RecombiMAb anti-mouse IL-10R (CD210) (LALA-PG)

Clone Catalog # Category
1B1.3A-CP073 CP073
USD 541 - USD 7325

About RecombiMAb anti-mouse IL-10R (CD210) (LALA-PG)

The 1B1.3A-CP073 monoclonal antibody is a recombinant, chimeric version of the original 1B1.3A antibody. The variable domain sequences are identical but the constant region sequences have been switched from Rat IgG1, κ to mouse IgG2a, κ for use in murine models. Additionally, 1B1.3A-CP073 contains LALA-PG mutations in the heavy chain Fc fragment rendering it unable to bind endogenous murine FcγR or C1q to induce antibody-dependent, cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). Antibodies with active Fc regions can engage immune cells via FcγRs, leading to the depletion of antigen expressing cells through mechanisms like ADCC or complement activation. Fc-silenced antibodies do not trigger these pathways and can block signaling without killing or depleting target cells. Using Fc-silent antibodies allows researchers to distinguish between the effects of receptor blockade and those of cell depletion or Fc-mediated immune activation. Additionally, species-matched chimeric antibodies demonstrate reduced immunogenicity and formation of anti-drug antibodies (ADAs) compared to xenogenic antibodies in animal models. The highly controlled sequence and lack of genetic drift in recombinant antibodies provide more reliable and reproducible results over hybridoma derived antibodies. 1B1.3A-CP073 monoclonal antibody reacts with mouse IL-10R (IL-10 receptor) also known as CD210. The IL-10R is a class II cytokine receptor and is expressed by a variety of cell types including thymocytes, T lymphocytes, B lymphocytes, NK cells, monocytes, and macrophages. Upon binding IL-10, IL-10R stimulation results in many pleiotropic effects in immunoregulation and inflammation. IL-10R downregulates the expression of pro-inflammatory cytokines, MHC class II antigens, and co-stimulatory molecules on macrophages. It also enhances B lymphocyte survival, proliferation, and antibody production. IL-10R signaling can block NF-κB activity and is involved in the regulation of the JAK-STAT signaling pathway. The 1B1.3A antibody is a neutralizing antibody and has been shown to block the binding of human IL-10, which cross-reacts with the mouse IL-10R. However, this clone does not recognize the human IL-10R.

RecombiMAb anti-mouse IL-10R (CD210) (LALA-PG) Specifications

IsotypeMouse IgG2a LALA-PG
ImmunogenRecombinant ligand-binding domain of mouse IL-10R
Reported ApplicationsFlow Cytometry Western blot in vitro blocking of IL-10R signaling in vivo blocking of IL-10/IL-10R signaling *Reported for the original rat IgG1 1B1.3A antibody. For information on in vivo applications, please contact technicalservice@bioxcell.com
FormulationPBS, pH 7.0 Contains no stabilizers or preservatives
Endotoxin<1EU/mg (<0.001EU/μg) Determined by LAL gel clotting assay
Aggregation<5% Determined by SEC
Purity>95% Determined by SDS-PAGE
Sterility0.2 μm filtration
ProductionPurified from HEK293 cell supernatant in an animal-free facility
PurificationProtein G
Molecular Weight150 kDa
StorageThe antibody solution should be stored at the stock concentration at 4°C. Do not freeze.

Application References

RecombiMAb anti-mouse IL-10R (CD210) (LALA-PG) (CLONE: 1B1.3A-CP073)

Mindt BC, Kim J, Warren T, Song Y, DiGiandomenico A (2023). "Differential in vivo labeling with barcoded antibodies allows for simultaneous transcriptomic profiling of airway, lung tissue and intravascular immune cells" Front Immunol . PubMed

Single-cell RNA sequencing (scRNA-seq) is the state-of-the-art approach to study transcriptomic signatures in individual cells in respiratory health and disease. However, classical scRNA-seq approaches provide no spatial information and are performed using either bronchoalveolar lavage fluid (BAL) or lung single cell suspensions to assess transcript levels in airway and tissue immune cells, respectively. Herein we describe a simple method to simultaneously characterize transcriptomic features of airway, lung parenchymal and intravascular immune cells based on differential in vivo labeling with barcoded antibodies. In addition to gaining basic spatial information, this approach allows for direct comparison of cells within different anatomical compartments. Furthermore, this method provides a time- and cost-effective alternative to classical scRNA-seq where lung and BAL samples are processed individually, reducing animal and reagent use. We demonstrate the feasibility of this approach in a preclinical mouse model of bacterial lung infection comparing airway, parenchymal and vasculature neutrophils early after infection.

Saha A, Hyzy S, Lamothe T, Hammond K, Clark N, Lanieri L, Bhattarai P, Palchaudhuri R, Gillard GO, Proctor J, Riddle MJ, Panoskaltsis-Mortari A, MacMillan ML, Wagner JE, Kiem HP, Olson LM, Blazar BR (2022). "A CD45-targeted antibody-drug conjugate successfully conditions for allogeneic hematopoietic stem cell transplantation in mice" Blood 139(11):1743-1759. PubMed

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment of patients with nonmalignant or malignant blood disorders. Its success has been limited by graft-versus-host disease (GVHD). Current systemic nontargeted conditioning regimens mediate tissue injury and potentially incite and amplify GVHD, limiting the use of this potentially curative treatment beyond malignant disorders. Minimizing systemic nontargeted conditioning while achieving alloengraftment without global immune suppression is highly desirable. Antibody-drug-conjugates (ADCs) targeting hematopoietic cells can specifically deplete host stem and immune cells and enable alloengraftment. We report an anti-mouse CD45-targeted-ADC (CD45-ADC) that facilitates stable murine multilineage donor cell engraftment. Conditioning with CD45-ADC (3 mg/kg) was effective as a single agent in both congenic and minor-mismatch transplant models resulting in full donor chimerism comparable to lethal total body irradiation (TBI). In an MHC-disparate allo-HSCT model, pretransplant CD45-ADC (3 mg/kg) combined with low-dose TBI (150 cGy) and a short course of costimulatory blockade with anti-CD40 ligand antibody enabled 89% of recipients to achieve stable alloengraftment (mean value: 72%). When CD45-ADC was combined with pretransplant TBI (50 cGy) and posttransplant rapamycin, cyclophosphamide (Cytoxan), or a JAK inhibitor, 90% to 100% of recipients achieved stable chimerism (mean: 77%, 59%, 78%, respectively). At a higher dose (5 mg/kg), CD45-ADC as a single agent was sufficient for rapid, high-level multilineage chimerism sustained through the 22 weeks observation period. Therefore, CD45-ADC has the potential utility to confer the benefit of fully myeloablative conditioning but with substantially reduced toxicity when given as a single agent or at lower doses in conjunction with reduced-intensity conditioning.

Chen PM, Wilson PC, Shyer JA, Veselits M, Steach HR, Cui C, Moeckel G, Clark MR, Craft J (2020). "Kidney tissue hypoxia dictates T cell-mediated injury in murine lupus nephritis" Sci Transl Med 12(538):eaay1620. PubMed

The kidney is a frequent target of autoimmune injury, including in systemic lupus erythematosus; however, how immune cells adapt to kidney's unique environment and contribute to tissue damage is unknown. We found that renal tissue, which normally has low oxygen tension, becomes more hypoxic in lupus nephritis. In the injured mouse tissue, renal-infiltrating CD4+ and CD8+ T cells express hypoxia-inducible factor-1 (HIF-1), which alters their cellular metabolism and prevents their apoptosis in hypoxia. HIF-1-dependent gene-regulated pathways were also up-regulated in renal-infiltrating T cells in human lupus nephritis. Perturbation of these environmental adaptations by selective HIF-1 blockade inhibited infiltrating T cells and reversed tissue hypoxia and injury in murine models of lupus. The results suggest that targeting HIF-1 might be effective for treating renal injury in autoimmune diseases.

Palchaudhuri R, Saez B, Hoggatt J, Schajnovitz A, Sykes DB, Tate TA, Czechowicz A, Kfoury Y, Ruchika F, Rossi DJ, Verdine GL, Mansour MK, Scadden DT (2016). "Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin" Nat Biotechnol 34(7):738-45. PubMed

Hematopoietic stem cell transplantation (HSCT) offers curative therapy for patients with hemoglobinopathies, congenital immunodeficiencies, and other conditions, possibly including AIDS. Autologous HSCT using genetically corrected cells would avoid the risk of graft-versus-host disease (GVHD), but the genotoxicity of conditioning remains a substantial barrier to the development of this approach. Here we report an internalizing immunotoxin targeting the hematopoietic-cell-restricted CD45 receptor that effectively conditions immunocompetent mice. A single dose of the immunotoxin, CD45-saporin (SAP), enabled efficient (>90%) engraftment of donor cells and full correction of a sickle-cell anemia model. In contrast to irradiation, CD45-SAP completely avoided neutropenia and anemia, spared bone marrow and thymic niches, enabling rapid recovery of T and B cells, preserved anti-fungal immunity, and had minimal overall toxicity. This non-genotoxic conditioning method may provide an attractive alternative to current conditioning regimens for HSCT in the treatment of non-malignant blood diseases.

Wulf GG, Luo KL, Goodell MA, Brenner MK (2003). "Anti-CD45-mediated cytoreduction to facilitate allogeneic stem cell transplantation" Blood 101(6):2434-9. PubMed

The CD45 antigen is present on all cells of the hematopoietic lineage. Using a murine model, we have determined whether a lytic CD45 monoclonal antibody can produce persistent aplasia and whether it could facilitate syngeneic or allogeneic stem cell engraftment. After its systemic administration, we found saturating quantities of the antibody on all cells expressing the CD45 antigen, both in marrow and in lymphoid organs. All leukocyte subsets in peripheral blood were markedly diminished during or soon after anti-CD45 treatment, but only the effect on the lymphoid compartment was sustained. In contrast to the prolonged depletion of T and B lymphocytes from the thymus and spleen, peripheral blood neutrophils began to recover within 24 hours after the first anti-CD45 injection and marrow progenitor cells were spared from destruction, despite being coated with saturating quantities of anti-CD45. Given the transient effects of the monoclonal antibody on myelopoiesis and the more persistent effects on lymphopoiesis, we asked whether this agent could contribute to donor hematopoietic engraftment following nonmyeloablative transplantation. Treatment with anti-CD45 alone did not enhance syngeneic engraftment, consistent with its inability to destroy progenitor cells and permit competitive repopulation with syngeneic donor stem cells. By contrast, the combination of anti-CD45 and an otherwise inactive dose of total-body irradiation allowed engraftment of H2 fully allogeneic donor stem cells. We attribute this result to the recipient immunosuppression produced by depletion of CD45(+) lymphocytes. Monoclonal antibodies of this type may therefore have an adjunctive role in nonmyeloablative conditioning regimens for allogeneic stem cell transplantation.