SSR4 Human

Signal Sequence Receptor, Delta Human Recombinant
Shipped with Ice Packs
In Stock

Description

SSR4 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 144 amino acids (24-144 a.a.) and having a molecular mass of 16.1kDa.
SSR4 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
SSR4, a member of the TRAP-delta family, serves as the delta subunit within the translocon-associated protein complex. This complex plays a crucial role in protein translocation across the endoplasmic reticulum membrane. Located in the Xq28 region, the SSR4 gene exhibits a compact head-to-head arrangement with the gene encoding isocitrate dehydrogenase 3 (NAD+) gamma. Notably, both genes share a common CpG-embedded bidirectional promoter.
Description
Recombinant human SSR4, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 144 amino acids (specifically, residues 24-144). With a molecular weight of 16.1 kDa, this protein features a 23-amino acid His-tag fused to its N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, sterile, and filtered solution.
Formulation
The SSR4 protein solution is provided at a concentration of 1 mg/ml. It is formulated in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To ensure optimal stability during long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA. It is important to avoid repeated freeze-thaw cycles.
Purity
The purity of the SSR4 protein is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
TRAPD, Translocon-associated protein subunit delta, TRAP-delta, Signal sequence receptor subunit delta SSR-delta.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEACLEPQ ITPSYYTTSD AVISTETVFI VEISLTCKNR VQNMALYADV GGKQFPVTRG QDVGRYQVSW SLDHKSAHAG TYEVRFFDEE SYSLLRKAQR NNEDISIIPP LFTVSVDHRG TWNG.

Q&A

What is the genomic location and structure of the human SSR4 gene?

SSR4 is a protein-coding gene located on the X chromosome in the Xq28 region. It is arranged head-to-head with the IDH3G gene, sharing a bidirectional promoter located between them. The gene consists of six exons and spans approximately 70 kb. While alternative splicing of exon 5 has not been observed in humans, transcript variants lacking this exon have been documented in other species including Xenopus laevis and Mus musculus . When investigating SSR4 structure, researchers should employ genomic analysis techniques including targeted sequencing and comparison with orthologous genes across species to identify conserved domains.

What is the primary function of SSR4 protein in human cells?

SSR4 encodes the Translocon-associated protein subunit delta (TRAP-delta), which plays a critical role in protein secretion pathways . Functionally, SSR4 appears essential for proper glycosylation processes, as evidenced by the glycosylation abnormalities observed in patients with SSR4 mutations. To study SSR4 function experimentally, researchers should consider subcellular localization techniques using fusion proteins (such as GFP::SSR4) combined with specific cellular compartment markers. Additionally, analyzing protein-protein interactions through co-immunoprecipitation or proximity labeling approaches can reveal functional partners of SSR4 in secretory pathways.

How does SSR4 contribute to normal glycosylation processes?

SSR4 mutations result in abnormal transferrin glycosylation profiles, specifically type 1 serum sialotransferrin patterns, indicating defects in early glycosylation pathways located in the cytosol or endoplasmic reticulum (designated as CDG-I) . Methodologically, researchers investigating SSR4's role in glycosylation should employ glycoprotein analysis techniques such as isoelectric focusing of transferrin, mass spectrometry of N-glycans, and pulse-chase experiments with glycosylation precursors to track the specific steps affected by SSR4 dysfunction.

What laboratory methods are most effective for diagnosing SSR4-CDG?

Diagnosis of SSR4-CDG relies on a combination of biochemical and genetic approaches. Transferrin isoelectric focusing should be performed first to detect the characteristic type 1 pattern (CDG-I) . This should be followed by molecular genetic testing, particularly whole-exome sequencing focused on the X chromosome, to identify mutations in the SSR4 gene. Researchers should be aware that in many cases, the abnormal glycosylation pattern in serum transferrin may be only slightly above normal cutoff ranges, requiring careful laboratory analysis and interpretation . When designing diagnostic studies, researchers should include controls with other types of CDG to establish clear differentiating criteria specific to SSR4-CDG.

What is the spectrum of clinical presentations in SSR4-CDG patients?

SSR4-CDG patients consistently present with a constellation of clinical features including hypotonia, failure to thrive, developmental delay, and dysmorphic traits . Notably, patients also exhibit connective tissue abnormalities resembling those seen in connective tissue disorders, including redundant skin, joint laxity, blue sclerae, and vascular tortuosity . Research protocols investigating SSR4-CDG should employ standardized neurodevelopmental assessments, detailed physical examinations focusing on connective tissue features, and longitudinal monitoring to document the natural history of the disease.

How can researchers differentiate SSR4-CDG from other congenital disorders of glycosylation?

Differentiating SSR4-CDG from other CDGs requires a multifaceted approach. While all CDGs may present with overlapping clinical features, SSR4-CDG specifically demonstrates X-linked inheritance, consistent connective tissue abnormalities, and a characteristic type 1 transferrin pattern that may be only slightly abnormal . To distinguish SSR4-CDG in research studies, investigators should implement comprehensive glycomics approaches including:

Diagnostic ApproachApplication in SSR4-CDGDistinguishing Features
Transferrin Isoform AnalysisPrimary screening testMild type 1 pattern (CDG-I)
Genetic TestingConfirmation of diagnosisX-linked mutations in SSR4
Clinical PhenotypingPatient classificationConnective tissue involvement
Glycoprotein AnalysisBiochemical characterizationSpecific N-glycan abnormalities

What molecular mechanisms underlie the connective tissue abnormalities in SSR4-CDG?

The connective tissue abnormalities in SSR4-CDG patients (redundant skin, joint laxity, blue sclerae, and vascular tortuosity) suggest impaired glycosylation of extracellular matrix proteins . To investigate these mechanisms, researchers should employ fibroblast cultures from patients to analyze collagen and elastin production, glycosylation, and secretion. Experimental approaches should include:

  • Quantitative PCR and western blotting to assess expression levels of key extracellular matrix proteins

  • Mass spectrometry to characterize glycosylation patterns of secreted proteins

  • Mechanical testing of patient-derived fibroblast matrices to assess structural integrity

  • 3D organoid models to evaluate tissue architecture and mechanical properties

These methodologies would provide insights into how SSR4 deficiency affects extracellular matrix composition and function.

What is the role of SSR4 in cancer development and progression?

Recent research indicates that SSR4 may serve as a biomarker in esophageal squamous cell carcinoma (ESCC) . Unlike adjacent non-cancerous tissues, SSR4 is overexpressed in ESCC tissues, as validated by both RT-qPCR and immunohistochemical staining. Expression levels correlate with N stage, lymph node metastasis, and AJCC TNM classification stage, with patients exhibiting low SSR4 expression demonstrating more favorable prognosis . Single-cell analysis reveals highest SSR4 expression in tumor plasma cells, suggesting potential immune system involvement.

Researchers investigating SSR4's role in cancer should implement:

  • Comprehensive transcriptomic and proteomic profiling of SSR4 in diverse tumor types

  • Single-cell RNA sequencing to identify cell populations with differential SSR4 expression

  • CRISPR-based functional assays to assess the impact of SSR4 knockdown/overexpression on cancer cell phenotypes

  • Correlative studies with clinical outcomes across multiple cancer cohorts

How does SSR4 interact with the immune system in disease contexts?

Analysis of immune infiltration has highlighted associations between SSR4 gene expression and the infiltration of specific immune cells, particularly plasma cells . When investigating immune interactions in SSR4-related pathologies, researchers should employ:

  • CellChat or similar computational tools to analyze cell-cell communication networks

  • Flow cytometry and single-cell transcriptomics to characterize immune cell populations

  • Cytokine profiling in patient samples and experimental models

  • Co-culture systems combining SSR4-deficient/overexpressing cells with immune cells

These approaches would help elucidate the immunological implications of SSR4 dysregulation in both developmental disorders and cancer.

What are the optimal models for studying SSR4 function in vitro and in vivo?

Selecting appropriate experimental models is crucial for SSR4 research. Based on available literature, researchers should consider:

In vitro models:

  • Patient-derived fibroblasts from SSR4-CDG individuals

  • CRISPR-engineered SSR4 knockout/knockdown cell lines

  • Inducible expression systems to control SSR4 levels

  • iPSC-derived organoids to model tissue-specific effects

In vivo models:

  • Conditional SSR4 knockout mice (note that complete knockout may be embryonic lethal)

  • Xenopus models, given the documented alternative splicing in this organism

  • Drosophila models for high-throughput genetic interaction studies

For subcellular localization studies, researchers should employ fusion proteins (such as GFP::SSR4) combined with nucleus-specific dyes like DAPI, followed by visualization through laser scanning confocal microscopy (LSCM) .

What techniques are most effective for measuring SSR4 expression and function?

When quantifying SSR4 expression and function, researchers should employ multiple complementary approaches:

  • Real-time quantitative PCR (qPCR) using specific primers for SSR4, with 18S rRNA as internal control

  • Western blotting with monoclonal antibodies specific to SSR4

  • Immunohistochemistry for tissue localization

  • Functional assays measuring glycosylation efficiency using reporter proteins

  • Protein-protein interaction studies via co-immunoprecipitation or proximity labeling

For gene editing experiments, homologous recombination approaches have been successfully employed for SSR4 deletion and complementation .

How can researchers resolve contradictory findings in SSR4 research?

Contradictory findings in SSR4 research may arise from differences in experimental systems, detection methods, or biological contexts. To address these, researchers should:

  • Standardize experimental conditions and readouts across laboratories

  • Employ multiple complementary techniques to validate key findings

  • Consider cell type-specific and developmental stage-specific effects

  • Account for potential compensatory mechanisms in knockout models

  • Implement systematic meta-analyses of published data to identify sources of variation

When inconsistencies persist, targeted experiments designed specifically to address the contradictions should be conducted, with careful attention to biological and technical variables.

How might single-cell technologies advance our understanding of SSR4 biology?

Single-cell technologies offer unprecedented opportunities to elucidate SSR4 functions in heterogeneous cell populations. Analysis of SSR4 expression in ESCC has already revealed highest expression levels in tumor plasma cells . Researchers should consider:

  • Single-cell RNA sequencing to map SSR4 expression across development and disease states

  • Single-cell proteomics to assess post-translational modifications dependent on SSR4

  • Spatial transcriptomics to understand SSR4 expression in tissue context

  • Single-cell glycomics to characterize cell-specific glycosylation patterns affected by SSR4

These approaches would provide insights into cell type-specific roles of SSR4 and potentially identify new therapeutic targets.

What therapeutic strategies might target SSR4-related pathologies?

Developing therapeutic approaches for SSR4-related disorders requires understanding of underlying molecular mechanisms. Researchers should explore:

  • Gene therapy approaches for SSR4-CDG, considering its X-linked nature

  • Small molecule modulators of glycosylation pathways to bypass SSR4 deficiency

  • Targeted therapies for cancers with SSR4 overexpression

  • Dietary interventions to modify glycosylation substrate availability

  • Connective tissue-targeted therapies addressing the extracellular matrix abnormalities

Experimental designs should include both in vitro high-throughput screening and appropriate animal models that recapitulate key aspects of human SSR4-related pathologies.

Product Science Overview

Introduction

Signal Sequence Receptor, Delta (SSR4), also known as the delta subunit of the translocon-associated protein complex, plays a crucial role in the translocation of proteins across the endoplasmic reticulum (ER) membrane. This protein is essential for the proper functioning of cellular processes and is involved in various biological pathways.

Structure and Function

The SSR4 gene encodes the delta subunit of the translocon-associated protein complex. This complex is responsible for the translocation of newly synthesized proteins across the ER membrane. The delta subunit, along with other subunits, forms a channel through which proteins can pass into the ER lumen. The SSR4 protein is located in the Xq28 region of the human genome and is arranged in a compact head-to-head manner with the isocitrate dehydrogenase 3 (NAD+) gamma gene .

Recombinant Human SSR4

Recombinant human SSR4 is produced using recombinant DNA technology, which involves inserting the SSR4 gene into a suitable expression system, such as E. coli or HEK293 cells. The recombinant protein is then purified and used for various research applications. The recombinant human SSR4 protein typically includes a His-tag or other tags to facilitate purification and detection .

Applications

Recombinant human SSR4 is used in various research applications, including:

  • Protein translocation studies: Understanding the mechanism of protein translocation across the ER membrane.
  • Protein-protein interactions: Investigating interactions between SSR4 and other proteins involved in the translocation process.
  • Disease research: Studying the role of SSR4 in diseases related to protein translocation defects.
Clinical Significance

Mutations in the SSR4 gene can lead to various diseases, including congenital disorders of glycosylation (CDG). CDG is a group of inherited metabolic disorders that affect the glycosylation of proteins and lipids. Patients with SSR4 mutations may exhibit symptoms such as developmental delay, intellectual disability, and other systemic manifestations .

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.