CD47 Human

CD47 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of CD47

CD47 comprises three key domains:

  • Extracellular N-terminal IgV domain: Mediates interactions with SIRPα and TSP-1 .

  • Five transmembrane helices: Forms a tightly packed core critical for structural stability .

  • Short cytoplasmic tail: Splices into four isoforms with tissue-specific roles .

Table 1: CD47 Isoforms and Tissue Distribution

IsoformCytoplasmic Tail LengthPrimary Expression Sites
Form 116 amino acidsKeratinocytes
Form 234 amino acidsCirculating and immune cells
Form 32 amino acidsUndetermined
Form 436 amino acidsBrain and peripheral neurons

Structural studies reveal conserved residues in viral CD47-like proteins, suggesting evolutionary importance of its extracellular loop region (ECLR) for ligand binding .

Immune Regulation

  • "Don’t eat me" signal: CD47 binding to SIRPα on macrophages inhibits phagocytosis, enabling cancer cells to evade immune surveillance .

  • T cell modulation:

    • Promotes memory T cell survival by blocking macrophage clearance .

    • Suppresses Th1 differentiation by reducing IL-12 responsiveness .

Cellular Processes

  • Apoptosis: CD47 ligation regulates intrinsic apoptosis pathways in leukemia cells .

  • Metabolism: Modulates glycolysis, mitochondrial function, and nucleotide synthesis .

  • Migration: Integrin binding facilitates adhesion and tissue infiltration .

Overexpression and Prognosis

CD47 is overexpressed in >90% of solid tumors and hematologic malignancies . High CD47 mRNA levels correlate with poor survival in glioblastoma (HR = 2.1, 95% CI: 1.3–3.4) and ovarian cancer (HR = 1.8, 95% CI: 1.1–2.9) .

Table 2: CD47 in Human Cancers

Cancer TypeCD47 FunctionTherapeutic Target Potential
Acute Myeloid LeukemiaBlocks phagocytosis of leukemic stem cellsAnti-CD47 antibodies (e.g., magrolimab)
Lung CancerProtects cancer stem cells (CSCs)Anti-CD47 reduces tumor growth by 60% in murine models
GlioblastomaOverexpression linked to therapy resistanceCombinatorial anti-CD47/radiation trials underway

Mechanism of Immune Evasion

Tumor cells co-express CD47 and pro-phagocytic signals (e.g., calreticulin). CD47 dominance via SIRPα engagement overrides "eat me" signals, but antibody blockade reverses this balance, enabling macrophage-mediated clearance .

Preclinical Advances

  • Anti-CD47 antibodies: B6H12 and Hu5F9-G4 enhance phagocytosis of lung cancer cells by 3–5 fold in vitro and reduce CSC-driven tumorigenesis in vivo .

  • Combination therapies: Synergy with rituximab (anti-CD20) improves lymphoma regression rates by 40% compared to monotherapy .

Clinical Challenges

  • Toxicity: Transient anemia and neutropenia observed in murine models .

  • Resistance mechanisms: Tumor cell adaptation via upregulation of alternative immune checkpoints (e.g., PD-L1) .

Emerging Research Frontiers

  • Metabolic reprogramming: CD47 knockdown reduces glycolysis by 30% in breast cancer cells, suggesting metabolic dependency .

  • Viral CD47 homologs: Phylogenetic analysis reveals conserved functional motifs, offering insights into immune evasion strategies .

Product Specs

Introduction
CD47, also known as integrin-associated protein (IAP), is a transmembrane receptor that interacts with several ligands, including thrombospondin-1 (THBS1) and signal regulatory protein alpha (SIRPα). CD47 plays diverse roles in cell adhesion, immune regulation, and neuronal function. As an adhesion receptor for THBS1 on platelets, CD47 participates in cell-cell interactions and platelet aggregation. It also modulates integrin activity, influencing cell migration and adhesion to the extracellular matrix. In the immune system, CD47 acts as a 'don't eat me' signal by binding to SIRPα on macrophages, thereby inhibiting phagocytosis and promoting self-tolerance. This interaction is crucial for preventing the premature destruction of red blood cells and maintaining immune homeostasis. Dysregulation of CD47-SIRPα signaling has been implicated in cancer immune evasion, making it a potential target for immunotherapy. Furthermore, CD47 contributes to memory formation and synaptic plasticity in the hippocampus, suggesting a role in cognitive function.
Description
Recombinant CD47 protein was expressed in Sf9 insect cells using a baculovirus expression system. The protein encompasses amino acids 19-141 of the human CD47 sequence, with a 6-amino acid Histidine tag added at the C-terminus for purification purposes. The resulting protein has a total of 129 amino acids and a molecular weight of 14.7 kDa. SDS-PAGE analysis under reducing conditions reveals multiple bands between 18-28 kDa, indicative of glycosylation. The protein was purified using proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Clear, colorless liquid, free from particulate matter
Formulation
The CD47 protein is supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) with a pH of 7.4 and 10% glycerol as a cryoprotectant.
Stability
For short-term storage (up to 4 weeks), the CD47 protein solution should be stored at 4°C. For long-term storage, it is recommended to freeze the protein at -20°C. Adding a carrier protein like bovine serum albumin (BSA) or human serum albumin (HSA) to a final concentration of 0.1% can help to stabilize the protein during freezing and thawing. Repeated freeze-thaw cycles should be avoided to prevent protein degradation.
Purity
The purity of the CD47 protein is greater than 95% as assessed by SDS-PAGE analysis.
Synonyms
CD47 Molecule, Antigenic Surface Determinant Protein OA3, CD47 Antigen (Rh-Related Antigen, Integrin-Associated Signal Transducer), Antigen Identified By Monoclonal Antibody 1D8, Integrin Associated Protein, Integrin-Associated Protein, Rh-Related Antigen, CD47 Glycoprotein, MER6, IAP, Integrin-Associated Signal Transducer, Leukocyte Surface Antigen CD47, CD47 Antigen, Protein MER6, OA3, CD47.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
QLLFNKTKSV EFTFCNDTVV IPCFVTNMEA QNTTEVYVKW KFKGRDIYTF DGALNKSTVP TDFSSAKIEV SQLLKGDASL KMDKSDAVSH TGNYTCEVTE LTREGETIIE LKYRVVSWFS PNEHHHHHH.

Q&A

What is the molecular structure and basic function of human CD47?

CD47, also known as integrin-associated protein (IAP), is a ubiquitous 50 kDa multipass transmembrane protein with a single V-type Ig-like domain at its N-terminus. It belongs to the immunoglobulin superfamily and is encoded by the CD47 gene in humans. Structurally, CD47 contains five transmembrane domains with an extracellular N-terminal domain and a short C-terminal cytoplasmic tail. The protein functions primarily as a cellular marker that inhibits phagocytosis by binding to signal-regulatory protein alpha (SIRPα) on macrophages, essentially serving as a "don't eat me" signal that prevents the engulfment of viable cells .
Alternative splicing generates several isoforms with truncated cytoplasmic domains, allowing for tissue-specific functions. Within the N-terminal extracellular domain, human CD47 shares approximately 63% amino acid sequence identity with mouse and rat CD47, highlighting evolutionary conservation of this important immune regulatory protein .

What are the primary binding partners of CD47 and their functional significance?

CD47 interacts with multiple binding partners that mediate distinct cellular functions:

Binding PartnerPrimary FunctionCellular Implications
SIRPαInhibits phagocytosisCell survival; cancer immune evasion
Thrombospondin-1 (TSP-1)Modulates T cell responsesReduced T cell proliferation and inflammatory reactions
VEGFR2Induces VEGFR2 signalingAffects angiogenesis and T cell activation
IntegrinsCell adhesionInfluences cell migration and tissue architecture
The most well-characterized interaction is between CD47 and SIRPα, which triggers inhibitory signaling cascades in phagocytes. When CD47 binds to SIRPα on macrophages, it induces tyrosine phosphorylation of immunoreceptor tyrosine-based inhibition motifs (ITIMs) in the SIRPα cytoplasmic region, subsequently recruiting and activating SHP-1 and SHP-2 phosphatases. This signaling cascade inhibits myosin-IIA accumulation at the phagocytic synapse, preventing cytoskeletal rearrangements necessary for engulfment .
The thrombospondin interaction with CD47 on T cells has significant immunomodulatory effects, including reduced T cell proliferation and dampening of inflammatory reactions. This provides an additional layer of immune regulation beyond the canonical "don't eat me" signal function .

How does CD47 contribute to cancer progression and immune evasion?

CD47 is expressed on the surface of all human solid tumor cells, serving as a critical immune evasion mechanism. Cancer cells upregulate CD47 to prevent phagocytosis by macrophages, allowing tumors to evade immune surveillance and clearance. This mechanism operates through the CD47-SIRPα axis, where CD47 on cancer cells engages SIRPα on phagocytic cells to transmit inhibitory signals that prevent engulfment .
The ubiquitous expression of CD47 across diverse cancer types indicates its fundamental role in cancer cell survival. Research has demonstrated that CD47 upregulation correlates with poorer clinical outcomes in various malignancies. Interestingly, cancer cells often simultaneously upregulate both CD47 (the "don't eat me" signal) and calreticulin (an "eat me" signal), but the inhibitory CD47 signal dominates in the absence of therapeutic intervention .
Methodologically, researchers typically assess CD47 expression in cancer tissues using immunohistochemistry, flow cytometry, or Western blotting. Flow cytometric analysis is particularly valuable for quantifying CD47 surface expression levels, with increased mean fluorescence intensity (MFI) serving as a key measurement parameter when comparing malignant versus normal tissues .

What is the role of CD47 in neurodevelopmental disorders associated with macrocephaly?

Recent research has revealed an unexpected role for CD47 in neurodevelopmental disorders, particularly in 16p11.2 deletion syndrome that presents with macrocephaly. Studies on patient-derived induced pluripotent stem cells (iPSCs) show that CD47 is significantly overexpressed in neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs) from individuals with 16p11.2 deletion syndrome who exhibit brain enlargement .
The overexpression of CD47 in these neural cells disrupts normal phagocytic clearance mechanisms, contributing to brain overgrowth. Specifically, 16p11.2 deletion NPCs and OPCs show:

  • Significantly upregulated CD47 expression at both mRNA and protein levels

  • Increased cell surface expression of calreticulin (a prophagocytic "eat me" signal)

  • Reduced rates of phagocytosis by macrophages and microglia

  • Normalization of phagocytosis rates when treated with CD47-blocking antibodies
    These findings establish a novel connection between CD47 and neurodevelopmental disorders, suggesting that dysregulation of cellular clearance mechanisms may contribute to the pathophysiology of conditions associated with brain overgrowth. Importantly, CD47 expression and related phagocytosis rates were similar across control and 16p11.2 deletion groups at the pluripotent stage before differentiation, highlighting the developmental specificity of this mechanism .

What are the optimal techniques for detecting and quantifying CD47 expression in different tissue types?

Multiple complementary techniques can be employed to detect and quantify CD47 expression, each with specific advantages depending on the research question:

TechniquePrimary ApplicationAdvantagesLimitations
Flow CytometryQuantifying surface expressionSingle-cell resolution; quantitativeRequires fresh/viable cells
Western BlotTotal protein expressionDetects specific isoforms; semi-quantitativeLow spatial resolution
ImmunohistochemistryTissue localizationPreserves tissue architecture; clinical samplesSemi-quantitative
qPCRmRNA expressionHigh sensitivity; isoform specificDoesn't reflect protein levels
ImmunofluorescenceSubcellular localizationCo-localization studiesRequires optimization for specificity
For flow cytometry, human CD47 is typically detected using specific antibodies such as anti-CD47 APC-conjugated monoclonal antibodies (e.g., clone 472603), with isotype controls used to establish baseline fluorescence. When analyzing CD47 expression in human blood lymphocytes, it's important to use proper gating strategies to identify specific cell populations of interest .
Western blot analysis for CD47 often reveals bands at approximately 45-70 kDa, representing different glycosylation states and isoforms. To minimize protein aggregation, samples are sometimes not heated prior to gel loading. For immunohistochemical detection, paraffin-embedded tissue sections are commonly used with appropriate antigen retrieval methods and specific anti-CD47 antibodies .

How can researchers effectively block CD47 function in experimental models?

Blocking CD47 function is a critical approach in both basic research and therapeutic development. Several methodological approaches have proven effective:

  • Blocking antibodies: Anti-CD47 antibodies such as clone B6.H12 can effectively block the CD47-SIRPα interaction. When using blocking antibodies, it's essential to:

    • Titrate antibody concentrations (typically 5-20 μg/mL) to determine optimal blocking without non-specific effects

    • Include appropriate isotype controls

    • Confirm blocking efficacy using functional assays such as phagocytosis assays

  • Genetic approaches:

    • CRISPR/Cas9-mediated knockdown or knockout

    • shRNA or siRNA targeting CD47 mRNA

    • Dominant-negative CD47 constructs

  • Small molecule inhibitors:

    • Peptide mimetics of SIRPα that compete for CD47 binding

    • Small molecules that disrupt the CD47-SIRPα interface
      In phagocytosis assays, researchers typically co-culture target cells (e.g., NPCs, cancer cells) with macrophages or microglia at defined ratios (often 1:1 or 2:1) and quantify engulfment through fluorescent labeling and microscopy or flow cytometry. Treatment with CD47-blocking antibodies has been shown to restore phagocytosis of 16p11.2 deletion NPCs and OPCs to control levels, particularly in cells with increased surface expression of calreticulin .

What is the relationship between CD47 expression and cellular senescence?

Recent research has uncovered a significant relationship between CD47 expression and cellular senescence. Quantitative PCR analyses demonstrate that CD47 mRNA levels increase during replicative senescence and aging. This upregulation appears to be part of the senescence-associated secretory phenotype (SASP) and may contribute to age-related tissue dysfunction .
The increased expression of CD47 in senescent cells has several potential implications:

  • It may protect senescent cells from immune clearance, contributing to their accumulation in aging tissues

  • It could affect tissue homeostasis by altering the phagocytic removal of damaged cells

  • It might represent a potential therapeutic target for senolytic approaches
    Methodologically, researchers studying this relationship typically induce senescence through various means (replicative exhaustion, oncogene activation, irradiation, or chemical induction), followed by assessment of CD47 expression via qPCR, Western blotting, and flow cytometry. The relationship between senescence markers (p16, p21, SA-β-gal) and CD47 expression provides valuable insights into the role of this protein in aging processes .

How does the CD47-SIRPα axis interact with other phagocytic signals in determining cell fate?

The regulation of phagocytosis involves a complex interplay between pro-phagocytic "eat me" signals and anti-phagocytic "don't eat me" signals, with CD47-SIRPα representing a dominant inhibitory pathway. This complex system operates as follows:

  • Calreticulin (CRT): A major pro-phagocytic signal that translocates to the cell surface in response to cellular stress or damage. CRT binds to LDL-receptor-related protein (LRP) on phagocytes to promote engulfment.

  • Phosphatidylserine (PS): Exposed on the outer leaflet of the plasma membrane during apoptosis, PS is recognized by various receptors on phagocytes, including TIM-4, BAI1, and stabilin-2.

  • CD47-SIRPα interaction: Provides a dominant inhibitory signal that can override pro-phagocytic signals.
    Research in 16p11.2 deletion syndrome has revealed important insights into this balance. 16p11.2 deletion NPCs and OPCs simultaneously upregulate both CD47 and cell surface calreticulin, but the high levels of CD47 override the pro-phagocytic CRT signal. Importantly, treatment with CD47-blocking antibodies restored phagocytosis, particularly in cells with high CRT expression, demonstrating the dominance of the CD47 signal in this context .
    Methodologically, researchers investigating this axis often use flow cytometry to simultaneously measure surface expression of CD47, calreticulin, and other markers such as asialoglycan binding sites (detected with PHA-L). The balance between these signals determines cellular fate, with therapeutic manipulation of this balance showing promise for treating conditions ranging from cancer to neurodevelopmental disorders .

What are the current approaches for targeting CD47 in cancer immunotherapy?

The recognition of CD47 as a nearly universal tumor immune evasion mechanism has spurred significant therapeutic development. Current approaches include:

Therapeutic ApproachMechanismDevelopment StatusConsiderations
Anti-CD47 antibodiesBlock CD47-SIRPα interactionMultiple clinical trialsOn-target anemia; first-dose effects
SIRPα-Fc fusion proteinsCompetitive inhibition of CD47Early clinical developmentPotentially reduced red blood cell binding
Bispecific antibodiesTarget CD47 and tumor-specific antigensPreclinical/early clinicalImproved targeting specificity
CD47-ADCsTargeted cytotoxic deliveryPreclinicalCombines blocking and cell killing
Small molecule inhibitorsDisrupt CD47-SIRPα interfaceDiscovery phasePotential oral bioavailability
Methodologically, evaluation of anti-CD47 therapeutics typically involves in vitro phagocytosis assays, where target cancer cells are co-cultured with macrophages in the presence of therapeutic candidates. Efficacy is measured by quantifying the percentage of cancer cells engulfed. In vivo efficacy is assessed using xenograft or syngeneic tumor models, with tumor volume reduction and survival as primary endpoints .
One challenge in CD47-targeted therapy is the ubiquitous expression of CD47 on normal cells, particularly red blood cells, which can lead to anemia and antigen sink effects. Strategies to overcome this include tumor-directed bispecific antibodies and careful dosing regimens .

What potential exists for targeting CD47 in neurodevelopmental disorders?

The discovery of CD47 overexpression in 16p11.2 deletion syndrome with macrocephaly opens new therapeutic possibilities for neurodevelopmental disorders. Research has demonstrated that blocking CD47 with antibodies restores normal phagocytic clearance of neural progenitor cells and oligodendrocyte progenitor cells derived from individuals with 16p11.2 deletion syndrome .
This suggests several potential therapeutic approaches:

  • Targeted CD47 blockade: Specific delivery of CD47-blocking agents to the central nervous system during critical developmental windows

  • Modulation of downstream pathways: Targeting components of the CD47-SIRPα signaling cascade to normalize phagocytic function

  • Combinatorial approaches: Simultaneously enhancing pro-phagocytic signals while blocking CD47
    Significant research challenges remain, including the delivery of therapeutics across the blood-brain barrier, identifying optimal developmental windows for intervention, and ensuring specificity to abnormal neural cells. Future research directions should include comprehensive investigation of CD47 expression patterns across various neurodevelopmental disorders associated with brain overgrowth, development of CNS-penetrant CD47-targeting agents, and detailed characterization of the effects of CD47 modulation on neural circuit development .

How should researchers optimize flow cytometry protocols for accurate CD47 quantification?

Flow cytometric analysis of CD47 requires careful optimization to ensure accurate quantification, particularly given its ubiquitous expression and the importance of detecting subtle expression differences in pathological states:

  • Sample preparation:

    • Fresh isolation is preferred when possible

    • Careful enzymatic dissociation to preserve surface epitopes

    • Appropriate fixation (if needed) using paraformaldehyde at concentrations that maintain epitope integrity

  • Antibody selection and titration:

    • Use of validated anti-CD47 antibodies (e.g., clone 472603 for human CD47)

    • Proper titration to determine optimal antibody concentration

    • Inclusion of appropriate isotype controls to establish baseline fluorescence

  • Panel design:

    • Include markers for cell identification and viability

    • Consider simultaneous detection of related markers (SIRPα, calreticulin)

    • Careful compensation when using multiple fluorophores

  • Analysis considerations:

    • Report CD47 expression as mean fluorescence intensity (MFI)

    • Use consistent gating strategies between experiments

    • Consider the percentage of CD47+ cells and the intensity distribution
      Human peripheral blood lymphocytes can serve as positive controls for CD47 expression, as demonstrated in multiple studies using techniques such as flow cytometry with APC-conjugated monoclonal antibodies against CD47 .

What are the key considerations for designing valid phagocytosis assays to study CD47 function?

Phagocytosis assays are central to functional studies of CD47. Critical methodological considerations include:

  • Target cell preparation:

    • Fluorescent labeling (e.g., CFSE, CellTracker dyes) for tracking

    • Assessment of baseline CD47 and calreticulin expression

    • Standardization of cell numbers and viability

  • Phagocyte selection and preparation:

    • Source of phagocytes (primary macrophages, microglia, cell lines)

    • Consistent activation state (M0, M1, M2 polarization)

    • Fluorescent labeling distinct from target cells

  • Co-culture conditions:

    • Optimized target:phagocyte ratio (typically 1:1 to 5:1)

    • Appropriate incubation time (2-24 hours depending on system)

    • Culture medium selection to support both cell types

  • Analysis methods:

    • Flow cytometry (double-positive events indicating engulfment)

    • Confocal microscopy with Z-stack confirmation of internalization

    • Live cell imaging for kinetic studies

  • Controls and interventions:

    • CD47 blocking antibodies (e.g., B6.H12 clone)

    • SIRPα blocking antibodies

    • Cytochalasin D to inhibit all phagocytosis as negative control
      Phagocytosis is typically quantified as the percentage of phagocytes containing engulfed target cells. When studying 16p11.2 deletion NPCs and OPCs, researchers have demonstrated that treatment with CD47-blocking antibodies restores phagocytosis rates to control levels, particularly in cells with increased cell surface expression of calreticulin .

Product Science Overview

Structure and Expression

CD47 is a 47 kDa protein that appears at a position of 70 kDa upon SDS-PAGE due to its hyperglycosylated structure . It consists of:

  • An N-terminal extracellular IgV domain
  • Five transmembrane helices
  • A C-terminal cytoplasmic short variable spliced tail

There are four different spliced isoforms of CD47, with isoform 2 being the most abundantly expressed . CD47 is found on a variety of hematopoietic and non-hematopoietic cells, and its expression level varies depending on the cell type .

Function

CD47 is well-known for its immunoregulatory functions. It acts as a “don’t eat me” signal to prevent phagocytosis by macrophages . This receptor is crucial for maintaining immune system homeostasis by modulating cellular phagocytosis, transmigration of neutrophils, and activation of dendritic cells, T cells, and B cells .

In healthy cells, CD47 prevents phagocytosis, while aged and superfluous cells are removed through phagocytosis due to CD47 down-regulation . This regulatory function was first described in red blood cells but is now known to be widely expressed in other cell types as well .

Role in Cancer

CD47 is highly expressed in various types of cancer cells, allowing them to escape from the immune system . This makes CD47 a prominent target in cancer therapy. Blocking the interaction between CD47 and SIRPα can enhance cancer cell clearance by macrophages and increase antigen cross-presentation, leading to T-cell priming and an activated adaptive antitumor immune response .

Therapeutic Applications

Several strategies have been developed to target CD47 in cancer therapy, including:

  • Mimicry peptides
  • Antibodies
  • Gene silencing strategies

Among these, the use of anti-CD47 antibodies is the most advanced approach. These antibodies inhibit the CD47-SIRPα axis, enhancing cancer cell phagocytosis . Combining these antibodies with chemotherapy and immunotherapy has shown promise in improving the survival of cancer patients .

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.