InVivoMAb anti-mouse/human CD44

Clone IM7
Catalog # BE0039
Category InVivoMab Antibodies
Price
Size Regular Price
1 mg $ 150.00
5 mg $ 550.00
25 mg $ 1,840.00
50 mg $ 2,770.00
100 mg $ 3,920.00
About InVivoMAb anti-mouse/human CD44

The IM7 monoclonal antibody reacts with human and mouse CD44 also known as Hermes, HCAM, and Pgp-1. CD44 is an 80-95 kDa glycoprotein that is expressed on all leukocytes, endothelial cells, hepatocytes, and mesenchymal cells. As an adhesion molecule, CD44 participates in a wide variety of cellular functions including lymphocyte activation, recirculation and homing, and hematopoiesis. CD44 is a receptor for hyaluronic acid and can also interact with other ligands, such as osteopontin, collagens, and matrix metalloproteinases (MMPs). Additionally, CD44 is involved in tumor metastasis and targeting of CD44 by antibodies has been shown to reduce the malignant activities of various neoplasms. Interestingly, high levels of the adhesion molecule CD44 on leukemic cells are essential to generate leukemia. The IM7 antibody has been shown to neutralize CD44 in vivo

InVivoMAb anti-mouse/human CD44 Specifications
IsotypeRat IgG2b, κ
ImmunogenDexamethasone-induced myeloid leukemia M1 cells
Reported Applications

in vivo CD44 neutralization

Formulation
  • PBS, pH 7.0
  • Contains no stabilizers or preservatives
Endotoxin
  • <2EU/mg (<0.002EU/μg)
  • Determined by LAL gel clotting assay
Purity
  • >95%
  • Determined by SDS-PAGE
Sterility0.2 μM filtered
ProductionPurified from tissue culture supernatant in an animal free facility
PurificationProtein G
RRIDAB_1107649
Molecular Weight150 kDa
StorageThe antibody solution should be stored at the stock concentration at 4°C. Do not freeze.
Application References

INVIVOMAB ANTI-MOUSE/HUMAN CD44 (CLONE: IM7)

Guidotti, L. G., et al. (2015). “Immunosurveillance of the liver by intravascular effector CD8(+) T cells.” Cell 161(3): 486-500. PubMed

Effector CD8(+) T cells (CD8 TE) play a key role during hepatotropic viral infections. Here, we used advanced imaging in mouse models of hepatitis B virus (HBV) pathogenesis to understand the mechanisms whereby these cells home to the liver, recognize antigens, and deploy effector functions. We show that circulating CD8 TE arrest within liver sinusoids by docking onto platelets previously adhered to sinusoidal hyaluronan via CD44. After the initial arrest, CD8 TE actively crawl along liver sinusoids and probe sub-sinusoidal hepatocytes for the presence of antigens by extending cytoplasmic protrusions through endothelial fenestrae. Hepatocellular antigen recognition triggers effector functions in a diapedesis-independent manner and is inhibited by the processes of sinusoidal defenestration and capillarization that characterize liver fibrosis. These findings reveal the dynamic behavior whereby CD8 TE control hepatotropic pathogens and suggest how liver fibrosis might reduce CD8 TE immune surveillance toward infected or transformed hepatocytes.

 

Mott, P. J. and A. H. Lazarus (2013). “CD44 antibodies and immune thrombocytopenia in the amelioration of murine inflammatory arthritis.” PLoS One 8(6): e65805. PubMed

Antibodies to CD44 have been used to successfully ameliorate murine models of autoimmune disease. The most often studied disease model has been murine inflammatory arthritis, where a clear mechanism for the efficacy of CD44 antibodies has not been established. We have recently shown in a murine passive-model of the autoimmune disease immune thrombocytopenia (ITP) that some CD44 antibodies themselves can induce thrombocytopenia in mice, and the CD44 antibody causing the most severe thrombocytopenia (IM7), also is known to be highly effective in ameliorating murine models of arthritis. Recent work in the K/BxN serum-induced model of arthritis demonstrated that antibody-induced thrombocytopenia reduced arthritis, causing us to question whether CD44 antibodies might primarily ameliorate arthritis through their thrombocytopenic effect. We evaluated IM7, IRAWB14.4, 5035-41.1D, KM201, KM114, and KM81, and found that while all could induce thrombocytopenia, the degree of protection against serum-induced arthritis was not closely related to the length or severity of the thrombocytopenia. CD44 antibody treatment was also able to reverse established inflammation, while thrombocytopenia induced by an anti-platelet antibody targeting the GPIIbIIIa platelet antigen, could not mediate this effect. While CD44 antibody-induced thrombocytopenia may contribute to some of its therapeutic effect against the initiation of arthritis, for established disease there are likely other mechanisms contributing to its efficacy. Humans are not known to express CD44 on platelets, and are therefore unlikely to develop thrombocytopenia after CD44 antibody treatment. An understanding of the relationship between arthritis, thrombocytopenia, and CD44 antibody treatment remains critical for continued development of CD44 antibody therapeutics.

 

Hutas, G., et al. (2008). “CD44-specific antibody treatment and CD44 deficiency exert distinct effects on leukocyte recruitment in experimental arthritis.” Blood 112(13): 4999-5006. PubMed

CD44, the leukocyte adhesion receptor for hyaluronan, has been considered a therapeutic target on the basis of the robust anti-inflammatory effect of CD44-specific antibodies in animal models of immune-mediated diseases. However, CD44 deficiency does not provide substantial protection against inflammation. Using intravital video microscopy in a murine model of rheumatoid arthritis, we show that CD44 deficiency and anti-CD44 antibody treatment exert disparate effects on leukocyte recruitment in inflamed joints. Leukocyte rolling, which is increased in CD44-deficient mice, is promptly abrogated in anti-CD44-treated wild-type mice. CD44-specific antibodies also trigger platelet deposition on granulocytes and subsequent depletion of this leukocyte subset in the circulation. These in vivo effects require CD44 cross-linking and are reproducible with an antibody against Gr-1, a molecule that, like CD44, is highly expressed on granulocytes. Anticoagulant pretreatment, which prevents platelet deposition, mitigates both granulocyte depletion and the suppressive effect of CD44-specific antibody on joint swelling. Our observations suggest that cross-linking of prominent cell surface molecules, such as CD44 or Gr-1, can initiate a rapid self-elimination program in granulocytes through engagement of the coagulation system. We conclude that the robust anti-inflammatory effect of CD44-specific antibodies in arthritis is primarily the result of their ability to trigger granulocyte depletion.