TRAPPC4 Human

Trafficking Protein Particle Complex 4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to TRAPPC4 Human

TRAPPC4 (Trafficking Protein Particle Complex Subunit 4) is a core component of the TRAPP (Transport Protein Particle) complexes, which regulate membrane trafficking pathways critical for cellular function. In humans, it is encoded by the TRAPPC4 gene located on chromosome 11 (11q23.3) and plays pivotal roles in:

  • Vesicular transport: Facilitating ER-to-Golgi trafficking and recycling endosome dynamics.

  • Immune regulation: Modulating PD-L1 (Programmed Death-Ligand 1) expression in cancer cells.

  • Neurodevelopment: Maintaining neuronal function and preventing degenerative disorders.

This protein is implicated in both cancer immunology and neurodevelopmental diseases, underscoring its dual clinical significance.

TRAPP Complex Composition

TRAPPC4 is a subunit of TRAPPII and TRAPPIII complexes, which differ in their accessory subunits but share core components (e.g., TRAPPC1, TRAPPC2, TRAPPC3, TRAPPC5). These complexes act as Rab GTPase activators, enabling membrane tethering and fusion.

Key Interacting Proteins (String Database)

ProteinFunctionInteraction Score
TRAPPC6AMelanosome biogenesis0.999
TRAPPC8ER-to-Golgi trafficking, autophagy0.999
TRAPPC2Vesicular transport, cell survival0.999
TRAPPC5ER-to-Golgi trafficking0.999
TRAPPC3Vesicular transport, ER stress response0.999

Data sourced from STRING interaction network analysis .

Subcellular Localization

TRAPPC4 localizes primarily to recycling endosomes and the Golgi apparatus, where it interacts with RAB11 and PD-L1 to regulate membrane recycling .

Role in Cancer Immunology

TRAPPC4 promotes PD-L1 recycling to the tumor cell surface, enabling immune evasion. Key findings:

  • PD-L1 dependency: Knockdown of TRAPPC4 reduces PD-L1 protein levels by 40–60% in colorectal cancer cells .

  • Tumor microenvironment: Correlation between TRAPPC4 and PD-L1 expression in human colorectal cancer tissues (Spearman’s r = 0.8314) .

  • Therapeutic potential: Targeting TRAPPC4 could enhance anti-PD-1/PD-L1 immunotherapy efficacy .

PD-L1 Regulation Mechanism

ProcessTRAPPC4’s RoleOutcome
Recycling endosomeBinds PD-L1 and RAB11, facilitatingPD-L1 accumulation on
dynamicsvesicle recyclingcell surface
Lysosomal degradationProtects PD-L1 from degradationIncreased PD-L1 stability

Neurodevelopmental Disorders

A homozygous splice variant c.454+3A>G in TRAPPC4 causes NEDESBA (Neurodevelopmental Disorder with Epilepsy, Spasticity, and Brain Atrophy), characterized by:

  • Clinical features: Microcephaly, early-onset epilepsy, spastic tetraplegia, and progressive brain atrophy .

  • Molecular mechanism: Partial exon 3 skipping, leading to a premature stop codon and reduced protein levels .

Clinical Variant Overview

VariantEffectCarrier Frequency
c.454+3A>G (splice)Aberrant splicing, loss of function2.4–5.4 per 10,000
TRAPPC4 deficiencyImpaired autophagy, ER stressRare (recessive)

Data from exome sequencing and population studies .

Loss-of-Function Studies

  • PD-L1 reduction: siRNA-mediated TRAPPC4 knockdown in RKO cells decreased PD-L1 membrane expression by 70% .

  • T-cell activation: TRAPPC4-deficient tumor cells showed impaired PD-1 binding and enhanced cytotoxic T-cell activity .

Splice Variant Pathogenicity

  • RNA sequencing: Patient-derived RNA revealed partial exon 3 skipping, generating a truncated protein .

  • Yeast model: Temperature-sensitive TRS23 (yeast TRAPPC4 homolog) mutants exhibited defective secretion and autophagy .

Future Directions and Therapeutic Targets

  1. Cancer Therapy: Combining TRAPPC4 inhibitors with anti-PD-1 antibodies to overcome immune evasion.

  2. Genetic Screening: Identifying TRAPPC4 variants in populations with microcephaly or epilepsy.

  3. Biomarker Development: Validating TRAPPC4/PD-L1 co-expression as a predictive marker for immunotherapy response.

Product Specs

Introduction
Trafficking protein particle complex 4 (TRAPPC4), a component of the multisubunit TRAPP (transport protein particle) complex, interacts with SDC2 and plays a role in vesicular transport from the endoplasmic reticulum to the Golgi apparatus.
Description
Recombinant human TRAPPC4, expressed in E. coli, is a non-glycosylated polypeptide chain containing 242 amino acids (residues 1-219) with a molecular weight of 26.7 kDa. It includes a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile filtered solution.
Formulation
The TRAPPC4 protein solution (0.25 mg/ml) is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.2 M NaCl, 20% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Trafficking protein particle complex subunit 4, TRS23 homolog, Synbindin, Hematopoietic stem/progenitor cell protein 172.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAIFSVY VVNKAGGLIY QLDSYAPRAE AEKTFSYPLD LLLKLHDERV LVAFGQRDGI RVGHAVLAIN GMDVNGRYTA DGKEVLEYLG NPANYPVSIR FGRPRLTSNE KLMLASMFHS LFAIGSQLSP EQGSSGIEML ETDTFKLHCY QTLTGIKFVV LADPRQAGID SLLRKIYEIY SDFALKNPFY SLEMPIRCEL FDQNLKLALE VAEKAGTFGP GS.

Q&A

What is TRAPPC4 and what is its role in cellular trafficking?

TRAPPC4 (Transport Protein Particle Complex 4) is a core subunit of the TRAPP complex, which functions as a modular activator of Ypt/RAB GTPases. This protein is highly conserved from yeast to humans and plays an essential role in membrane trafficking between lipid organelles in a process called vesicular tethering . TRAPP complexes are critical for cellular processes including secretion and macroautophagy/autophagy . The function of TRAPPC4 specifically involves coordinating membrane trafficking pathways, acting as scaffolding proteins that facilitate the proper movement of vesicles within cells. As a core component, TRAPPC4 is required for the stability and function of the entire TRAPP complex, contributing to the regulation of both secretory pathways and autophagy processes .

How is TRAPPC4 structured and what are its functional domains?

TRAPPC4 is one of four core subunits essential for cell viability in the TRAPP complex. While the search results don't detail specific functional domains, research indicates that TRAPPC4 interacts with other TRAPP subunits to form a functional complex that serves as a guanine nucleotide exchange factor (GEF) for RAB GTPases . The protein's structure enables it to serve as a scaffold, as evidenced by its ability to interact simultaneously with PD-L1 and RAB11 in recycling endosomes . This scaffolding function is critical for coordinating protein-protein interactions that facilitate vesicular trafficking. Any structural alterations, such as those caused by the c.454+3A>G splicing variant, can disrupt these interactions and compromise cellular trafficking mechanisms .

How conserved is TRAPPC4 across species and what does this suggest about its evolutionary importance?

TRAPPC4 demonstrates remarkable evolutionary conservation from yeast to human cells, indicating its fundamental importance in eukaryotic cellular processes . In yeast, the homolog of TRAPPC4 is known as Trs23, and conditional mutations in this gene produce similar cellular defects to those observed with pathogenic human TRAPPC4 variants . This high degree of conservation suggests that TRAPPC4 performs essential cellular functions that have been maintained throughout eukaryotic evolution. The conservation extends not only to the protein sequence but also to its functional role in membrane trafficking and autophagy pathways. The fact that both yeast and human cells with TRAPPC4/Trs23 defects show similar patterns of impairment—with autophagy more severely affected than secretion—further underscores the evolutionary significance of this protein .

What is the genetic basis of TRAPPC4-associated neurological disorders?

The primary genetic cause of TRAPPC4-associated neurological disorders is a homozygous splicing variant, c.454+3A>G (located at genomic position hg38:11:119020256 A>G), which affects an intron-exon junction . This variant results in an incompletely penetrant splicing defect that reduces the levels of wild-type TRAPPC4 transcripts and subsequently the amount of full-length TRAPPC4 protein and intact TRAPP complex . The disorder follows an autosomal recessive inheritance pattern, requiring both alleles to carry the pathogenic variant for disease manifestation . Interestingly, this specific variant has been identified in multiple unrelated families from diverse ethnic backgrounds, including European, Mediterranean, Middle Eastern, and Indian ancestries . Carrier frequency is relatively common, ranging from 2.4 to 5.4 per 10,000 individuals worldwide, with higher frequency in Mediterranean and European populations .

What is the clinical spectrum of TRAPPC4-related neurodevelopmental disorders?

TRAPPC4-related disorders, sometimes referred to as NEDESBA (Neurodevelopmental Disorder with Epilepsy, Spasticity, and Brain Atrophy), present with a severe neurological phenotype characterized by:

  • Early-onset seizures, including infantile spasms

  • Progressive microcephaly

  • Severe developmental delay and regression

  • Spastic quadriparesis

  • Visual impairment and optic atrophy

  • Sensorineural hearing loss

  • Brain abnormalities, including progressive cortical atrophy

  • Dysmorphic facial features (bitemporal narrowing, thick eyebrows, long filtrum, thin upper lip, pointed chin)

Patients typically show normal development initially, followed by developmental stagnation and regression, often triggered by seizures . The condition is progressive, with neuroimaging showing increasing cerebral atrophy, particularly in frontotemporal regions . Some patients may also exhibit frequent infections and immunological abnormalities, such as low IgA levels . The prognosis is poor, with most affected individuals not surviving beyond the first decade of life (mean age of death around 8.8 years) .

How do researchers differentiate TRAPPC4-related disorders from other neurodegenerative conditions?

Differential diagnosis of TRAPPC4-related disorders requires a systematic approach:

  • Clinical features assessment: The combination of severe microcephaly, early-onset seizures, developmental regression, and progressive cortical atrophy is characteristic of TRAPPC4-related disorders .

  • Neuroimaging: Sequential MRI demonstrating progressive cortical atrophy, particularly in frontotemporal regions, with enlarged lateral ventricles and subarachnoid spaces .

  • EEG patterns: Very low voltage background activity and epileptiform abnormalities, often with generalized disorganization .

  • Metabolic screening: A thorough metabolic workup to exclude other neurodegenerative conditions such as GM1 and GM2 gangliosidosis, mitochondrial encephalopathies, organic acidurias, and Lesch Nyhan syndrome that may present with similar basal ganglia and cerebellar involvement .

  • Genetic testing: Whole-exome sequencing or whole-genome sequencing is crucial for definitive diagnosis, with targeted sequencing for the common c.454+3A>G variant in TRAPPC4 being particularly effective given its relatively high prevalence .

  • Dysmorphology assessment: Characteristic facial features can provide additional diagnostic clues .

The availability of next-generation sequencing has significantly reduced the "diagnostic odyssey" for these patients, with phenotype-guided genetic tests achieving diagnostic confirmation rates of up to 94% .

What are the molecular mechanisms by which TRAPPC4 mutations cause neurological disease?

The pathogenesis of TRAPPC4-related neurological disorders involves several interrelated mechanisms:

  • Reduced protein levels: The c.454+3A>G variant causes an incompletely penetrant splicing defect, resulting in decreased levels of full-length TRAPPC4 protein and the entire TRAPP complex .

  • Autophagy defects: Both yeast models with Trs23 (TRAPPC4 homolog) mutations and patient-derived fibroblasts demonstrate severe defects in autophagy pathways . This impairment in autophagy appears more pronounced than secretion defects and is likely the primary pathogenic mechanism.

  • Neuronal vulnerability: Neuronal cells are particularly sensitive to long-term autophagy defects, explaining the predominantly neurological phenotype . Autophagy is critical for maintaining neuronal homeostasis and clearing protein aggregates and damaged organelles.

  • Vesicular trafficking disruption: Defects in TRAPPC4 disrupt the TRAPP complex's function in vesicular tethering and membrane trafficking, affecting multiple cellular processes dependent on proper protein and lipid transport .

  • Developmental impacts: TRAPP proteins are essential for dendritic spine morphogenesis, and their dysfunction likely contributes to the microcephaly and neurodevelopmental abnormalities observed in affected individuals .

The differential impact of TRAPPC4 deficiency on autophagy versus secretion suggests that neurons, with their complex morphology and high metabolic demands, are especially vulnerable to autophagy disruptions over extended periods .

What experimental models are used to study TRAPPC4 function and pathology?

Researchers utilize multiple experimental models to investigate TRAPPC4:

  • Yeast models: Conditional mutations in Trs23 (the yeast homolog of TRAPPC4) provide valuable insights into fundamental cellular functions. These models demonstrate that lower levels of Trs23 result in decreased TRAPP complex formation with consequent defects in autophagy and, to a lesser extent, secretion .

  • Patient-derived fibroblasts: Primary fibroblasts from individuals with TRAPPC4 variants allow direct assessment of cellular phenotypes in human cells carrying the disease-causing mutation. These cells exhibit similar defects to yeast models, validating the cross-species conservation of TRAPPC4 function .

  • Cancer cell lines: Human cancer cell lines with TRAPPC4 knockdown have been used to study its role in PD-L1 trafficking and immune evasion mechanisms. These models revealed TRAPPC4's function as a scaffold between PD-L1 and RAB11 in recycling endosomes .

  • Murine tumor models: Mouse models overexpressing Trappc4 have demonstrated the protein's role in checkpoint therapy sensitivity, providing insights into potential therapeutic applications in cancer immunotherapy .

These complementary models allow researchers to investigate TRAPPC4 function across different biological contexts, from basic cellular mechanisms to disease-specific pathways in both neurodevelopmental disorders and cancer.

How does TRAPPC4 interact with other TRAPP complex components and downstream effectors?

TRAPPC4 functions as a core subunit of the modular TRAPP complex, interacting with other TRAPP components to form a functional unit that activates Ypt/RAB GTPases . While the search results don't provide detailed information on all interactions, several key aspects are evident:

These interactions position TRAPPC4 as a critical coordinator in vesicular trafficking pathways, linking the TRAPP complex to specific RAB GTPases and cargo proteins to regulate membrane transport processes.

What role does TRAPPC4 play in cancer immunology through PD-L1 regulation?

TRAPPC4 has emerged as a crucial regulator of programmed death-1 ligand 1 (PD-L1) in cancer cells, with significant implications for tumor immune evasion:

  • PD-L1 trafficking regulation: TRAPPC4 maintains PD-L1 expression levels by coordinating its intracellular trafficking and recycling. It acts as a scaffold for PD-L1 and RAB11 in recycling endosomes, promoting RAB11-mediated recycling of PD-L1 to the cell surface .

  • Protection from degradation: By facilitating recycling, TRAPPC4 protects PD-L1 from lysosomal degradation, thereby maintaining higher levels of this immune checkpoint protein on cancer cell membranes .

  • Post-translational regulation: Unlike other PD-L1 regulators, TRAPPC4 does not affect PD-L1 mRNA levels. Instead, it regulates PD-L1 through post-translational mechanisms involving protein trafficking and stability .

  • Immune checkpoint function: TRAPPC4-mediated maintenance of surface PD-L1 enhances PD-1/PD-L1 binding, promoting T-cell tolerance and immune evasion. Knockdown of TRAPPC4 significantly impairs PD-1 binding to tumor cells .

  • Response to inflammatory signals: TRAPPC4 is particularly important for sustaining PD-L1 upregulation in response to IFN-γ stimulation, a key inflammatory signal in the tumor microenvironment .

These findings identify TRAPPC4 as a major regulator of immune checkpoint function in cancer cells through its specific role in vesicular trafficking pathways.

How might targeting TRAPPC4 enhance cancer immunotherapy approaches?

Targeting TRAPPC4 offers several promising strategies for enhancing cancer immunotherapy:

  • Disruption of immune evasion: Inhibition of TRAPPC4 reduces surface PD-L1 expression on tumor cells, potentially diminishing their ability to evade T cell-mediated cytotoxicity .

  • Synergy with checkpoint inhibitors: TRAPPC4 depletion sensitizes tumor cells to immune checkpoint blockade therapy, suggesting that combinatorial approaches targeting both TRAPPC4 and PD-1/PD-L1 directly could enhance therapeutic efficacy .

  • Specificity advantage: TRAPPC4 appears to have unique functions in PD-L1 trafficking compared to other TRAPP subunits, making it a potentially specific target with fewer off-target effects .

  • Post-translational intervention: Since TRAPPC4 regulates PD-L1 post-translationally rather than at the transcriptional level, targeting this pathway might overcome resistance mechanisms involving transcriptional upregulation of immune checkpoint molecules .

  • Target across cancer types: TRAPPC4's role in maintaining PD-L1 expression has been observed in multiple cancer cell lines, suggesting broad applicability across different tumor types .

Experimental evidence from murine tumor models indicates that modulating TRAPPC4 expression affects tumor response to checkpoint therapy, providing proof-of-concept for therapeutic approaches targeting this protein .

What experimental techniques are used to study TRAPPC4's role in vesicular trafficking?

Researchers employ various sophisticated techniques to investigate TRAPPC4's function in vesicular trafficking:

  • RNA interference: siRNA-mediated knockdown of TRAPPC4 is used to assess its functional importance in maintaining PD-L1 levels and membrane distribution. This approach revealed that TRAPPC4 depletion significantly reduces PD-L1 expression without affecting its mRNA levels .

  • Flow cytometry: This technique quantifies surface protein expression, allowing researchers to measure the impact of TRAPPC4 manipulation on membrane PD-L1 levels and PD-1 binding capacity .

  • Co-immunoprecipitation: Used to identify protein-protein interactions, this method demonstrated TRAPPC4's direct interaction with PD-L1 and RAB11 in recycling endosomes .

  • Immunofluorescence microscopy: This imaging technique visualizes the co-localization of TRAPPC4 with PD-L1 and vesicular markers to track intracellular trafficking pathways .

  • Mass spectrometry: Proteomic analysis identified TRAPPC4 as a predominant regulator of PD-L1 expression in cancer cells .

  • Functional T-cell assays: These assess the impact of TRAPPC4 manipulation on T cell-mediated cytotoxicity against tumor cells, providing insights into immunological consequences .

  • Murine tumor models: In vivo models evaluate how TRAPPC4 expression affects tumor growth and response to checkpoint inhibitor therapy .

These complementary approaches have collectively established TRAPPC4's critical role in maintaining PD-L1 expression through regulation of vesicular trafficking pathways.

What therapeutic approaches might address TRAPPC4-related neurological disorders?

Potential therapeutic strategies for TRAPPC4-related neurological disorders could include:

  • Splicing modulation: Since the common c.454+3A>G variant affects splicing, antisense oligonucleotides or small molecules that correct splicing defects might restore proper TRAPPC4 expression .

  • Autophagy enhancement: Given that autophagy defects appear to be the primary pathogenic mechanism, therapies that enhance alternative autophagy pathways might ameliorate neuronal dysfunction .

  • Gene therapy: Delivering functional copies of TRAPPC4 to affected tissues, particularly the central nervous system, could potentially restore TRAPP complex function .

  • Protein stabilization: Small molecules that stabilize the reduced levels of TRAPPC4 protein might enhance the function of remaining TRAPP complexes .

  • Early seizure management: Since seizures appear to trigger or accelerate neurodegeneration in these patients, aggressive early seizure control might slow disease progression .

  • Neuroprotective strategies: Approaches that protect neurons from stress caused by trafficking and autophagy defects could potentially slow neurodegeneration .

Current management is largely supportive, focusing on seizure control, nutritional support, and management of complications. The development of disease-modifying therapies will require deeper understanding of molecular pathways downstream of TRAPPC4 dysfunction .

How might genetic screening for TRAPPC4 variants be implemented in clinical practice?

Implementation of genetic screening for TRAPPC4 variants could follow several approaches:

  • Targeted variant screening: Given the relatively high carrier frequency of the c.454+3A>G variant (2.4-5.4 per 10,000 individuals), targeted screening for this specific variant could be cost-effective, particularly in populations with higher carrier rates such as Mediterranean and European ancestries .

  • Inclusion in neurological gene panels: TRAPPC4 should be included in gene panels for early-onset epilepsy, progressive microcephaly, and neurodegenerative disorders in childhood .

  • Prenatal and carrier testing: For families with affected individuals or identified carriers, prenatal diagnosis and extended family carrier testing could be offered .

  • Newborn screening consideration: In populations with higher carrier frequencies, inclusion in expanded newborn screening programs might be considered if early interventions prove beneficial .

  • Phenotype-guided testing: For patients presenting with the characteristic triad of severe microcephaly, early-onset seizures, and progressive cortical atrophy, direct testing for the TRAPPC4 c.454+3A>G variant might be warranted before proceeding to broader genomic testing .

Implementation should include genetic counseling regarding the autosomal recessive inheritance pattern and the incomplete penetrance of the splicing defect, which may complicate genotype-phenotype correlations .

What are the most pressing unanswered questions in TRAPPC4 research?

Several critical questions remain in TRAPPC4 research:

  • Tissue specificity: Why do TRAPPC4 mutations predominantly affect the nervous system despite the protein's ubiquitous expression? Understanding the tissue-specific vulnerability is crucial for therapeutic development .

  • Functional domains: What are the specific functional domains of TRAPPC4 that mediate interactions with other TRAPP components, RAB GTPases, and cargo proteins? Structural insights could guide targeted therapeutic approaches .

  • Mechanistic details: How exactly does TRAPPC4 coordinate RAB11-mediated recycling, and what other RAB GTPases might it regulate? A more complete understanding of the molecular mechanisms would expand therapeutic possibilities .

  • Phenotypic variability: What factors contribute to the variable severity observed among patients with identical TRAPPC4 variants? Identifying genetic modifiers or environmental factors could improve prognostication .

  • Therapeutic windows: Is there a critical developmental period during which therapeutic intervention would be most effective? Determining if neurodegeneration can be halted or reversed at different disease stages is essential .

  • Cancer connections: Beyond PD-L1 regulation, does TRAPPC4 influence other aspects of tumor biology through its trafficking functions? Exploring broader roles in cancer could uncover additional therapeutic applications .

  • Broader immunological functions: Does TRAPPC4 regulate trafficking of other immunologically relevant proteins? This could expand its relevance in immunological disorders and immunotherapy .

Addressing these questions will require interdisciplinary approaches combining structural biology, cell biology, neuroscience, and clinical research.

What are the methodological challenges in studying TRAPPC4 protein function?

Investigating TRAPPC4 function presents several technical challenges:

  • Protein complex integrity: As a component of the multisubunit TRAPP complex, studying TRAPPC4 in isolation may not reflect its native function. Maintaining the integrity of protein-protein interactions during experimental manipulations is challenging .

  • Functional redundancy: Potential compensation by other trafficking pathways when TRAPPC4 is depleted may mask phenotypes in acute experimental models, necessitating careful interpretation of knockdown/knockout studies .

  • Tissue-specific effects: TRAPPC4 may have different functions in different cell types, requiring diverse experimental models to capture its tissue-specific roles, particularly in neurons which are challenging to culture .

  • Temporal dynamics: Vesicular trafficking occurs rapidly, requiring high-resolution imaging techniques to capture the dynamic processes mediated by TRAPPC4 .

  • Distinguishing direct from indirect effects: Determining whether phenotypes result directly from TRAPPC4 dysfunction or from downstream consequences requires careful experimental design and multiple complementary approaches .

  • Translating between models: Findings from yeast or cancer cell models may not directly translate to human neuronal function, necessitating validation across multiple systems .

Overcoming these challenges requires combining genetic, biochemical, and imaging approaches across different model systems to build a comprehensive understanding of TRAPPC4 function.

How can researchers effectively model TRAPPC4-related disorders in experimental systems?

Effective modeling of TRAPPC4-related disorders requires multifaceted approaches:

  • Patient-derived cellular models: Fibroblasts from affected individuals provide directly relevant cellular systems for studying disease mechanisms. These can be reprogrammed into induced pluripotent stem cells (iPSCs) and differentiated into neurons to model neurological aspects of the disease .

  • CRISPR-engineered cell lines: Introducing the c.454+3A>G variant or other TRAPPC4 mutations using CRISPR/Cas9 in relevant cell types can create isogenic models that control for genetic background .

  • Conditional knockout models: Since complete TRAPPC4 loss is likely lethal, conditional knockout systems in specific tissues or at defined developmental stages may better recapitulate the disease .

  • Splicing reporter assays: To specifically study the impact of the c.454+3A>G variant on splicing, minigene constructs containing the relevant exon-intron boundaries can be developed .

  • Yeast complementation studies: Testing whether human TRAPPC4 variants can rescue phenotypes in Trs23 mutant yeast provides functional validation across species .

  • Mouse models: Generating mouse models with the equivalent of the human c.454+3A>G variant would allow investigation of developmental and behavioral phenotypes relevant to the human condition .

  • Organoid models: Brain organoids derived from patient cells or engineered stem cells can recapitulate three-dimensional tissue architecture relevant to neurodevelopmental processes .

The most effective approach would integrate findings across these complementary models, each addressing different aspects of TRAPPC4 biology and disease pathology.

What technological advances might accelerate TRAPPC4 research?

Several emerging technologies could significantly advance TRAPPC4 research:

  • Super-resolution microscopy: Advanced imaging techniques could provide unprecedented insights into the spatial organization and dynamics of TRAPPC4 within vesicular trafficking pathways .

  • Single-cell multi-omics: Combining transcriptomics, proteomics, and metabolomics at the single-cell level could reveal cell-specific responses to TRAPPC4 dysfunction across various tissues .

  • Cryo-electron microscopy: Structural determination of the TRAPP complex with atomic resolution would illuminate how TRAPPC4 interacts with other components and how disease variants disrupt these interactions .

  • Optogenetic tools: Developing tools to manipulate TRAPPC4 function with spatiotemporal precision could reveal its acute roles in specific cellular compartments .

  • Advanced CRISPR screening: Genome-wide CRISPR screens in the context of TRAPPC4 deficiency could identify genetic modifiers and compensatory pathways, potentially revealing therapeutic targets .

  • Microfluidic organ-on-chip technologies: These could model complex tissue interactions relevant to TRAPPC4 function in both neurodevelopmental contexts and cancer microenvironments .

  • AI-driven protein function prediction: Computational approaches could predict the functional impact of TRAPPC4 variants and identify potential small-molecule binding sites for therapeutic development .

  • In vivo imaging of protein trafficking: Development of techniques to visualize protein trafficking in living organisms could bridge the gap between cellular models and organismal phenotypes .

These technological advances, particularly when applied in combination, have the potential to address fundamental questions about TRAPPC4 biology and accelerate therapeutic development for TRAPPC4-related disorders.

Product Science Overview

Introduction

Trafficking Protein Particle Complex 4 (TRAPPC4) is a crucial component of the TRAPP (Transport Protein Particle) complex, which plays a significant role in intracellular vesicular trafficking. This complex is involved in various cellular processes, including secretion, autophagy, and vesicle-mediated transport from the endoplasmic reticulum to the Golgi apparatus .

Structure and Function

TRAPPC4 is a highly conserved protein that acts as a guanine nucleotide exchange factor (GEF) for Rab1 GTPase . This activity is essential for the regulation of vesicular trafficking events. The protein is also known to interact with other components of the TRAPP complex, facilitating the formation and function of the complex .

Biological Significance

The TRAPP complex, including TRAPPC4, is vital for maintaining cellular homeostasis. It ensures the proper transport of proteins and lipids within the cell, which is crucial for various cellular functions. Disruptions in the function of TRAPPC4 can lead to severe cellular dysfunctions and are associated with several neurodevelopmental disorders .

Recombinant Human TRAPPC4

Recombinant human TRAPPC4 is produced using recombinant DNA technology, where the TRAPPC4 gene is cloned and expressed in a suitable host system, such as E. coli. The recombinant protein is then purified using conventional chromatography techniques . This recombinant form is used in various research applications to study the protein’s function and its role in cellular processes.

Applications in Research

Recombinant human TRAPPC4 is widely used in biochemical and cellular studies to understand its role in vesicular trafficking and autophagy. It is also used to investigate the molecular mechanisms underlying neurodevelopmental disorders associated with TRAPPC4 dysfunction .

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.