TRAPPC2 Human

Trafficking Protein Particle Complex 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

TRAPPC2 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 163 amino acids (1-140 a.a.) and having a molecular mass of 18.8kDa.
TRAPPC2 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Trafficking Protein Particle Complex 2 (TRAPPC2) is believed to play a role in the intricate process of transporting cargo within cells. It is thought to be part of a larger complex that helps move vesicles, small sacs that carry molecules, from the endoplasmic reticulum (where proteins are made) to the Golgi apparatus (where proteins are modified and sorted). This protein may also interact with other proteins involved in controlling gene activity. Mutations in the TRAPPC2 gene can lead to a skeletal disorder called spondyloepiphyseal dysplasia tarda (SEDT).
Description
This product consists of the human TRAPPC2 protein, manufactured in a lab using E. coli bacteria. It is a single chain of 163 amino acids, with a molecular weight of 18.8kDa. The protein is not glycosylated, meaning it does not have sugar molecules attached. For purification and detection purposes, a 23 amino acid His-tag is attached to its N-terminus.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
This solution contains 0.25mg/ml of TRAPPC2 protein in a buffer composed of 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 40% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), keep the solution refrigerated at 4°C. For longer periods, freeze the solution at -20°C. Adding a carrier protein (like HSA or BSA) at a concentration of 0.1% is recommended for long-term storage. Avoid repeatedly freezing and thawing the solution.
Purity
The purity of this product is over 90%, as assessed by SDS-PAGE analysis.
Synonyms
Trafficking Protein Particle Complex 2, Sedlin, SEDL, Trafficking Protein Particle Complex Subunit 2, Spondyloepiphyseal Dysplasia Late, TRAPPC2P1, HYP38334, ZNF547L, MIP2A, TRS20, SEDT, TRAPPC2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSGSFYF VIVGHHDNPV FEMEFLPAGK AESKDDHRHL NQFIAHAALD LVDENMWLSN NMYLKTVDKF NEWFVSAFVT AGHMRFIMLH DIRQEDGIKN FFTDVYDLYI KFSMNPFYEP NSPIRSSAFD RKVQFLGKKH LLS.

Q&A

What is TRAPPC2 and what is its primary cellular function?

TRAPPC2 encodes sedlin, an essential component of the trafficking protein particle (TRAPP) complex that mediates vesicular transport between cellular compartments. Specifically, sedlin plays a crucial role in transporting proteins from the endoplasmic reticulum (ER) to the Golgi apparatus. Its most documented function involves facilitating the movement of large proteins, particularly procollagens, out of the ER so they can be processed into mature collagen molecules . Sedlin is also known to interact with MBP1 (myc promoter-binding protein 1) and can block its transcriptional repression capability, suggesting potential roles beyond vesicular trafficking .

The TRAPPC2 gene is located on the X chromosome at position 22 (Xp22) between base-pairs 13,712,241 to 13,734,634 . Notably, processed pseudogenes of TRAPPC2 have been identified on chromosomes 8, 19, and the Y chromosome, although these do not produce functional protein .

How are mutations in TRAPPC2 linked to human disease?

Mutations in the TRAPPC2 gene cause X-linked spondyloepiphyseal dysplasia tarda (X-linked SEDT), a rare skeletal disorder with an estimated prevalence of 1 in 150,000 to 200,000 people worldwide . This condition impairs bone growth and occurs almost exclusively in males due to its X-linked recessive inheritance pattern .

Approximately 90% of X-linked SEDT cases are attributed to TRAPPC2 mutations that result in nonfunctional sedlin protein . The defective sedlin disrupts procollagen transport out of the ER, leading to decreased mature collagen formation and subsequent impairment of bone and cartilage development . Interestingly, despite the congenital nature of the mutation, skeletal problems typically manifest only in childhood (around ages 6-8), suggesting complex developmental regulation of TRAPPC2 function .

What are the typical clinical manifestations of TRAPPC2 mutations?

Patients with TRAPPC2 mutations primarily present with disproportionate short stature and skeletal dysplasia. Clinical manifestations typically include:

  • Delayed linear growth beginning around 6-8 years of age

  • Flattening of vertebral bodies with characteristic humping

  • Progressive joint and back pain, particularly in adulthood

  • Development of early-onset osteoarthritis

  • Disproportionate short stature (short trunk relative to limbs)

These symptoms result from improper bone formation due to disrupted collagen processing and transport. In rare cases, female carriers of TRAPPC2 mutations may develop early-onset osteoarthritis, though they typically do not display the full spectrum of X-linked SEDT symptoms .

What methodologies are recommended for detecting TRAPPC2 variants in clinical and research settings?

For comprehensive TRAPPC2 variant detection, targeted next-generation sequencing (NGS) represents the gold standard approach. This methodology should be implemented with the following considerations:

  • Targeted panel design: Include the complete TRAPPC2 gene, including non-coding regions that may contain regulatory elements

  • Advanced bioinformatics pipeline: Implement algorithms capable of detecting single nucleotide variants, small insertions/deletions, and copy number variations

  • Validation strategy: Confirm novel variants using Sanger sequencing to eliminate false positives

When interpreting variants, researchers should employ established classification frameworks such as the American College of Medical Genetics and Genomics (ACMG) criteria to determine pathogenicity . For example, the novel c.260A>C variant in TRAPPC2 described in one case study was classified as pathogenic based on:

  • In silico prediction tools (SIFT, Polyphen2, MutationTaster, REVEL) consistently indicating damaging effects

  • Protein structure modeling using SWISS-model to predict structural alterations

  • Segregation analysis confirming X-linked inheritance pattern

For genetic counseling purposes, cascade screening of family members should be considered, particularly for females of childbearing age who may be carriers .

What is the spectrum of known pathogenic TRAPPC2 variants and their functional consequences?

The mutational spectrum of TRAPPC2 is relatively narrow, with most pathogenic variants resulting in loss of sedlin function. Based on current research, the following patterns have been observed:

Variant TypeFrequencyFunctional ImpactExamples
MissenseRare (~5 reported)Protein misfolding affecting TRAPP complex assemblyc.260A>C (p.H87P), c.239A>G (p.H80R), c.248T>C (p.F83S)
NonsenseCommonTruncated, non-functional proteinMultiple variants reported
FrameshiftCommonTruncated, non-functional proteinMultiple variants reported
Splice-siteReportedAberrant mRNA processingLimited data available

Interestingly, missense variants are relatively rare in the TRAPPC2 mutational spectrum, with only a few documented cases . The novel c.260A>C (p.H87P) variant reported in a 13-year-old Chinese Han boy is adjacent to previously reported pathogenic variants c.239A>G (p.H80R) and c.248T>C (p.F83S), suggesting this region may be a mutational hotspot with critical importance for protein function .

Functional studies suggest that pathogenic variants in TRAPPC2 lead to sedlin protein misfolding, affecting either Golgi integrity or collagen trafficking pathways . This disruption ultimately impairs the secretion of extracellular matrix proteins by chondrocytes, explaining the skeletal manifestations of X-linked SEDT .

How should researchers approach inheritance analysis for TRAPPC2-related disorders?

TRAPPC2-related disorders follow an X-linked recessive inheritance pattern, requiring specific methodological approaches for pedigree analysis:

  • Carrier detection: Female carriers typically have one normal and one mutated TRAPPC2 allele, necessitating heterozygosity testing. NGS with sufficient depth is crucial to detect allelic imbalances

  • X-inactivation studies: Consider analyzing X-chromosome inactivation patterns in female carriers, as skewed inactivation may explain rare cases of mild symptoms in carriers

  • Pedigree mapping: Document multiple generations focusing on maternal lineage transmission, noting that affected males cannot transmit the condition to sons

  • Genetic counseling methodology: For families with identified TRAPPC2 mutations, offer testing to females of childbearing age. In one documented case, two female family members (III-1 and III-2) underwent targeted NGS and tested negative, ruling out carrier status and the possibility of having affected children

  • Mosaicism consideration: In cases with no apparent family history, consider gonadal mosaicism in the mother as a potential explanation

Given the X-linked inheritance pattern, males require only one altered copy of the gene to manifest the condition, while females require mutations in both copies (extremely rare). This explains why X-linked SEDT occurs almost exclusively in males .

What experimental approaches can be used to characterize the functional impact of novel TRAPPC2 variants?

For rigorous characterization of novel TRAPPC2 variants, researchers should employ a multi-tiered experimental approach:

  • Structural biology techniques:

    • Generate 3D protein models using tools like SWISS-model to predict how mutations affect protein folding and interaction domains

    • Consider X-ray crystallography or cryo-EM to determine precise structural changes in the mutant protein

    • Implement molecular dynamics simulations to predict functional consequences of amino acid substitutions

  • Cellular trafficking assays:

    • Develop fluorescently tagged procollagen constructs to quantify ER-to-Golgi transport in cells expressing wild-type versus mutant TRAPPC2

    • Employ pulse-chase experiments to measure secretion rates of extracellular matrix proteins

    • Use live-cell imaging to visualize vesicular trafficking dynamics in real-time

  • Biochemical interaction studies:

    • Perform co-immunoprecipitation experiments to assess how mutations affect TRAPPC2 interactions with known partners like Alpha-enolase and CLIC1

    • Use yeast two-hybrid or proximity labeling techniques to identify potentially disrupted protein-protein interactions

    • Quantify TRAPP complex assembly and stability using size-exclusion chromatography

  • Transcriptomics approaches:

    • Implement RNA-seq to identify dysregulated genes and pathways in cells expressing mutant TRAPPC2

    • Consider examining the impact on MBP1 target genes, given TRAPPC2's reported role in blocking MBP1-mediated transcriptional repression

These methodologies should be applied in relevant cellular models such as primary chondrocytes, osteoblasts, or patient-derived induced pluripotent stem cells differentiated into skeletal lineages to maintain physiological relevance.

What are the methodological challenges in developing therapeutic interventions for TRAPPC2-related disorders?

Developing therapeutic interventions for TRAPPC2-related disorders presents several methodological challenges that researchers must address:

  • Growth hormone therapy considerations:

    • Although recombinant human growth hormone (rhGH) has been explored as a potential treatment for the short stature associated with X-linked SEDT, serious concerns exist regarding metabolic side effects

    • In one documented case, a 13-year-old boy with X-linked SEDT achieved 2.1 cm height gain over three months of rhGH treatment, but therapy was terminated due to increased glucose levels that normalized after discontinuation

    • Researchers hypothesize that X-linked SEDT patients may be particularly susceptible to hyperglycemia with rhGH treatment, suggesting a need for careful glucose monitoring protocols in any future trials

    • The pubertal growth spurt may confound assessment of rhGH efficacy, necessitating careful study design with appropriate controls

  • Gene therapy approach limitations:

    • The ubiquitous expression of TRAPPC2 across tissues poses challenges for targeted delivery

    • X-linked inheritance pattern creates complexities for germline modification approaches

    • The delayed onset of symptoms (appearing around ages 6-8) complicates timing of intervention and clinical endpoint selection

  • Drug development considerations:

    • The rarity of the condition (1 in 150,000-200,000) creates challenges for clinical trial recruitment

    • The progressive nature of joint pain and osteoarthritis symptoms requires long-term follow-up

    • Compounds targeting vesicular trafficking may have wide-ranging cellular effects beyond TRAPPC2 function

Given these challenges, researchers exploring therapeutic interventions should consider establishing international registries and biobanks for this rare condition, implementing standardized outcome measures, and developing robust preclinical models for initial efficacy testing.

How can researchers design cell-based assays to study TRAPPC2 function in collagen trafficking?

Developing robust cell-based assays to study TRAPPC2's role in collagen trafficking requires careful methodological considerations:

  • Selection of appropriate cellular models:

    • Primary chondrocytes represent the most physiologically relevant cell type but are challenging to maintain in culture

    • Immortalized chondrocyte cell lines offer reproducibility but may have altered trafficking dynamics

    • Patient-derived fibroblasts maintain the genetic background but may show tissue-specific differences

    • Genetically engineered cells with TRAPPC2 knockout/knockin can provide controlled experimental systems

  • Fluorescent reporter systems design:

    • Generate constructs expressing fluorescently tagged procollagen to visualize trafficking in real-time

    • Consider dual-color systems with organelle markers (e.g., ER-Tracker, Golgi-GFP) to precisely locate procollagen during transport

    • Implement photoactivatable fluorescent proteins to enable pulse-chase visualization of specific protein cohorts

  • Quantification methodologies:

    • Develop high-content imaging workflows with automated segmentation of cellular compartments

    • Implement FRAP (Fluorescence Recovery After Photobleaching) to measure dynamic transport rates

    • Utilize flow cytometry for large-scale quantification of surface-expressed versus intracellular collagen

  • Perturbation strategies:

    • Create isogenic cell lines with CRISPR-Cas9 encoding various TRAPPC2 mutations

    • Implement inducible expression systems to study acute versus chronic loss of TRAPPC2 function

    • Develop small molecule inhibitors of specific TRAPPC2 interactions as chemical biology tools

These assays should incorporate appropriate controls, including rescue experiments with wild-type TRAPPC2 to confirm specificity of observed defects, and comparison to other trafficking defects to establish specificity for the collagen secretory pathway.

What are the unexplored aspects of TRAPPC2 function beyond vesicular trafficking?

While TRAPPC2's role in vesicular trafficking is well-established, several unexplored aspects of its biology warrant investigation:

  • Transcriptional regulation functions:

    • TRAPPC2 can bind MBP1 and block its transcriptional repression capability, suggesting potential nuclear functions independent of trafficking

    • Research methodologies should include ChIP-seq to identify potential genomic binding sites and RNA-seq to characterize transcriptional programs affected by TRAPPC2 depletion

    • Protein localization studies using fractionation and imaging approaches should examine potential nuclear-cytoplasmic shuttling of TRAPPC2

  • Signaling pathway integration:

    • Investigate potential roles of TRAPPC2 in mechanotransduction signaling relevant to bone development

    • Examine interactions with established skeletal regulatory pathways such as BMP, Wnt, and FGF signaling

    • Consider phosphoproteomic approaches to identify potential post-translational modifications of TRAPPC2 in response to different stimuli

  • Developmental timing regulation:

    • The delayed onset of X-linked SEDT symptoms (appearing around ages 6-8) suggests developmental regulation of TRAPPC2 function

    • Temporal expression analysis across different developmental stages and tissues may reveal regulatory mechanisms

    • Investigation of potential interactions with growth plate signaling during childhood growth acceleration

  • Tissue-specific functions:

    • Beyond skeletal tissues, explore TRAPPC2 functions in other collagen-rich tissues like skin, tendons, and blood vessels

    • Implement tissue-specific conditional knockout models to distinguish primary from secondary effects

    • Consider single-cell approaches to identify cell populations particularly dependent on TRAPPC2 function

These investigations require interdisciplinary approaches combining biochemical, cellular, and in vivo methodologies to fully elucidate TRAPPC2's multifaceted biological roles.

How can advanced sequencing technologies enhance our understanding of TRAPPC2 regulation?

Next-generation sequencing technologies offer powerful approaches for investigating TRAPPC2 regulation at multiple levels:

  • Non-coding regulatory element identification:

    • Implement ATAC-seq to identify open chromatin regions that may contain enhancers controlling TRAPPC2 expression

    • Use ChIP-seq to map transcription factor binding sites in the TRAPPC2 promoter and enhancer regions

    • Consider chromosome conformation capture methods (Hi-C, 4C) to identify distant regulatory elements that interact with the TRAPPC2 locus

  • Post-transcriptional regulation analysis:

    • Apply RNA-seq with specialized library preparation to identify alternative splicing events affecting TRAPPC2

    • Implement CLIP-seq to identify RNA-binding proteins that regulate TRAPPC2 mRNA stability or translation

    • Consider ribosome profiling to examine translational efficiency of TRAPPC2 under different conditions

  • Epigenetic regulation profiling:

    • Perform bisulfite sequencing to map DNA methylation patterns at the TRAPPC2 locus

    • Use CUT&RUN or CUT&Tag to profile histone modifications associated with TRAPPC2 expression

    • Investigate X-chromosome inactivation patterns in female tissues to understand escape phenomena

  • Single-cell approaches:

    • Apply single-cell RNA-seq to identify cell populations with particularly high TRAPPC2 expression

    • Consider spatial transcriptomics to map TRAPPC2 expression patterns within complex tissues like growth plates

    • Implement trajectory analysis to understand dynamic regulation during differentiation processes

These advanced sequencing approaches should be coupled with functional validation experiments to confirm the biological significance of identified regulatory mechanisms.

What methodological approaches can be used to study the structure-function relationship of the TRAPPC2 protein?

Elucidating the structure-function relationship of TRAPPC2 (sedlin) requires a comprehensive set of methodological approaches:

  • High-resolution structural determination:

    • X-ray crystallography of wild-type and mutant sedlin proteins

    • Cryo-electron microscopy of the complete TRAPP complex to understand sedlin's position and interactions

    • NMR spectroscopy to characterize dynamic regions and binding interfaces

    • Hydrogen-deuterium exchange mass spectrometry to identify conformationally flexible regions

  • Structure-guided mutagenesis:

    • Generate systematic alanine scanning mutants across the protein to identify functionally critical residues

    • Create chimeric proteins by swapping domains with related TRAPP components to determine specificity determinants

    • Introduce known disease-causing mutations (e.g., c.260A>C, p.H87P) to correlate structural changes with functional defects

  • Protein-protein interaction mapping:

    • Perform crosslinking mass spectrometry to identify interaction interfaces within the TRAPP complex

    • Implement FRET-based biosensors to detect conformational changes upon binding partners

    • Consider BioID or APEX proximity labeling to identify the sedlin interactome in living cells

  • Molecular dynamics simulations:

    • Conduct all-atom simulations to predict how disease-causing mutations affect protein stability and dynamics

    • Implement coarse-grained simulations to model interactions with membrane systems

    • Use enhanced sampling techniques to identify potential cryptic binding sites for future drug development

These methodologies should be applied in a complementary manner to build a comprehensive understanding of how TRAPPC2's structure dictates its function in cellular trafficking and potentially other biological processes.

What approaches can researchers use to develop improved diagnostic criteria for TRAPPC2-related disorders?

Developing robust diagnostic criteria for TRAPPC2-related disorders requires integration of clinical, radiographic, and molecular data:

  • Standardized clinical assessment tools:

    • Develop validated questionnaires specifically addressing X-linked SEDT symptoms

    • Establish growth chart references specific for X-linked SEDT patients to accurately characterize growth patterns

    • Create scoring systems for severity assessment incorporating degree of vertebral flattening, joint involvement, and functional limitations

  • Advanced imaging protocols:

    • Implement standardized radiographic positioning to ensure comparable vertebral assessments

    • Consider quantitative methods for measuring vertebral flattening and epiphyseal changes

    • Establish timing recommendations for radiographic evaluation based on age-of-onset patterns

  • Molecular diagnostic algorithms:

    • Develop tiered testing strategies starting with targeted TRAPPC2 sequencing

    • Include methodologies for detecting non-coding and regulatory region variants

    • Establish standards for variant interpretation specific to TRAPPC2 pathogenicity

  • Biomarker development:

    • Investigate collagen processing markers in blood or urine as potential non-invasive diagnostic indicators

    • Explore metabolomic profiles that might distinguish TRAPPC2-related disorders from other skeletal dysplasias

    • Consider proteomics approaches to identify secretory pathway disruptions indicative of TRAPPC2 dysfunction

These approaches should be validated in well-characterized patient cohorts and updated regularly as new knowledge about TRAPPC2-related phenotypes emerges.

What considerations should guide the development of patient registries for TRAPPC2-related disorders?

Establishing effective patient registries for TRAPPC2-related disorders requires careful methodological planning:

  • Registry structure and governance:

    • Implement international, multi-center design to maximize patient inclusion given the rarity of the condition (1 in 150,000-200,000)

    • Establish clear data ownership, sharing, and privacy policies compliant with international regulations

    • Develop sustainable funding models considering the long-term nature of registry maintenance

  • Data collection standardization:

    • Create uniform case report forms capturing key clinical variables, including:

      • Detailed growth measurements and proportions

      • Standardized radiographic assessments

      • Pain and functional limitation scoring

      • Quality of life measures

    • Establish minimum dataset requirements versus optional extended data collection

    • Implement standardized terminology and coding systems

  • Longitudinal follow-up methodologies:

    • Design protocols for regular patient assessment at defined intervals

    • Develop strategies to minimize loss to follow-up in this lifelong condition

    • Implement systems to capture treatment interventions and outcomes, such as growth hormone therapy results

  • Integration with biobanking:

    • Establish protocols for collection and storage of biological samples

    • Standardize processing methods for DNA, RNA, plasma, and potentially tissue samples

    • Create systems linking biospecimens with clinical data while maintaining privacy

  • Patient-reported outcome measures:

    • Incorporate validated quality of life assessments specific to skeletal disorders

    • Develop systems for remote data collection to reduce burden on patients with mobility issues

    • Include measures of psychosocial impact and educational/occupational outcomes

These registries should be designed with input from patients, clinicians, and researchers to ensure they meet the needs of all stakeholders while advancing knowledge about TRAPPC2-related disorders.

How can animal models contribute to understanding TRAPPC2 function and developing therapies?

Developing and utilizing animal models for TRAPPC2-related disorders requires consideration of several methodological approaches:

  • Selection of appropriate model organisms:

    • Mouse models: Generate conditional and inducible Trappc2 knockout mice to study tissue-specific and temporal requirements

    • Zebrafish models: Utilize CRISPR-Cas9 to create trappc2 mutants for high-throughput screening and visualization of skeletal development

    • Drosophila models: Exploit the evolutionary conservation of the TRAPP complex to study fundamental trafficking mechanisms

  • Disease-relevant phenotyping protocols:

    • Implement micro-CT analysis for detailed skeletal phenotyping

    • Develop methods for quantifying growth plate architecture and chondrocyte organization

    • Establish behavioral assessments for pain and mobility limitations

    • Consider gait analysis for functional evaluation of skeletal changes

  • Molecular and cellular analysis approaches:

    • Apply in vivo imaging of collagen trafficking using transgenic fluorescent reporters

    • Implement laser capture microdissection to isolate specific cell populations from growth plates

    • Conduct single-cell RNA-seq of growth plate chondrocytes to identify dysregulated pathways

  • Therapeutic testing paradigms:

    • Establish clear outcome measures and treatment windows based on disease progression

    • Consider combinatorial approaches targeting both trafficking defects and downstream consequences

    • Implement pharmacokinetic/pharmacodynamic studies to optimize delivery to skeletal tissues

  • Humanized model considerations:

    • Generate knock-in models with specific human TRAPPC2 mutations (e.g., c.260A>C, p.H87P)

    • Consider xenograft approaches with patient-derived cells in immunodeficient hosts

    • Develop chimeric models with human skeletal elements for increased translational relevance

These animal models should be developed with careful attention to human disease features and used in complementary fashion to maximize translational potential.

What methodological innovations might advance therapeutic development for TRAPPC2-related disorders?

Several innovative methodological approaches could accelerate therapeutic development for TRAPPC2-related disorders:

  • RNA-based therapeutic approaches:

    • Explore antisense oligonucleotides to modify splicing in cases with splice-site mutations

    • Consider targeted RNA editing to correct point mutations in TRAPPC2 mRNA

    • Investigate small activating RNAs to upregulate compensatory trafficking pathways

  • Small molecule screening strategies:

    • Develop high-throughput phenotypic screens based on collagen trafficking readouts

    • Implement fragment-based drug discovery targeting critical TRAPPC2 protein interactions

    • Consider repurposing approaches focusing on approved drugs that modulate secretory pathways

  • Advanced delivery technologies:

    • Explore bone-targeting nanoparticles to concentrate therapeutics in skeletal tissues

    • Investigate extracellular vesicle-based delivery systems for nucleic acid therapies

    • Consider implantable drug delivery systems for sustained local release

  • Pathway-based intervention approaches:

    • Target downstream consequences of TRAPPC2 dysfunction rather than the primary defect

    • Investigate modulation of ER stress responses to mitigate effects of protein trafficking defects

    • Consider anti-inflammatory approaches to address joint pain and osteoarthritis development

  • Precision medicine frameworks:

    • Implement variant-specific therapeutic strategies based on molecular mechanism

    • Develop biomarker-guided treatment selection algorithms

    • Consider combination therapies addressing multiple aspects of disease pathophysiology

Unlike growth hormone therapy, which showed concerning effects on glucose homeostasis in an X-linked SEDT patient , these novel therapeutic approaches should be designed with careful consideration of potential off-target effects and evaluated in appropriate preclinical models before clinical translation.

How can systems biology approaches enhance our understanding of TRAPPC2 in cellular homeostasis?

Systems biology offers powerful frameworks for integrating multiple data types to understand TRAPPC2 function within broader cellular networks:

  • Multi-omics integration methodologies:

    • Combine transcriptomics, proteomics, and metabolomics data from TRAPPC2-deficient models

    • Implement computational methods to identify convergent pathways across different data types

    • Develop network biology approaches to position TRAPPC2 within the cellular interactome

  • Quantitative pathway modeling:

    • Develop mathematical models of vesicular trafficking incorporating TRAPPC2 function

    • Implement ordinary differential equation-based models of collagen processing pathways

    • Use agent-based modeling to simulate emergent properties of skeletal development

  • Perturbation biology approaches:

    • Conduct systematic genetic interaction screens using CRISPR interference in TRAPPC2-deficient backgrounds

    • Implement chemical-genetic approaches to identify synthetic interactions

    • Develop scalable phenotypic readouts for high-dimensional perturbation studies

  • Temporal dynamics analysis:

    • Apply time-series experiments to characterize system responses to TRAPPC2 perturbation

    • Implement pulse-chase proteomics to quantify protein turnover rates in trafficking pathways

    • Develop livecell imaging approaches with computational image analysis for trafficking dynamics

  • Multi-scale modeling integration:

    • Link molecular dynamics simulations of TRAPPC2 structure with cellular trafficking models

    • Connect cellular phenotypes to tissue-level growth and development models

    • Develop mechanistic models explaining the delayed onset of skeletal manifestations in X-linked SEDT

Product Science Overview

Introduction

Trafficking Protein Particle Complex 2 (TRAPPC2), also known as Sedlin, is a crucial component of the trafficking protein particle (TRAPP) complex. This complex plays a significant role in the transport of proteins between cellular compartments, particularly between the endoplasmic reticulum (ER) and the Golgi apparatus .

Function and Importance

TRAPPC2 is involved in the transport of large proteins, such as procollagens, from the ER to the Golgi apparatus. This transport is essential for the proper processing and modification of these proteins, which are later transformed into mature collagen. Collagen is vital for the strength and support of connective tissues, including skin, bone, cartilage, tendons, and ligaments .

Genetic Implications

Mutations in the TRAPPC2 gene can lead to X-linked spondyloepiphyseal dysplasia tarda, a condition that impairs bone growth and predominantly affects males. This condition usually manifests between the ages of 6 and 10 and is characterized by a lack of functional Sedlin protein, which disrupts the transport of procollagen and reduces the amount of mature collagen in cells .

Preparation Methods

The preparation of recombinant TRAPPC2 involves the expression of the protein in a suitable host, such as HEK293T cells. The recombinant protein is then purified using affinity chromatography techniques. The protein is typically stored at -80°C to maintain its stability and avoid repeated freeze-thaw cycles .

Synthetic Routes

The synthetic routes for TRAPPC2 involve the use of cDNA clones or amino acid sequences to express the protein in host cells. The protein is then captured through anti-DDK affinity columns followed by conventional chromatography steps to ensure high purity and concentration .

Chemical Reactions Analysis

TRAPPC2 is part of the TRAPP complex, which regulates multiple membrane trafficking pathways. The TRAPP complexes exist in three forms, each activating specific GTPases such as Ypt1 or RAB1. These complexes play a crucial role in the secretory pathway, ensuring the directionality and fidelity of protein transport from the ER to the Golgi complex and beyond .

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.