DYNLT1 serves as a non-catalytic accessory subunit of cytoplasmic dynein 1, enabling:
Protein | Role in Dynein Complex | Functional Impact |
---|---|---|
DYNC1I2 | Intermediate chain for cargo binding | Membrane trafficking, Golgi dynamics |
DYNLL1 | Light chain for motor regulation | Apoptosis, estrogen receptor signaling |
TCTEX1D4 | Tctex1 domain-containing protein | Ciliary retrograde transport |
DYNLT1 is implicated in tumor progression and metastasis:
Breast Cancer (BC):
Immune Checkpoint Therapy:
Parameter | High DYNLT1 vs. Low DYNLT1 | P-Value |
---|---|---|
TNM Stage | Higher III/IV prevalence | <0.05 |
Relapse-Free Survival | 45% vs. 72% at 5 years | 0.015 |
Distant Metastasis | 60% reduction with knockdown | <0.0001 |
Spermatogenic Defects: Reduced DYNLT1 mRNA and protein levels observed in oligozoospermic and teratozoospermic males
Mechanism: Regulates dynein-independent GEF-H1 signaling, critical for sperm motility
Diagnostic Biomarker: Overexpression in BC tissues (validated via IHC) distinguishes malignant from normal samples
Therapeutic Target: siRNA-mediated knockdown suppresses tumor growth (in vivo) and metastatic potential
Fertility Applications: Potential marker for idiopathic male infertility screening
Dynein light chain Tctex-type 1 (DYNLT1), a member of the dynein light chain Tctex-type family, is a dynein light chain involved in cargo binding. As one of several non-catalytic accessory components of the cytoplasmic dynein 1 complex, DYNLT1 is believed to play a role in linking dynein to cargos and adapter proteins that regulate dynein function. Cytoplasmic dynein, a crucial motor protein complex, is responsible for minus-end, microtubule-based motile processes. Each dynein complex comprises two heavy chains with ATPase and motor activities, along with a group of accessory polypeptides.
Recombinant Human DYNLT1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 133 amino acids (1-113 a.a.). With a molecular weight of 14.6 kDa, DYNLT1 is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
The DYNLT1 protein solution (1 mg/ml) contains 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, 30% glycerol, and 0.1 M NaCl.
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. To ensure optimal long-term stability, adding a carrier protein (0.1% HSA or BSA) is advisable. Avoid repeated freeze-thaw cycles.
Dynein light chain Tctex-type 1, Protein CW-1, T-complex testis-specific protein 1 homolog, DYNLT1, TCTEL1, TCTEX-1, TCTEX1, CW-1.
MGSSHHHHHH SSGLVPRGSH MEDYQAAEET AFVVDEVSNI VKEAIESAIG GNAYQHSKVN QWTTNVVEQT LSQLTKLGKP FKYIVTCVIM QKNGAGLHTA SSCFWDSSTD GSCTVRWENK TMYCIVSAFG LSI.
DYNLT1 (Dynein light chain Tctex-type 1) is a protein component of the cytoplasmic dynein motor protein complex, encoded by the DYNLT1 gene located on human chromosome 6. It functions primarily as part of the dynein complex responsible for minus-end, microtubule-based motile processes. Within this complex, DYNLT1 serves as a dynein light chain involved in cargo binding to the dynein motor .
Methodologically, researchers typically study DYNLT1's function through protein-protein interaction assays, immunoprecipitation, and immunofluorescence microscopy to visualize its association with microtubules and binding partners. For functional studies, DYNLT1 knockdown or knockout models using shRNA or CRISPR/Cas9 approaches are commonly employed to observe resulting phenotypes.
DYNLT1 expression varies across different human tissues, with notable expression in neural tissues. The DYNLT1 regulatory element has been identified as selectively marking nestin+/GFAP+/Sox2+ neural stem-like cells in both developing and adult brain .
To study DYNLT1 regulation, researchers typically employ:
RT-qPCR for quantitative measurement of mRNA expression
Western blotting for protein expression analysis
Promoter-reporter assays using GFP placed under the control of the DYNLT1 promoter to analyze transcriptional regulation
Chromatin immunoprecipitation (ChIP) to identify transcription factors binding to the DYNLT1 promoter
For clinical samples, multiple complementary approaches yield the most reliable results:
RT-qPCR: For quantitative mRNA expression analysis in blood samples or tissue biopsies. This method has been successfully employed in studies examining DYNLT1 expression in Huntington's disease patients .
Immunohistochemistry (IHC): Optimal for tissue sections, using antibodies against DYNLT1 (e.g., Proteintech, 11954-1-AP, 1:500 dilution). Scoring systems typically combine staining intensity (0-3) and frequency of positive cells (0-4) to generate a composite score (0-12) .
Western blotting: For protein expression quantification in tissue lysates.
RNA-seq: For unbiased transcriptome-wide analysis, which can place DYNLT1 expression in broader pathway contexts.
DYNLT1 engages with multiple cellular partners beyond the dynein complex, functioning in diverse signaling networks:
G-protein βγ subunits: DYNLT1 interaction regulates initial neurite sprouting, axonal specification, and elongation of hippocampal neurons .
GEF-H1: DYNLT1 binds GEF-H1 to microtubules, and its release without microtubule depolymerization is mediated through interaction with G proteins .
Protein-protein interaction network: Analysis using the String database (https://www.string-db.org/) reveals multiple potential interacting proteins .
For identifying novel interactions, co-immunoprecipitation followed by mass spectrometry, yeast two-hybrid screening, or proximity labeling methods (BioID, APEX) are recommended methodological approaches.
DYNLT1 serves as a novel marker for neural progenitors in adult brain, with its regulatory elements selectively marking nestin+/GFAP+/Sox2+ neural stem-like cells in both developing and adult brain . Genetic knockdown of DYNLT1 in radial precursors promotes neurogenesis, indicating its role in neural progenitor fate determination .
In Huntington's disease (HD), DYNLT1 gene expression is significantly downregulated in patient blood samples, with this downregulation correlating with disease progression. Studies comparing DYNLT1 expression between HD patients and healthy age-matched controls, analyzed at multiple time points, suggest DYNLT1 could serve as a peripheral prognostic indicator for HD .
Methodologically, researchers investigating DYNLT1 in neurological contexts should consider:
Patient-derived iPSCs differentiated into neural lineages
Conditional knockout mouse models
Longitudinal expression studies in patient samples
Brain organoid models to study developmental roles
DYNLT1 shows complex associations with cancer outcomes, varying by cancer type:
DYNLT1 is overexpressed in BC tissues compared to normal breast tissue
DYNLT1 promotes BC development by enhancing cell proliferation, migration, and metastasis
Expression correlates with multiple clinicopathological features including HER-2 status, PAM50 subtype, Scarff-Bloom-Richardson grade, and Nottingham prognostic index
To study DYNLT1's role in cancer progression, recommended methodologies include:
Colony formation assays to assess proliferation
Transwell assays to evaluate invasion and migration
In vivo xenograft models using DYNLT1-knockdown cancer cells
Analysis of metastatic lesions using H&E staining of affected organs
Correlation of DYNLT1 expression with mutational landscape and immune infiltration patterns
DYNLT1 promotes cell migration and metastasis in breast cancer through mechanisms that likely involve:
Cytoskeletal regulation: As a component of the dynein complex, DYNLT1 can influence microtubule dynamics and cellular transport processes essential for migration.
Cell proliferation promotion: Experiments with DYNLT1 knockdown in MDA-MB-231 breast cancer cells demonstrate reduced colony formation .
Metastatic capability: Animal models show that DYNLT1 knockdown reduces metastatic spread to organs such as lungs and liver .
For investigating these mechanisms, researchers should consider:
Live-cell imaging of cytoskeletal dynamics
Assessment of focal adhesion turnover
Analysis of Rho GTPase activity (especially given DYNLT1's interaction with GEF-H1)
Proteomic analysis of migration-associated complexes
In vivo metastasis models with tissue-specific DYNLT1 manipulation
For reliable functional studies, researchers can modulate DYNLT1 expression through multiple approaches:
shRNA-mediated knockdown using lentiviral vectors. Example sequences that have proven effective include:
Cloning DYNLT1 into expression vectors such as pEGFP-N1
DYNLT1 (Dynlt1b; Accession No. CCDS49936.1) can be amplified (342 bp) from total cDNA using gene-specific primers with appropriate restriction sites
qPCR for mRNA expression changes
Western blot for protein level alterations
Functional readouts appropriate to the biological context being studied
DYNLT1 shows promising potential as a biomarker in multiple disease contexts:
DYNLT1 gene expression is downregulated in blood samples from HD patients
This downregulation correlates with disease progression
Expression analysis at multiple time points suggests DYNLT1 could serve as a peripheral prognostic indicator for HD
Overexpression predicts poor relapse-free survival
Expression correlates with multiple clinicopathological features
Standardization of detection methods across clinical laboratories
Establishment of reference ranges in diverse populations
Correlation with existing clinical parameters and outcomes
Development of minimally invasive sampling procedures (liquid biopsies)
Integration with other biomarkers for improved predictive power
For cancer studies: Use matched pairs of normal/cancer cell lines with validated DYNLT1 expression differences
For neurological studies: Consider neural progenitor cells, differentiated neurons, or patient-derived iPSCs
For reproductive studies: Utilize appropriate reproductive tissue cell models given DYNLT1's association with male factor infertility
SCID mice have been successfully used for xenograft studies with DYNLT1-manipulated cancer cells
Consider conditional knockout models for tissue-specific DYNLT1 deletion
For neurological studies, consider models allowing visualization of neural progenitor development
Multiple independent DYNLT1 knockdown or overexpression constructs
Rescue experiments to confirm specificity of observed phenotypes
Parallel approaches (e.g., pharmacological and genetic) when possible
Appropriate controls for each experimental system
For effective DYNLT1 cloning and expression:
DYNLT1 can be amplified (342 bp) from mature mouse total testicular cDNA using gene-specific primers:
Subcloning into TOPO vectors: PCR products can be subcloned into pCR4-TOPO vectors for sequence verification
Expression vectors: For tagged expression, HindIII and SalI restriction sites can be incorporated into primers for directional cloning into vectors like pEGFP-N1
Lentiviral vectors: For stable expression or knockdown, pLKO.1-based vectors are effective
Colony PCR using vector-specific primers
Restriction enzyme digestion
DNA sequencing using automated sequencing with Big Dye Terminator v3.1 Cycle Sequencing Kit
Bioinformatic analysis of DYNLT1 pathway associations can be conducted through multiple complementary approaches:
Input DYNLT1 into the String database (https://www.string-db.org/) to generate PPI networks
Analyze direct and indirect interactions to identify functional modules
Perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses using R software with the "clusterProfiler" package
Consider terms with false discovery rate (FDR) < 0.05 as significant
Use tools like CAMOIP (http://camoip.net/) to perform GSEA analysis with cancer datasets
Compare pathway activation patterns between DYNLT1-high and DYNLT1-low samples
Analyze correlations between DYNLT1 expression and mutation patterns using tools like cBioPortal (http://www.cbioportal.org/)[4]
Examine associations with DNA damage repair pathway genes
Given DYNLT1's role in the dynein motor complex, specialized techniques for studying transport functions include:
Fluorescently tagged DYNLT1 for real-time visualization
Dual-color imaging with cargo proteins to analyze co-transport
Photo-activatable proteins to track directional movement
Immunoprecipitation of DYNLT1 followed by mass spectrometry
Proximity labeling methods (BioID, APEX) to identify transient interactions
In vitro binding assays with purified proteins
Golgi fragmentation and reassembly assays
Endosomal trafficking analysis
Mitochondrial transport in neurons
Vesicle motility tracking
Optical trapping to measure force generation
Single-molecule tracking of labeled DYNLT1-containing complexes
In vitro reconstitution of transport complexes
DYNLT1 shows context-dependent effects across cancer types, with both pro-tumorigenic and anti-tumorigenic roles reported . When facing conflicting data, researchers should:
Consider tissue context: Perform tissue-specific analysis separating different cancer types and subtypes.
Examine molecular subtypes: In breast cancer, DYNLT1 expression varies with molecular features like HER-2 status and PAM50 subtype .
Analyze pathway intersections: Use bioinformatic approaches to identify differentially activated pathways between cancer types showing opposite DYNLT1 effects.
Control for confounding factors: Genetic background, treatment history, and tumor stage can influence DYNLT1's apparent role.
Direct comparative studies: Design experiments directly comparing DYNLT1 manipulation across multiple cancer cell lines under identical conditions.
This table summarizes reported DYNLT1 effects across contexts:
To validate DYNLT1 as a robust clinical biomarker, researchers should address:
Assay reproducibility across different laboratories
Standardization of collection, storage, and processing protocols
Establishment of reference ranges in healthy populations
Inter-assay and intra-assay coefficient of variation measurement
Prospective studies in target patient populations
Comparison with existing biomarkers and clinical parameters
Longitudinal sampling to assess temporal dynamics (particularly important for Huntington's disease progression monitoring)
Multivariate analysis with other clinically relevant factors
Cost-effectiveness analysis compared to existing diagnostic/prognostic approaches
Assessment of clinical decision impact (would DYNLT1 testing change management?)
Implementation requirements (specialized equipment, training)
Regulatory pathway planning
DYNLT1 displays diverse functions across cellular contexts, functioning both within and independently of the dynein complex. To reconcile these diverse roles:
Protein interaction mapping: Identify context-specific interaction partners that might direct DYNLT1 toward different functions.
Post-translational modification analysis: Investigate whether phosphorylation, ubiquitination, or other modifications switch DYNLT1's functional modes.
Subcellular localization studies: Track whether DYNLT1 localizes differently in various cell types or under different conditions.
Isoform-specific analysis: Determine whether alternative splicing creates context-specific DYNLT1 variants.
Integrated multi-omics approach: Combine transcriptomics, proteomics, and metabolomics to build comprehensive models of DYNLT1's role in different cellular networks.
Evolutionary analysis: Examine conservation patterns to identify functionally important domains that might serve distinct roles.
When analyzing DYNLT1 expression in clinical datasets, appropriate statistical approaches include:
Student's t-test for comparing two groups with normally distributed data
Mann-Whitney U test for non-normally distributed data
ANOVA with post-hoc tests for multiple group comparisons
Chi-square test for categorical variables (e.g., high vs. low expression)
Kaplan-Meier curves with log-rank tests for comparing survival between DYNLT1-high and DYNLT1-low groups
Cox proportional hazards models for multivariate analysis, adjusting for confounding clinical factors
Competing risk analysis when multiple outcome types are possible
Receiver operating characteristic (ROC) curve analysis to determine optimal cutoff values
Calculation of sensitivity, specificity, positive and negative predictive values
Net reclassification improvement (NRI) analysis when adding DYNLT1 to existing biomarker panels
Mixed effects models for repeated measures over time (particularly relevant for Huntington's disease progression)
Time series analysis for expression pattern changes
Based on current evidence, several therapeutic strategies targeting DYNLT1 warrant investigation:
Upregulation of DYNLT1 expression might counteract the observed downregulation in HD patients
Development of small molecules that mimic DYNLT1 function in neural tissues
Gene therapy approaches to restore DYNLT1 levels in affected regions
Development of DYNLT1 inhibitors to reduce proliferation and metastasis
Combination approaches targeting DYNLT1 alongside established therapies
Stratification of patients based on DYNLT1 expression for personalized treatment selection
Small molecule inhibitors of DYNLT1-protein interactions
Peptide-based inhibitors targeting specific binding interfaces
Antisense oligonucleotides or siRNA for DYNLT1 knockdown in cancer contexts
AAV-based gene therapy for neurological applications
Emerging technologies with potential to deepen our understanding of DYNLT1 include:
Single-cell RNA-seq to identify cell populations with differential DYNLT1 expression
Single-cell proteomics to track DYNLT1 protein levels and modifications at cellular resolution
Spatial transcriptomics to map DYNLT1 expression patterns within tissues
Super-resolution microscopy to visualize DYNLT1-containing complexes below the diffraction limit
Lattice light-sheet microscopy for long-term live imaging of DYNLT1 dynamics
Correlative light and electron microscopy to link DYNLT1 function to ultrastructural features
CRISPR base editing for precise modification of DYNLT1 regulatory elements
CRISPR activation/repression systems for temporal control of DYNLT1 expression
CRISPR screening to identify synthetic lethal interactions with DYNLT1 manipulation
Deep learning for prediction of DYNLT1 binding partners and regulatory networks
Molecular dynamics simulations to understand structural basis of DYNLT1 interactions
Multi-scale modeling of DYNLT1's role in cellular transport and signaling
DYNLT1 research shows promising intersections with several cutting-edge biomedical fields:
DYNLT1 expression in blood samples correlates with Huntington's disease progression
Potential utility for monitoring other conditions where DYNLT1 plays a role
Integration with circulating tumor cell analysis for cancer monitoring
DYNLT1's role in neural progenitor cells suggests applications in regenerative medicine
Potential target for promoting neurogenesis after injury or in neurodegenerative conditions
Applications in directing neural stem cell fate in tissue engineering
DYNLT1 expression patterns correlate with breast cancer subtypes and features
Potential for improving patient stratification and treatment selection
Integration with immune profiling for comprehensive tumor characterization
DYNLT1's role in neural development suggests broader applications in understanding embryogenesis
Potential implications for congenital disorders
Applications in directing differentiation of pluripotent stem cells
The cytoplasmic dynein motor complex consists of two heavy chains that have ATPase and motor activities, along with several accessory polypeptides, including the light chain Tctex-type 1 . The primary role of DYNLT1 is to act as a non-catalytic accessory component of the dynein complex, linking dynein to various cargos and adapter proteins that regulate its function .
DYNLT1 is involved in the intracellular retrograde motility of vesicles and organelles along microtubules. It binds to transport cargos and plays a crucial role in apical cargo transport, such as rhodopsin-bearing vesicles in polarized epithelia . Additionally, it may serve as an accessory component of axonemal dynein .
DYNLT1 has several dynein-independent functions, including roles in G protein signaling activation and neuronal growth . It is selectively enriched in proliferating neural progenitors of both embryonic and adult brains . This protein also interacts with viral proteins, such as the minor capsid protein L2 of human papillomavirus, and is required for dynein-mediated delivery of viral nucleic acid to the host nucleus .
Moreover, DYNLT1 interacts with oncogenic nucleoporins to disrupt gene regulation and cause leukemic transformation . It plays a role in neuronal morphogenesis, independent of cytoplasmic dynein, by enhancing Rac1 activity and regulating the actin cytoskeleton .
Recombinant human DYNLT1 is used in research to study its function and interactions within the cell. It is also utilized in assays to investigate its role in disease mechanisms and to develop potential therapeutic strategies.