Recombinant Human Trafficking protein particle complex subunit 11 (TRAPPC11), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice is requested in advance. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The specific tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
TRAPPC11; C4orf41; Trafficking protein particle complex subunit 11
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
TRAPPC11
Uniprot No.

Target Background

Function
Involved in the early stages of endoplasmic reticulum to Golgi apparatus protein trafficking.
Gene References Into Functions
  1. Recessive TRAPPC11 and GOSR2 mutations are linked to congenital muscular dystrophy and alpha-dystroglycan hypoglycosylation. PMID: 29855340
  2. A novel TRAPPC11 mutation expands the previously described myopathy phenotype, notably characterized by achalasia, alacrima, and neurological and muscular manifestations. PMID: 27707803
  3. TRAPPC11 plays a role in protein glycosylation and lipid-linked oligosaccharides biosynthesis. PMID: 26912795
  4. Homozygous TRAPPC11 mutations cause a form of autosomal-recessive, slowly progressive limb girdle muscular dystrophy with childhood onset and elevated serum creatine kinase levels. PMID: 23830518
Database Links

HGNC: 25751

OMIM: 614138

KEGG: hsa:60684

STRING: 9606.ENSP00000335371

UniGene: Hs.443240

Involvement In Disease
Limb-girdle muscular dystrophy 2S (LGMD2S)
Protein Families
TRAPPC11 family
Subcellular Location
Golgi apparatus. Golgi apparatus, cis-Golgi network.

Q&A

What is the domain structure of TRAPPC11 and what functions do these domains serve?

TRAPPC11 contains several functionally significant domains that have been characterized through mutation studies and functional analyses:

  • Foie gras domain (amino acids 263-561): This evolutionarily conserved region is critical for proper protein function. In-frame deletions within this domain (p.Ala372_Ser429del) have been identified in patients with myopathy and neurodevelopmental disorders . This domain appears particularly important for post-Golgi trafficking, as mutations affect protein exit from the Golgi to the cell surface .

  • Gryzun domain: A well-conserved 60-amino acid stretch near the carboxy terminus. Missense mutations in this domain (p.Gly980Arg) have been identified in patients with limb girdle muscular dystrophy (LGMD) . This domain appears critical for proper binding to other TRAPP complex components .

  • Carboxy-terminal region: Recent evidence indicates this region is important for recruiting the ATG2B-WIPI4 complex required for autophagosome formation . Frameshift mutations affecting this region (p.Asp1127Valfs*47) significantly impair autophagy .

Multisequence alignments demonstrate that while the entire protein is well-conserved among mammals, the carboxy terminus shows reduced conservation in more distant species, suggesting higher eukaryotes may have evolved unique autophagy-related functions in this region .

What cellular processes does TRAPPC11 participate in?

TRAPPC11 participates in several essential cellular processes:

  • Membrane trafficking: TRAPPC11 is essential for proper ER-to-Golgi transport and protein exit from the Golgi to the cell surface. Fibroblasts from patients with TRAPPC11 mutations show delayed transport kinetics .

  • Golgi structure maintenance: TRAPPC11 is required for maintaining normal Golgi morphology. Depletion or mutation of TRAPPC11 leads to Golgi fragmentation visible by immunostaining with markers like GM130 .

  • Protein N-glycosylation: TRAPPC11 plays a unique role in protein glycosylation that appears distinct from other TRAPP complex components. Mutations affect glycosylation of various proteins including TRAP-α (a resident ER protein), alpha-dystroglycan, serum transferrin, and apoCIII .

  • Autophagy: TRAPPC11, particularly its carboxy terminus, is involved in autophagosome formation through recruitment of the ATG2B-WIPI4 complex .

These functions are interconnected but can be differentially affected by specific mutations, allowing researchers to dissect domain-specific roles.

What phenotypes are associated with TRAPPC11 mutations in humans?

TRAPPC11 mutations cause a spectrum of disorders characterized by:

  • Muscular phenotypes (present in all reported cases):

    • Limb girdle muscular dystrophy (LGMD)

    • Generalized myopathy

    • Progressive muscle weakness

  • Neurological features (variable presentation):

    • Intellectual disability

    • Infantile hyperkinetic movements

    • Ataxia

    • Cerebral atrophy

  • Additional systemic manifestations:

    • Scoliosis

    • Alacrima (inability to produce tears)

    • Hepatic steatosis

    • Eye development defects

Different mutations produce distinct phenotypic patterns. For example, the homozygous c.2938G>A (p.Gly980Arg) mutation within the gryzun domain has been identified in individuals with predominantly LGMD features, while the c.1287+5G>A splice-site mutation resulting in a 58 amino acid in-frame deletion in the foie gras domain is associated with a more complex phenotype including myopathy, movement disorders, and intellectual disability .

How can researchers experimentally assess TRAPPC11's role in membrane trafficking?

To investigate TRAPPC11's role in membrane trafficking, researchers can employ several complementary approaches:

  • Trafficking kinetics assays:

    • Measure the transit time of cargo proteins through the secretory pathway

    • Transferrin receptor recycling assays to assess recycling endosome function

    • VSV-G protein trafficking from ER to Golgi and plasma membrane

  • Golgi morphology assessment:

    • Immunostaining with Golgi markers such as GM130

    • Quantification of Golgi fragmentation patterns

    • Electron microscopy for ultrastructural analysis

  • Rescue experiments:

    • Transfection of TRAPPC11-deficient cells with wild-type TRAPPC11 to assess restoration of normal trafficking

    • Domain-specific constructs to identify critical regions for trafficking function

  • Protein-protein interaction studies:

    • Co-immunoprecipitation to assess binding to other TRAPP complex components

    • Comparison of wild-type versus mutant TRAPPC11 interaction profiles

TRAPPC11 variants display distinct trafficking defects. For example, cells harboring the p.Ala372_Ser429del variant show normal ER-to-Golgi transport but dramatically delayed exit from the Golgi to the cell surface , while other variants may affect multiple trafficking steps .

What are the mechanistic links between TRAPPC11 dysfunction and defective protein glycosylation?

TRAPPC11 plays a unique role in protein glycosylation that appears distinct from its membrane trafficking function. The mechanisms include:

  • Effects on glycosylation precursors:

    • Zebrafish trappc11 mutants show reduced levels of lipid-linked oligosaccharides

    • Upregulation of genes involved in terpenoid biosynthesis (precursor for dolichol, the carbohydrate carrier in glycosylation)

  • Protein-specific glycosylation defects:

    • TRAP-α (ER-resident protein) shows partial glycosylation in patient fibroblasts with various TRAPPC11 mutations

    • Alpha-dystroglycan glycosylation defects in muscle tissue

    • Altered glycosylation of serum transferrin and apoCIII

  • TRAPPC11-specific effect:

    • The glycosylation defect appears specific to TRAPPC11 dysfunction and is not observed with mutations in other TRAPP components like TRAPPC12

  • Connection to ER stress:

    • Modest increases in unfolded protein response (UPR) gene expression (including XBP1) are observed in some TRAPPC11 variants

    • This suggests glycosylation defects may trigger ER stress pathways

Experimental approaches to study this connection include glycosylation pattern analysis using glycosylation-sensitive antibodies, glycosidase sensitivity assays, and analysis of UPR pathway activation markers.

How does TRAPPC11 contribute to autophagy and what experimental approaches can be used to study this?

TRAPPC11 plays a role in autophagosome formation, with its carboxy terminus being particularly important:

  • Functional role:

    • The carboxy terminus appears to recruit the ATG2B-WIPI4 complex

    • This complex is required for sealing isolation membranes into enclosed autophagosomes

    • Not all TRAPPC11 variants affect autophagy equally; those affecting the carboxy terminus (e.g., p.Asp1127Valfs*47) show the most significant autophagy impairment

  • Experimental approaches:

    MethodDescriptionAdvantagesLimitations
    LC3-II/LC3-I ratioWestern blot analysis of autophagosome marker conversionQuantitative, widely acceptedSnapshot measurement
    p62/SQSTM1 accumulationWestern blot or immunofluorescence of autophagy substrateIndicates autophagic fluxCan be affected by expression changes
    Tandem fluorescent-tagged LC3mRFP-GFP-LC3 to distinguish autophagosomes from autolysosomesDifferentiates formation vs. fusion defectsRequires specialized microscopy
    Electron microscopyUltrastructural analysis of autophagosome morphologyDirect visualization of structuresLabor-intensive, requires expertise
    ATG2B-WIPI4 recruitmentCo-immunoprecipitation or fluorescence microscopyDirectly tests TRAPPC11 functionComplex to interpret
  • Domain-specific analysis:

    • Structure-function studies using TRAPPC11 truncations or domain-specific mutations

    • Rescue experiments with wild-type vs. mutant constructs in TRAPPC11-deficient cells

These approaches can help researchers understand the specific role of TRAPPC11 in autophagy and how defects in this process contribute to disease pathogenesis.

How do different TRAPPC11 variants affect protein interactions and complex formation?

TRAPPC11 variants can differentially impact protein interactions and TRAPP complex formation:

  • Domain-specific effects:

    • Gryzun domain mutations (e.g., p.Gly980Arg): Impair binding to other TRAPP components despite normal TRAPPC11 protein levels

    • Foie gras domain mutations: May have less impact on complex formation but affect downstream functions

    • Carboxy-terminal mutations: Disrupt both complex formation and specific interactions with autophagy machinery

  • Functional consequences:

    • Reduced GEF (guanine nucleotide exchange factor) activity for Rab GTPases

    • Altered Golgi morphology and trafficking

    • Compromised autophagosome formation

  • Protein stability effects:

    • Some mutations (e.g., c.1287+5G>A in subject S4) result in unstable protein that is undetectable by western analysis

    • Others (p.Gly980Arg) produce stable protein but with impaired function

To study these interactions, researchers can use co-immunoprecipitation, proximity labeling techniques, and structural biology approaches. Understanding how different mutations affect protein interactions can provide insights into the molecular basis of phenotypic variability in TRAPPC11-related disorders.

What is the evidence for digenic inheritance involving TRAPPC11?

Recent research has identified potential digenic inheritance involving TRAPPC11:

  • TRAPPC11 and TTN co-segregating variants:

    • A family with limb-girdle muscular dystrophy showed co-segregation of TRAPPC11 c.3092C>G (p.Pro1031Arg) and TTN c.19481T>G (p.Leu6494Arg) variants with disease phenotype

    • Both variants are rare in population databases (frequencies <0.001 in 1000 Genomes, NHLBI-ESP6500, and ExAC)

    • Bioinformatic predictions suggest both variants are potentially pathogenic ("disease causing" by MutationTaster and "possibly damaging" by PolyPhen-2)

  • Functional connections:

    • TRAPPC11 is co-expressed with annexin A7 gene (ANXA7) and centrosomal protein 135 gene (CEP135), both of which physically interact with TTN

    • This suggests potential functional relationships between these proteins

  • Experimental approaches to study digenic inheritance:

    • Generate cellular or animal models with mutations in each gene individually and in combination

    • Compare phenotypic severity between single and double mutants

    • Investigate molecular interactions between the affected pathways

    • Test whether correcting either genetic defect alone is sufficient to rescue the phenotype

This emerging evidence highlights the importance of considering genetic modifiers and digenic inheritance when studying TRAPPC11-related disorders.

What cellular models are most appropriate for studying TRAPPC11 function?

Several cellular models have been employed to study TRAPPC11 function, each with distinct advantages:

  • Patient-derived fibroblasts:

    • Contain naturally occurring mutations at physiological expression levels

    • Allow direct correlation with patient phenotypes

    • Limitations include potential tissue-specificity issues (fibroblasts vs. affected tissues like muscle)

  • Established cell lines with TRAPPC11 knockdown/knockout:

    • HeLa cells with siRNA knockdown of TRAPPC11

    • CRISPR/Cas9-engineered knockout lines

    • Provide cleaner genetic background for mechanistic studies

    • Allow controlled expression of wild-type or mutant rescue constructs

  • Drosophila S2 cells:

    • RNAi knockdown models demonstrate conservation of TRAPPC11 function

    • Shows TRAPPC11 requirement for Golgi exit

  • Primary muscle cells or differentiated myotubes:

    • More relevant for studying muscular pathology

    • Can be derived from patient biopsies or through iPSC differentiation

  • iPSC-derived models:

    • Patient-specific cells can be differentiated into relevant cell types

    • Allow study of tissue-specific effects in neurons or muscle cells

When selecting a model system, researchers should consider the specific aspect of TRAPPC11 function they wish to study, as different mutations may have cell type-specific effects.

How can researchers differentiate between primary and secondary effects of TRAPPC11 dysfunction?

Distinguishing primary from secondary effects of TRAPPC11 dysfunction is crucial for understanding disease mechanisms:

  • Time-course experiments:

    • Use inducible knockdown or rapid protein degradation systems

    • Monitor cellular changes chronologically to identify earliest effects

  • Domain-specific mutational analysis:

    • Compare effects of mutations in different domains

    • Common effects across mutations likely represent primary consequences

    • Domain-specific effects help delineate specific functions

  • Rescue experiments:

    • Complementation with wild-type TRAPPC11 should reverse both primary and secondary effects

    • Domain-specific constructs can identify critical regions for specific functions

    • Timing of rescue can help distinguish immediate vs. downstream effects

  • Comparison with other TRAPP component mutations:

    • Effects common to multiple TRAPP component mutations likely reflect core complex functions

    • Effects specific to TRAPPC11 suggest unique roles

  • Pathway inhibition studies:

    • Use of chemical inhibitors or genetic manipulation of downstream pathways

    • If inhibiting a pathway prevents a phenotype in TRAPPC11-deficient cells, that phenotype is likely secondary

These approaches can help researchers build a mechanistic understanding of how TRAPPC11 dysfunction leads to disease, potentially identifying therapeutic targets.

What techniques can be used to study protein glycosylation defects in TRAPPC11-deficient cells?

Several techniques can be employed to characterize and quantify glycosylation defects:

  • Western blot analysis of glycoprotein mobility:

    • Assess migration patterns of glycoproteins like TRAP-α

    • Compare with enzymatically deglycosylated controls

    • Observe for intermediate forms indicating partial glycosylation

  • Glycosidase sensitivity assays:

    • Treatment with endoglycosidases (PNGase F, Endo H)

    • Different sensitivity patterns indicate glycosylation type and completeness

  • Lectin binding assays:

    • Different lectins bind specific glycan structures

    • Flow cytometry or microscopy-based quantification

    • Western blotting with lectin probes

  • Mass spectrometry:

    • Detailed characterization of glycan structures

    • Quantitative comparison of glycoforms

    • Site-specific glycosylation analysis

  • Metabolic labeling of glycans:

    • Pulse-chase experiments with radioactive or chemical labels

    • Track glycan synthesis and processing kinetics

  • Analysis of specific glycoproteins:

    • TRAP-α (resident ER protein with trafficking-independent glycosylation)

    • Alpha-dystroglycan (relevant to muscle pathology)

    • Serum proteins like transferrin and apoCIII

These techniques can help researchers characterize the specific nature and extent of glycosylation defects caused by different TRAPPC11 variants.

What are the most promising therapeutic approaches for TRAPPC11-related disorders?

Based on our current understanding of TRAPPC11 function and disease mechanisms, several therapeutic approaches merit investigation:

  • Targeting protein glycosylation:

    • Enhancing N-glycosylation pathway components

    • Stabilizing partially glycosylated proteins

    • Addressing downstream consequences of glycosylation defects

  • Enhancing membrane trafficking:

    • Small molecules that promote vesicular transport

    • Targeting compensatory trafficking pathways

    • Reducing ER stress caused by trafficking defects

  • Modulating autophagy:

    • Autophagy enhancers for variants with autophagy defects

    • Reducing accumulation of autophagic substrates

  • Gene therapy approaches:

    • AAV-mediated gene replacement for muscle-specific expression

    • Exon skipping for specific splice-site mutations

    • CRISPR-based correction of common mutations

  • Combinatorial approaches for digenic cases:

    • Simultaneous targeting of multiple affected pathways

    • Personalized approaches based on specific variant combinations

These therapeutic strategies should be evaluated in appropriate model systems before clinical translation. The tissue-specific manifestations of TRAPPC11 dysfunction suggest that targeted delivery to affected tissues may be necessary for effective treatment.

What are the key unresolved questions in TRAPPC11 research?

Despite significant advances, several important questions remain in TRAPPC11 research:

  • Structure-function relationships:

    • Complete structural characterization of TRAPPC11 domains

    • Mechanism of interaction with other TRAPP components

    • Structural basis for different functions (trafficking vs. autophagy)

  • Tissue specificity:

    • Why mutations predominantly affect muscle despite ubiquitous expression

    • Role in specialized cell types (neurons, muscle, liver)

  • Developmental aspects:

    • Function during development vs. adult homeostasis

    • Developmental origins of neurodevelopmental phenotypes

  • Genotype-phenotype correlations:

    • Mechanisms underlying phenotypic variability among patients with similar mutations

    • Role of genetic modifiers and environmental factors

  • Therapeutic targets:

    • Most promising molecular targets for intervention

    • Biomarkers for monitoring disease progression and treatment response

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 2025 TheBiotek. All Rights Reserved.