Anti-Human CD11a – Purified in vivo GOLD™ Functional Grade

Anti-Human CD11a – Purified in vivo GOLD™ Functional Grade

Product No.: C373

[product_table name="All Top" skus="C373"]

- -
- -
Clone
38
Target
CD11a
Formats AvailableView All
Product Type
Monoclonal Antibody
Alternate Names
LFA-1α chain, ITGAL
Isotype
Mouse IgG2a
Applications
Costim
,
FC
,
in vivo
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Mouse
Recommended Isotype Controls
Recommended Dilution Buffer
Immunogen
Fibronectin purified monocytes.
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% monomer by analytical SEC
>95% by SDS Page
Formulation
This monoclonal antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
Product Preparation
Functional grade preclinical antibodies are manufactured in an animal free facility using in vitro cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Country of Origin
USA
Shipping
Next Day 2-8°C
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for this 38 antibody for staining cells in flow cytometry is ≤ 1 μg per 106 cells in a volume of 100 μl or 100μl of whole blood. Titration of the reagent is recommended for optimal performance for each application.
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
Clone 38 recognizes an epitope on human CD11a.
Background
LFA-1α (CD11a) and CD18 are the Integrin alpha-L and beta-2 chains respectively that combine to form LFA-1, a glycoprotein and a member of the Integrin family. Integrin alpha-L/beta-2 is a receptor for ICAM1, ICAM2, ICAM3, ICAM4 and for F11R. LFA-1 participates in the immunological synapses between CD8+ T lymphocytes and antigen-presenting cells. The absence of LFA-1α or ß may induce LAD. The antigen contributes to natural killer cell cytotoxicity, and is involved in various immune phenomena such as leukocyte-endothelial cell interaction, cytotoxic T-cell mediated killing, and antibody dependent killing by granulocytes and monocytes. The CD11b/CD18 antigen is a heterodimeric surface glycoprotein on leukocytes and belongs to the ß2 integrin family. CD11b functions as a receptor for C3bi complement, clotting factor X, fibrinogen and ICAM-1. CD11c forms an α/ß heterodimeric glycoprotein (CD11c/CD18 complex) which belongs to the ß2 integrin family. The complex binds fibrinogen and reportedly serves as a receptor for iC3b and ICAM-1. During inflammatory responses, it mediates cell to cell interaction and is important in both monocyte adhesion and chemotaxis.
Antigen Distribution
CD11a is present on thymocytes, blood lymphocytes, bone marrow cells and certain lymphoma and macrophage-like cell lines.
Ligand/Receptor
ICAM-1(CD54), ICAM-2(CD102), ICAM-3(CD50)
PubMed
NCBI Gene Bank ID
Research Area
Cell Adhesion
.
Cell Biology
.
Costimulatory Molecules
.
Immunology
.
Innate Immunity
.
Neuroinflammation
.
Neuroscience

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Clone 38 does not have a universally recognized definition in mouse in vivo research; the query likely refers either to the MC38 murine colorectal cancer cell line or an anti-CD38 antibody used in mice. The most common in vivo applications of the MC38 clone in mice are as follows:

  • Syngeneic Tumor Model: The MC38 cell line is widely implanted into immunocompetent C57BL/6 mice to create subcutaneous or orthotopic tumors for the study of anti-tumor immunity, cancer progression, and evaluation of novel immunotherapies.
  • Typically, MC38 cells are injected subcutaneously into the flank of C57BL/6 mice, allowing for easy tumor monitoring and reproducibility.
  • MC38 tumors serve to model advanced human colorectal carcinomas that have escaped immune selection, and the immune microenvironments of these tumors are studied using histology, immunohistochemistry, and flow cytometry.
  • Variants such as MC38 engineered to express human antigens (e.g., PSMA, EPCAM) or loss/gain of function mutations are also implanted for efficacy studies and immunotherapy research.

If referring to anti-CD38 clone antibodies (such as NIMR5 or OKT10), common in vivo applications include:

  • Modulation of Immune Cells: Anti-mouse CD38 (e.g., clone NIMR5) is used to stimulate or deplete CD38-expressing cells (such as B cells, NK cells, some T cells) in vivo, contributing to immune modulation studies.
  • Cancer Therapy Research: Human CD38 antibodies (e.g., OKT10) have been used as targeting vehicles for radioimmunotherapy and other cancer treatments in mouse xenograft models.

If clarification is needed for the exact “clone 38” (e.g., for an antibody or other clone), please provide additional details or context, as the term is ambiguous and most commonly encountered in reference to the MC38 tumor cell line in murine immuno-oncology.

Commonly used antibodies or proteins paired with CD38 antibodies in the literature—especially for immunophenotyping, clinical diagnostics, and research in hematology—include markers used to identify plasma cells and distinguish them from other immune populations. The most frequently reported are:

  • CD138: Often combined with CD38 to enumerate plasma cells in both diagnostic and minimal residual disease (MRD) panels, since both are highly expressed on plasma cells.
  • CD19 and CD27: Used alongside CD38 to help separate plasma cells (which are CD38⁺, CD138⁺, CD19⁻, and often CD27⁺) from B cells and other lymphocytes.
  • CD229, CD269 (BCMA), and CD319 (SLAMF7): These emerging markers can either substitute for or complement CD38 in flow cytometry, particularly when anti-CD38 therapies may interfere with CD38 detection.
  • Other hematological markers: In red blood cell compatibility testing following daratumumab or other anti-CD38 therapies, extensive phenotyping involves antigen groups such as ABO, RhD, Kell, Kidd, Duffy, and MNS, but these pertain more to transfusion compatibility rather than direct co-staining or protein interaction with CD38.

In summary, the most common antibodies or proteins used in panels together with CD38 are:

  • CD138
  • CD19
  • CD27
  • CD229 (Ly9)
  • CD269 (BCMA)
  • CD319 (SLAMF7)

Their selection depends on the immunophenotyping purpose, disease context (such as multiple myeloma), and potential interference from therapeutic anti-CD38 antibodies.

The key findings related to "clone 38" citations in scientific literature are unclear, as no search result directly addresses a clone specifically named "clone 38." If you are referring to a specific antibody clone, genetic clone, or a highly cited scientific publication associated with "clone 38," such information does not explicitly appear in the provided results.

However, here are the important contextual points relevant to the broader topic of scientific citation, clone detection, and reference accuracy:

  • Citation Accuracy in Science: A study found that the most common form of inaccurate citation in frequently cited biomedical papers was the citation of nonexistent findings (38.4%), followed by inaccurately cited numerical data/results (16.6%). This suggests a substantial issue with misattribution or errors in frequently cited science.
  • Clone Detection and Evolution in Computer Science: Systematic reviews summarize the evolution and detection of code clones (duplicated software code segments), with 47 distinct works found over past decades, but do not mention a "clone 38".
  • Cloning in Biology and Medicine: Literature on biological cloning discusses breakthroughs and challenges in the field, including the creation of embryonic stem cell lines and related ethical, legal, and technical considerations, but no result references a "clone 38".
  • Citation Metrics: In general, the evaluation of scientific impact via citation counts is nuanced, and citation manipulation or inflation is a concern within the scientific community.

If your query is about a specific scientific reagent, antibody, or genetic construct labeled as "clone 38," more context or a different set of search results may be needed to precisely identify its key findings or impact in literature. If you intended a particular publication or technical clone, please provide further details.

Dosing regimens of clone 38 in mouse models can vary significantly depending on the type of antibody, mouse strain, disease model, and experimental goal. However, your query lacks specificity as to which "clone 38" is referenced (e.g., an anti-PD-1, anti-CD38, or another antibody), which is crucial since multiple monoclonal antibodies could be assigned "38" as their clone identifier in the literature. Assuming you are asking about a well-established antibody such as anti-CD38 or other common research antibodies used in mice, here is how dosing regimens can differ across models:

  • Dose amount per mouse: Common in vivo antibody doses for mice usually range from 100–500 μg per mouse, typically delivered via intraperitoneal (IP) injection.
  • Dosing frequency: Doses are often given every 3–4 days or 2–3 times per week in chronic treatment studies.
  • Experimental model sensitivity: Doses and schedules can be adjusted according to the tumor type (e.g., MC38, CT26, B16) or immune depletion goal (T-cell, neutrophil, etc.), which may be necessary to achieve therapeutic or depletion efficacy.
  • Pharmacokinetic and efficacy matching: For mimicking humanized exposure, mouse dosing is sometimes adjusted to match human area under the curve (AUC) or peak concentration (Cmax), and this can lead to regimens such as 293 mg/kg every 6 hours for certain drugs in leukopenic models to mirror clinical pharmacokinetics.
  • Route of administration: While IP injection is most common, other routes—such as intravenous, subcutaneous, or intratumoral—can be used, based on model and experimental design.

Example Table: Typical Antibody Dosing Regimens (Syngeneic Mouse Tumor Models)

Antibody CloneTargetStandard DoseRouteScheduleComments
RMP1-14PD-1200–500 μg/mouseIntraperitoneal (IP)Every 3–4 daysUsed in MC38, B16 models
29F.1A12PD-1100–200 μg/mouseIPEvery 3 days ×3 dosesSyngeneic tumor models
OKT10CD3850–250 μg/mouse*IP or IV2–3 times per week*Varied by experiment
9D9CTLA-4100–250 μg/mouseIPEvery 3 daysDepletes Tregs in tumors

Critical Model Variation Factors

  • Strain and immune status: Immunodeficient vs. immunocompetent mice often require dose or schedule modifications.
  • Disease context: Acute vs. chronic, autoimmunity vs. tumor models.
  • Pharmacodynamic endpoints: When matching human PK/PD, higher-frequency dosing may be applied, especially in xenograft or immunodeficient mouse models.

If you are asking about anti-human CD38 (clone OKT10), for instance, the dosing regimens are experiment-dependent, with dose levels and schedules optimized for the antibody's half-life, the disease model (e.g., humanized mice, tumor models), and therapeutic readout.

There is no universally fixed regimen for "clone 38"; it is tailored to the antibody in question, the mouse model, experimental aims, and desired immunological effect. If you specify which clone 38 and application, a more precise regimen can be outlined.

References & Citations

1. Stern, LJ. et al. (2005) Proc Natl Acad Sci U S A.102(10):3744-9 PubMed
2. Taher, A. et al. (2008) Haematologica. 93(6):941-2. Article Link
3. Fliedner, TM et al. (1996) Cytometry.25(1):46-57. Article Link
4. Dransfield, I. et al. (1989) The EMBO Journal 8:3759
Costim
Flow Cytometry
in vivo Protocol
General Western Blot Protocol

Certificate of Analysis

- -
- -

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.