of cell cycle-related genes, either directly or as part of protein complexes.

  • Real-time cell analysis: Monitor cell proliferation kinetics in real-time using instruments like xCELLigence following DCTN2 modulation.

These methodological approaches will provide comprehensive insights into the specific mechanisms by which DCTN2 influences cell cycle progression, particularly through the G1/S phase as previously identified .

How does DCTN2 interact with the tumor immune microenvironment?

DCTN2's interaction with the tumor immune microenvironment represents an emerging area of research. Gene Set Enrichment Analysis (GSEA) has revealed that DCTN2 expression negatively correlates with anti-tumor immune signaling pathways, particularly interferon-gamma (IFN-γ) response . This suggests that DCTN2 may contribute to immune evasion mechanisms in cancer.

To investigate this relationship, researchers should consider:

  • Correlation analysis between DCTN2 expression and immune cell infiltration markers using computational approaches on transcriptomic data.

  • In vitro co-culture experiments with immune cells (e.g., T cells, NK cells) and cancer cells with modulated DCTN2 expression to assess functional impacts on immune recognition and cytotoxicity.

  • Examination of major histocompatibility complex (MHC) expression and presentation pathway components in relation to DCTN2 levels.

  • Analysis of immunosuppressive cytokine production in DCTN2-high versus DCTN2-low cancer cells.

These approaches will help elucidate how DCTN2 contributes to immune evasion and potentially identify strategies to overcome DCTN2-mediated immunosuppression in cancer therapy.

What are the potential therapeutic implications of targeting DCTN2 in cancer?

Targeting DCTN2 presents promising therapeutic potential based on its established role in tumor progression. The evidence that DCTN2 silencing dramatically suppresses both proliferative and metastatic capacities of tumor cells in vitro suggests it could be an effective therapeutic target .

Potential therapeutic approaches include:

  • RNA interference technologies: Development of siRNA or shRNA delivery systems specifically targeting DCTN2, potentially using nanoparticle-based delivery systems as mentioned in patent literature for circular RNA therapeutics targeting other cancer-related molecules .

  • Small molecule inhibitors: Design of compounds that disrupt DCTN2's interaction with key binding partners or inhibit its function in promoting AKT signaling.

  • Combination therapies: Integration of DCTN2 targeting with existing chemotherapeutics or immunotherapies, particularly given its negative correlation with anti-tumor immune pathways.

  • Biomarker-guided treatment: Utilization of DCTN2 expression as a stratification marker to identify patients who might benefit from specific treatment regimens.

The patent literature supports the feasibility of such approaches, noting that circular RNA polynucleotides can have "an in vivo duration of therapeutic effect in human greater than that of an equivalent" conventional treatment, and that nanoparticle delivery systems with targeting moieties can be developed for such therapeutics .

How can researchers address contradictory findings regarding DCTN2 function across different cancer types?

The search results indicate that while DCTN2 is overexpressed in many cancers including HCC and glioblastoma, it shows decreased expression in others like breast, colorectal, and ovarian cancers . To address these apparently contradictory findings, researchers should:

  • Perform tissue-specific functional studies: Conduct parallel functional experiments in multiple cancer types to determine if the consequences of DCTN2 modulation are consistent or context-dependent.

  • Investigate tissue-specific interactors: Identify protein interaction partners of DCTN2 in different tissue contexts through techniques like co-immunoprecipitation followed by mass spectrometry.

  • Examine isoform expression: Analyze whether different DCTN2 isoforms predominate in different tissues, potentially explaining divergent functions.

  • Assess genomic and epigenomic landscape: Compare the regulatory elements controlling DCTN2 expression across tissue types, as well as the methylation status of its promoter and enhancers.

  • Consider cellular origins: Evaluate whether differences in cell of origin or differentiation state across cancer types might explain varied DCTN2 functions.

By systematically addressing these aspects, researchers can reconcile contradictory findings and develop a more nuanced understanding of DCTN2's context-dependent roles in cancer biology.

What are the key unanswered questions in DCTN2 research?

Despite significant advances in understanding DCTN2's role in cancer, several critical questions remain unanswered:

  • What is the precise mechanism by which DCTN2 activates the AKT pathway in cancer cells?

  • How does DCTN2 contribute to tumor metastasis beyond its effects on cell proliferation?

  • What is the relationship between DCTN2 and drug resistance in cancer treatment?

  • Are there specific isoforms or post-translational modifications of DCTN2 that are particularly relevant to its oncogenic functions?

  • How do germline or somatic genetic variations in DCTN2 affect cancer susceptibility or progression?

  • What epigenetic mechanisms regulate DCTN2 expression across different tissue contexts?

Addressing these questions will require integrated approaches combining genomic, transcriptomic, proteomic, and functional analyses in both pre-clinical models and clinical samples.

How might emerging technologies advance DCTN2 research?

Emerging technologies offer exciting opportunities to address current knowledge gaps in DCTN2 research:

  • CRISPR-Cas9 gene editing: Precise modification of DCTN2 at the genomic level to study domain-specific functions or create isogenic cell lines for comparative studies.

  • Single-cell sequencing: Characterization of DCTN2 expression heterogeneity within tumors and correlation with cell states or developmental trajectories.

  • Spatial transcriptomics: Analysis of DCTN2 expression in the spatial context of the tumor microenvironment to understand regional variations and microenvironmental influences.

  • Proteomics and interactomics: Comprehensive mapping of the DCTN2 interactome in different cancer contexts to identify critical protein-protein interactions.

  • Patient-derived organoids: Development of three-dimensional culture systems that better recapitulate tumor heterogeneity for functional studies of DCTN2.

  • In vivo CRISPR screens: Systematic identification of synthetic lethal interactions with DCTN2 to uncover potential combination therapy targets.

Product Science Overview

Structure and Composition

Dynactin 2 is a 50 kDa subunit that is present in 4-5 copies per dynactin molecule . It is composed of three short alpha-helical coiled-coil domains, which facilitate its association with other dynactin subunits . The human recombinant form of Dynactin 2 (1-403 a.a.) is produced in Escherichia coli and is a single, non-glycosylated polypeptide chain containing 426 amino acids (1-403 a.a.) with a molecular mass of 46.9 kDa . This recombinant protein is fused to a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques .

Function and Role

Dynactin 2 plays a pivotal role in the dynactin complex by binding to both microtubules and cytoplasmic dynein . This interaction is essential for various cellular functions, including:

  • ER-to-Golgi transport
  • Centripetal movement of lysosomes and endosomes
  • Spindle formation and chromosome movement during mitosis
  • Nuclear positioning and axonogenesis

Dynactin 2 directly interacts with the largest subunit (p150) of the dynactin complex, anchoring it in place and modulating cytoplasmic dynein binding to organelles . It also plays a role in prometaphase chromosome alignment and spindle organization during mitosis . Additionally, Dynactin 2 is involved in anchoring microtubules to centrosomes and has a role in synapse formation during brain development .

Physical Properties and Storage

The recombinant Dynactin 2 protein is provided as a sterile filtered colorless solution. It is formulated in a solution containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, 0.15 M NaCl, and 1 mM DTT . The protein should be stored at 4°C if it will be used within 2-4 weeks. For longer storage, it should be frozen at -20°C, and it is recommended to add a carrier protein (0.1% HSA or BSA) to avoid multiple freeze-thaw cycles .

Applications

Dynactin 2 (1-403 a.a.) (Human Recombinant) is primarily used for laboratory research purposes. It is not intended for use as a drug, agricultural or pesticidal product, food additive, or household chemical .

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.