SH2D1A Human

SH2 domain containing 1A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

SH2D1A regulates immune responses through:

  • SLAM receptor modulation: Competes with phosphatases (SHP-2, SHIP) to enhance signaling via CD244, LY9, and SLAMF1

  • Cytotoxic lymphocyte regulation: Essential for NK/T-cell activation and apoptosis

  • Fyn kinase recruitment: Forms trimeric complexes with SLAM receptors to amplify downstream signaling

Key pathways influenced:

  1. NF-κB activation: Independent of SH2 domain, driving lymphocyte survival

  2. Dok1 interaction: Modulates tyrosine kinase signaling in hematopoietic cells

Disease Associations: X-Linked Lymphoproliferative Disease (XLP1)

Over 70 pathogenic SH2D1A mutations cause XLP1, characterized by:

  • EBV susceptibility: 58% develop fatal infectious mononucleosis

  • Lymphoproliferation: 25% risk of B-cell lymphoma

  • Dysgammaglobulinemia: Low IgG in 51.8% of patients

Common mutation types:

Mutation TypeClinical ImpactFrequency (%)Source
Missense (e.g., R55X)Impaired SAP-SLAM binding32%
Frameshift/TruncationsNonfunctional protein28%
Large deletionsComplete SAP loss (severe phenotypes)20%

Diagnostic and Therapeutic Insights

Genetic testing challenges:

  • NGS limitations: 71-bp deletions/16-bp insertions (e.g., c.162_201+31delinsTACAAGGACATATACA) require RNA studies for confirmation

  • Screening recommendations: Prioritize males with EBV-driven HLH or lymphoma

Treatment outcomes:

InterventionSurvival RateKey Findings
Allogeneic HSCT81.4%Restores immune function; 52-month median follow-up
Immunoglobulin therapy62.5%Temporary control for non-transplanted patients

Recent Research Advances

  • Structural studies: SH2D1A’s SH2 domain binds SLAM and Fyn simultaneously, enabling dual signaling

  • Novel variants: A 2022 case identified a complex splicing variant altering exon 2, linked to atypical EBV-negative XLP1

  • Therapeutic targets: SH2D1A-Fyn interaction inhibitors show promise in preclinical models

Product Specs

Introduction
SH2D1A, also known as Signaling Lymphocytic Activation Molecule (SLAM)-associated protein, functions as an inhibitor of SLAM self-association. Highly expressed in the thymus and lungs, with lower expression in the spleen and liver, SH2D1A plays a critical role in the bidirectional stimulation of T and B cells. Structurally, it comprises an SH2 domain and a short tail. Defects in the SH2D1A gene are linked to X-linked lymphoproliferative disease (XLPD), also known as Duncan disease, a rare congenital immunodeficiency characterized by severe complications following Epstein-Barr virus (EBV) infection.
Description
This product consists of the human recombinant SH2D1A protein, inclusive of a 20 amino acid His tag positioned at the N-terminus. Expressed in E. coli, it presents as a single, non-glycosylated polypeptide chain, encompassing 148 amino acids (specifically, amino acids 1 to 128). With a molecular weight of 16.3 kDa, the SH2D1A protein undergoes purification through proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The SH2D1A protein is supplied in a solution at a concentration of 1 mg/ml. The solution is buffered with 20mM Tris-HCl at a pH of 7.5 and further contains 1mM DTT and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept refrigerated at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (either HSA or BSA) to a final concentration of 0.1%. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the SH2D1A protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
SH2 domain-containing protein 1A, Duncan disease SH2-protein, Signaling lymphocytic activation molecule-associated protein, SLAM-associated protein, T-cell signal transduction molecule SAP, SH2D1A, DSHP, SAP, LYP, XLP, EBVS, IMD5, XLPD, MTCP1, FLJ18687, FLJ92177, SAP/SH2D1A.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MDAVAVYHGK ISRETGEKLL LATGLDGSYL LRDSESVPGV YCLCVLYHGY IYTYRVSQTE TGSWSAETAP GVHKRYFRKI KNLISAFQKP DQGIVIPLQY PVEKKSSARS TQGTTGIRED PDVCLKAP.

Q&A

What is the basic structure and normal function of the SH2D1A protein?

The SH2D1A gene encodes the signaling lymphocyte activation molecule (SLAM) associated protein (SAP), which is a 128-amino acid protein containing one Src homology 2 (SH2) domain . SAP primarily functions by interacting with SLAM family receptors to activate signaling pathways involved in immune cell control. At the molecular level, SAP competitively binds to SLAMs via its SH2 domain to regulate multiple immune processes .

The protein plays critical roles in:

  • Regulating cytotoxic lymphocytes

  • Development of natural killer T cells (iNKT cells)

  • Facilitating clearance of EBV-infected B cells via cytotoxic T lymphocytes and NK cells

  • Development of germinal centers

  • Production of immunoglobulin

  • Regulation of T cell restimulation-induced cell death

  • Maintenance of T cell homeostasis

SAP is primarily expressed in T cells, NK cells, and some EBV-positive Burkitt lymphoma-derived B cells .

How does SAP interact with other immune signaling molecules?

SAP functions as an adaptor protein that mediates protein-protein interactions within immune cells. It critically interacts with phosphorylated receptors of the SLAM family, including CD244 (2B4) . The SH2 domain of SAP recognizes and binds to specific phosphorylated tyrosine residues on these receptors.

These interactions regulate multiple downstream signaling cascades:

  • SAP can competitively bind to SLAMs via the SH2 domain, blocking the recruitment of other SH2-domain-containing molecules like SHP-2

  • SAP serves as a critical mediator in bidirectional stimulation of T and B cells

  • Recent evidence suggests potential involvement with PI3K-AKT-mTOR signaling pathway in the context of XLP-1

The binding affinity of SAP to phosphorylated receptors appears critical for its function, as demonstrated by the SH2D1A c.49G > A (p.E17K) variant, which showed >95% reduction in binding to phosphorylated CD244 receptor .

What types of SH2D1A mutations are associated with XLP-1, and how do they affect protein function?

More than 70 different mutations in the SH2D1A gene have been identified in patients with X-linked lymphoproliferative disease type 1 (XLP-1) . These mutations can be categorized based on their effects on the SAP protein:

Mutation TypeEffect on ProteinFunctional ConsequenceExample
Missense mutationsAltered amino acid sequenceImpaired binding to receptorsc.96G > T (p.R32S) - reduced SAP expression to 10.28%
Truncating mutationsAbnormally short proteinUnstable or nonfunctional protein-
Complete gene deletionsNo protein productionAbsence of SAP function-
Splice-site mutationsAbnormal splicingVariable protein expression-

Functionally, these mutations can disrupt SAP's:

  • Binding to SLAM family receptors

  • Competitive inhibition of other signaling molecules

  • Regulation of immune cell development and function

  • Control of apoptosis in lymphocytes

The c.96G > T mutation demonstrates how single amino acid substitutions can significantly impact protein structure and function, creating a hydrogen bond turnover at the mutation site that affects the protein's stability and binding capacity .

How does SAP deficiency lead to the characteristic immune dysregulation seen in XLP-1?

SAP deficiency disrupts immune regulation through several mechanisms:

  • Impaired EBV-specific immune responses: Without functional SAP, patients cannot mount appropriate cytotoxic responses against EBV-infected B cells, leading to uncontrolled proliferation of these cells and severe, sometimes fatal EBV infections .

  • Dysregulated NK and T cell function: SAP is crucial for NK and cytotoxic T cell functions, including cytotoxicity against virus-infected cells. Deficiency leads to impaired clearance of EBV-infected cells and excessive immune activation .

  • Abnormal lymphocyte apoptosis: SAP normally helps control immune reactions by triggering self-destruction (apoptosis) of lymphocytes when they are no longer needed. Loss of this function may contribute to lymphoproliferation and lymphoma development .

  • Defective germinal center formation: SAP deficiency impairs germinal center formation, contributing to the dysgammaglobulinemia observed in XLP-1 patients .

  • Aberrant PI3K-AKT-mTOR signaling: Recent research indicates that the PI3K-AKT-mTOR signaling pathway is fully activated in XLP-1 patients but inactive or only partially activated in healthy individuals or HLH patients without XLP-1, suggesting a role for this pathway in disease pathogenesis .

What methodologies are most effective for detecting and characterizing SH2D1A mutations?

Researchers employ multiple complementary approaches to detect and characterize SH2D1A mutations:

  • Genetic Sequencing:

    • Whole exome sequencing (WES) to identify novel mutations

    • Sanger sequencing for validation and familial testing

    • GATK (Genome Analysis TK) and ANNOVAR for variant detection and annotation

  • Protein Expression Analysis:

    • Flow cytometry to quantify SAP protein expression in lymphocytes

    • Western blot analysis for protein size and abundance assessment

  • Functional Assays:

    • Binding assays to assess SAP interaction with SLAM family receptors

    • Cellular cytotoxicity assays to evaluate NK and T cell function

    • Apoptosis assays to assess lymphocyte survival regulation

  • Structural Analysis:

    • Bioinformatics tools like PolyPhen-2 to predict mutation pathogenicity

    • SWISS-MODEL and SWISS-PDB Viewer for protein structure modeling

    • Analysis of hydrogen bonding and structural alterations

The combination of genetic, protein, functional, and structural analyses provides comprehensive characterization of SH2D1A variants and their biological significance. For example, in one study, researchers used flow cytometry to demonstrate that SAP expression was reduced to 10.28% in a patient with a novel c.96G > T mutation, while the patient's father and mother had 87.28% and 80.31% expression, respectively .

What cellular and animal models are available for studying SH2D1A function and XLP pathogenesis?

Several research models have been developed to study SH2D1A function and XLP pathogenesis:

Cellular Models:

  • Patient-derived primary cells: Lymphocytes from XLP patients provide direct insight into disease mechanisms but may be limited by availability and heterogeneity.

  • Cell lines with engineered SH2D1A mutations:

    • CRISPR/Cas9-modified T and NK cell lines

    • Lymphoblastoid cell lines from XLP patients

    • Cell lines with inducible SAP expression systems

  • EBV-challenged immune cell models: Systems to study the specific responses to EBV in the context of SAP deficiency.

Animal Models:

  • SH2D1A knockout mice: These recapitulate many aspects of human XLP, including:

    • Impaired NK and CD8+ T cell cytotoxicity

    • Defective NKT cell development

    • Abnormal cytokine production

    • Dysgammaglobulinemia

  • Humanized mouse models: Immunodeficient mice reconstituted with human immune cells (with or without SH2D1A mutations) can model human-specific aspects of disease, particularly EBV infection responses.

  • Knock-in models of specific mutations: These allow the study of particular SH2D1A variants found in patients, helping distinguish between complete loss-of-function and hypomorphic mutations.

When selecting models, researchers should consider the specific aspects of SAP function they wish to study, as different models may better recapitulate certain disease features than others.

How does the PI3K-AKT-mTOR pathway interact with SAP in normal and pathological conditions?

Recent research has revealed important connections between SAP and the PI3K-AKT-mTOR signaling pathway:

In normal conditions, SAP appears to modulate PI3K-AKT-mTOR signaling through its interactions with SLAM family receptors. The exact molecular mechanisms remain under investigation, but data suggest SAP may serve as a negative regulator of this pathway in certain contexts .

In XLP-1 pathological conditions:

  • The PI3K-AKT-mTOR signaling pathway is fully activated in XLP-1 patients

  • This pathway remains inactive or only partially activated in healthy individuals or non-XLP HLH patients

  • This differential activation suggests the pathway may play a significant role in XLP-1 pathogenesis

The activation status of this pathway may contribute to:

  • Excessive lymphocyte proliferation

  • Altered cytokine production

  • Resistance to apoptosis

  • Metabolic reprogramming of immune cells

These findings suggest potential therapeutic opportunities targeting PI3K-AKT-mTOR pathway components in XLP-1 patients, though further research is needed to fully elucidate the relationship between SAP deficiency and pathway dysregulation .

What protein-protein interactions are critical for SAP function and how are they disrupted in disease states?

SAP functions primarily through protein-protein interactions that are critical for immune regulation:

Key Interactions:

  • SAP-SLAM family receptor interactions: SAP binds via its SH2 domain to phosphorylated tyrosine motifs on SLAM family receptors (including SLAM/CD150, 2B4/CD244, NTB-A/SLAMF6, Ly9/CD229, and CRACC/CD319) .

  • Competitive binding with SHP-2: SAP competes with the phosphatase SHP-2 for binding to SLAM receptors, blocking SHP-2 recruitment and modifying downstream signaling .

  • Interaction with Fyn kinase: SAP can recruit and activate the Fyn tyrosine kinase to SLAM receptors, initiating downstream signaling cascades.

Disruption in Disease:
Mutations can disrupt these interactions through several mechanisms:

InteractionNormal FunctionDisruption in DiseaseConsequence
SAP-SLAM bindingSignal transduction initiationReduced binding affinity (e.g., >95% reduction with p.E17K variant) Impaired activation of downstream signaling
SAP-SHP-2 competitionPrevention of inhibitory signalingFailed competitionExcessive inhibitory signaling
SAP-Fyn recruitmentActivation of signaling cascadesFailed recruitmentBlocked downstream pathway activation

The c.49G > A variant demonstrates how subtle mutations can have profound effects: this variant produces normal levels of SAP protein, but binding to phosphorylated CD244 receptor is reduced by >95%, highlighting the critical importance of these protein-protein interactions in disease pathogenesis .

What are the challenges in diagnosing XLP-1, and how can functional assays improve diagnostic accuracy?

Diagnosing XLP-1 presents several challenges:

Diagnostic Challenges:

  • Variable expressivity: The same SH2D1A mutation can manifest differently in different individuals, even within the same family. For example, the SH2D1A c.49G > A variant caused fatal disease in one family member but three brothers carrying the same variant remained healthy .

  • Phenotypic overlap: XLP-1 shares features with other immunodeficiencies and lymphoproliferative disorders, including HLH from other causes.

  • Genetic complexity: Interpreting novel SH2D1A variants can be difficult, particularly for missense mutations where functional impact is uncertain.

  • Pre-EBV diagnosis: Diagnosing XLP-1 before EBV exposure is crucial but challenging without a positive family history.

Improving Diagnostic Accuracy Through Functional Assays:

These functional assays are especially important for variants of uncertain significance, where bioinformatic prediction alone may be insufficient to determine pathogenicity .

How can researchers differentiate between XLP-1 (SH2D1A deficiency) and XLP-2 (XIAP deficiency) in experimental settings?

Distinguishing between XLP-1 and XLP-2 is crucial for both research and clinical applications. Researchers can differentiate these conditions through several approaches:

Genetic Analysis:

  • XLP-1: Mutations in SH2D1A gene on Xq25

  • XLP-2: Mutations in XIAP (also known as BIRC4) gene on Xq25

Protein Expression Analysis:

  • Flow cytometry:

    • XLP-1: Reduced or absent SAP expression in lymphocytes

    • XLP-2: Reduced or absent XIAP expression in lymphocytes

    • Note: Some mutations may affect function without altering expression levels

Clinical and Pathological Features:

  • Lymphoma development:

    • XLP-1: Associated with lymphoma development (especially after EBV infection)

    • XLP-2: Lymphoma development is not typically observed

  • Recurrent HLH:

    • XLP-1: HLH typically occurs once, usually after EBV infection

    • XLP-2: Recurrent episodes of HLH are more common, and may occur without EBV infection

  • Gastrointestinal manifestations:

    • XLP-1: Not typically observed

    • XLP-2: Inflammatory bowel disease-like manifestations are common

Functional Assays:

  • NKT cell development:

    • XLP-1: Defective NKT cell development

    • XLP-2: Normal NKT cell development

  • Apoptosis pathway analysis:

    • XLP-1: Defects in SLAM-mediated signaling

    • XLP-2: Defects in NOD2-RIP2 signaling and TNF-mediated apoptosis

A comprehensive approach incorporating genetic, protein, clinical, and functional analyses provides the most accurate differentiation between these related but distinct disorders.

What are the most promising therapeutic targets emerging from SH2D1A research?

Research into SH2D1A function and XLP-1 pathogenesis has revealed several promising therapeutic targets:

  • PI3K-AKT-mTOR pathway inhibitors: Recent evidence showing full activation of the PI3K-AKT-mTOR pathway in XLP-1 patients suggests these components may be viable therapeutic targets . Specific inhibitors like:

    • PI3K inhibitors (e.g., idelalisib, duvelisib)

    • AKT inhibitors (e.g., MK-2206)

    • mTOR inhibitors (e.g., rapamycin, everolimus)

    These might help modulate the excessive immune activation in XLP-1.

  • SLAM receptor pathway modulation: Targeting SLAM family receptors or their downstream signaling components may compensate for SAP deficiency.

  • Gene therapy approaches: Delivering functional SH2D1A gene to patient-derived hematopoietic stem cells offers potential for long-term correction of the genetic defect.

  • EBV-directed therapies: Since EBV infection triggers severe manifestations in XLP-1, therapies targeting EBV, including:

    • EBV-specific T cell therapies

    • Anti-viral medications

    • Vaccination strategies

    These may help prevent or treat disease manifestations.

  • Targeted immunomodulation: Selective modulation of specific immune pathways dysregulated in XLP-1 may help control disease manifestations without complete immune suppression.

Each of these approaches merits further investigation in preclinical models before clinical translation. The PI3K-AKT-mTOR pathway appears particularly promising based on recent findings showing differential activation in XLP-1 patients compared to other conditions .

What methodological approaches are being developed to better model SH2D1A function in complex immune interactions?

Researchers are developing increasingly sophisticated approaches to model the complex immune interactions mediated by SAP:

  • Advanced organoid systems: Multi-cellular immune organoid systems incorporating various lymphocyte populations can better model the complex intercellular interactions mediated by SAP.

  • CRISPR/Cas9 genome editing: Precise engineering of specific SH2D1A mutations in relevant cell types allows direct study of mutation-specific effects on protein function and cellular phenotypes.

  • Single-cell analysis technologies:

    • Single-cell RNA sequencing to profile transcriptional changes in SAP-deficient cells

    • CyTOF (mass cytometry) to simultaneously analyze multiple signaling pathways at the single-cell level

    • Spatial transcriptomics to understand tissue-specific effects of SAP deficiency

  • Advanced imaging techniques:

    • Live-cell imaging to visualize SAP recruitment during immune synapse formation

    • FRET-based approaches to study SAP protein-protein interactions in real-time

    • Super-resolution microscopy to examine molecular-level interactions

  • Improved humanized mouse models: Next-generation humanized mice with more complete human immune reconstitution allow better modeling of human-specific aspects of SAP function, particularly in response to EBV infection.

  • Systems biology approaches: Integrating multi-omics data (genomics, transcriptomics, proteomics, metabolomics) provides comprehensive understanding of how SAP deficiency affects immune system function at multiple levels.

These methodological advances promise to provide deeper insights into the complex roles of SAP in immune regulation and may identify novel therapeutic targets for XLP-1 and related disorders.

Product Science Overview

Introduction

SH2 domain containing 1A (SH2D1A), also known as Signaling Lymphocytic Activation Molecule-Associated Protein (SLAM-associated protein or SAP), is a protein encoded by the SH2D1A gene in humans. This protein plays a crucial role in the immune system, particularly in the signaling pathways of T and B cells .

Structure and Function

The SH2D1A protein contains an SH2 (Src Homology 2) domain, which is a structurally conserved protein domain found in many intracellular signal-transducing proteins . The SH2 domain allows the protein to bind to phosphorylated tyrosine residues on other proteins, thereby modifying the function or activity of the SH2-containing protein . This binding is essential for the transmission of signals that control various cellular functions.

Role in Immune System

SH2D1A is primarily involved in the bidirectional stimulation of T and B cells. It associates with the signaling lymphocyte-activation molecule (SLAM), acting as an inhibitor by blocking the recruitment of the SH2-domain-containing signal-transduction molecule SHP-2 to its docking site . Additionally, SH2D1A can bind to other related surface molecules expressed on activated T, B, and NK cells, thereby modifying signal transduction pathways in these cells .

Clinical Significance

Mutations in the SH2D1A gene are associated with lymphoproliferative syndrome X-linked type 1 (XLP1), also known as Duncan disease . This rare immunodeficiency disorder is characterized by extreme susceptibility to infection with the Epstein-Barr virus (EBV), leading to severe mononucleosis and malignant lymphoma . Multiple transcript variants encoding different isoforms of SH2D1A have been identified, highlighting the complexity of its regulation and function .

Research and Applications

The recombinant form of SH2D1A is used in various research applications to study its role in immune signaling pathways and its involvement in diseases. Understanding the function and regulation of SH2D1A can provide insights into potential therapeutic targets for treating immune-related disorders.

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.