Mouse IgM Isotype Control — Purified in vivo GOLD™ Functional Grade

Mouse IgM Isotype Control — Purified in vivo GOLD™ Functional Grade

Product No.: I-1194

[product_table name="All Top" skus="I-1194"]

- -
- -
Product Type
Isotype Control
Isotype
Mouse IgM
Applications
in vivo

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Host Species
Mouse
Recommended Dilution Buffer
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
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Specificity
This Mouse IgM isotype control antibody has been tested against selected species' cells and tissues to assure minimal cross reactivity. Anti-0 0 recognizes 0 0. This

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.

Common In Vivo Applications of Cloned Mice

Cloned mice are widely used in various in vivo applications, particularly in genetic research and biomedical studies. Here are some common applications:

  1. Genome Editing and Genetic Studies: Cloned mice are used to study genetic mutations and their effects on organisms. They can be engineered to express specific genes or to model human diseases by introducing human or other species' genes into their genome.

  2. Cancer Research: Cloned mice can be used to study cancer progression and response to treatments. They are often engineered to carry specific genetic mutations related to cancer, allowing researchers to study cancer development in a controlled manner.

  3. Disease Modeling: Cloned mice are used to model human diseases such as diabetes, Alzheimer's disease, and more. This helps researchers understand disease mechanisms and test potential therapeutic strategies.

  4. Nanoparticle Delivery Studies: Mice, including cloned ones with humanized or primatized livers, are used to study the delivery and efficacy of nanoparticles, such as lipid nanoparticles (LNPs), in delivering mRNA or other therapeutic agents.

  5. Immune System Studies: Cloned mice with specific immune system modifications are used to study immune responses and develop new immunotherapies.

These applications highlight the versatility and utility of cloned mice in advancing biomedical research and understanding complex biological processes in vivo.

Nanoparticles are frequently conjugated with various antibodies and proteins across multiple biomedical applications. The literature reveals several categories of these biomolecules used in combination with nanoparticles for diagnostic, therapeutic, and imaging purposes.

Antibody Fragments and Engineered Variants

Nanobodies (Nbs) represent one of the most versatile protein fragments used with nanoparticles. These antibody fragments are derived from heavy-chain-only IgG antibodies found in the Camelidae family and cartilaginous fish. Nanobodies offer several advantages when conjugated to nanoparticles, including stability, diverse binding capabilities, absence of cross-reactive Fc regions, easy multivalency, and compatibility with gold nanoparticles. Specific examples include nanobodies conjugated to gold nanoparticles for lateral flow immunoassays, where clones P158 and P86 were able to detect SARS-CoV-2 spike protein variants.

VH domains of conventional antibodies share structural similarities with nanobodies and are also employed in nanoparticle conjugates. These single-domain antibodies provide similar benefits in terms of size and stability.

Therapeutic and Diagnostic Antibodies

Tumor-targeting antibodies are extensively conjugated with nanoparticles for cancer imaging and therapy. Gold nanoparticles conjugated with antibodies have been used to enhance imaging quality and provide anatomical information on tumor tissues. These conjugates enable non-invasive detection of overexpressed tumor surface antigens and help determine suitable therapeutic strategies.

Cell membrane proteins represent a unique approach where entire cellular membranes containing multiple surface antigens are coated onto nanoparticles. RBC membrane-coated nanoparticles retain natural cell membrane and surface antigens, serving as antibody decoys for treating autoimmune conditions like autoimmune hemolytic anemia. This biomimetic approach leverages the complete antigen profile rather than individual antibodies.

Disease-Specific Antibodies

Anti-parasite antibodies conjugated to nanoparticle carriers have shown promise in targeted drug delivery. High-affinity nanobodies against trypanosome variant surface glycoprotein (VSG) have been conjugated to poly(lactic-co-glycolic acid) or chitosan carriers loaded with pentamidine for treating parasitic infections. Similarly, nanobodies against Pfs230, a Plasmodium falciparum surface protein, have been developed for malaria transmission-blocking applications.

Anti-toxin and anti-venom antibodies have been engineered in nanobody formats for treating envenoming. These include nanobody-Fc conjugates against scorpion toxins and snake venom components, which can be produced in various expression systems.

Detection and Diagnostic Proteins

Pathogen detection antibodies are commonly conjugated to nanoparticles for immunoassays. Serum ferritin-specific nanobodies have been employed in sandwich ELISA formats achieving detection limits of 1.01 ng/mL. Nanobodies targeting hepatitis E virus ORF2 protein have demonstrated high sensitivity and specificity across multiple viral genotypes. Additionally, nanobodies for detecting Clostridioides difficile toxins TcdA and TcdB have been developed for both in vitro and in vivo diagnostic applications.

The diversity of antibodies and proteins used with nanoparticles reflects the broad applicability of this technology across oncology, infectious diseases, autoimmune disorders, and diagnostic platforms, with each application selecting biomolecules optimized for specific targeting, stability, and functional requirements.

From the search results, there is limited information directly related to "clone nan" citations in scientific literature. However, one relevant mention in the context of a novel voltage-gated sodium channel is as follows:

  • NaN (Novel Voltage-Gated Sodium Channel): The scientific literature discussing a clone named "NaN" focuses on a novel voltage-gated sodium channel α-subunit. Key findings include:
    • Expression and Characteristics: NaN is expressed preferentially in sensory neurons and is predicted to be tetrodotoxin-resistant (TTX-R) and voltage-gated. It shares structural features with both subfamilies 1 and 2 of sodium channels, suggesting an ancestral relationship or a possible third subfamily.
    • Molecular Features: NaN exhibits a unique amino acid sequence with significant similarity to other sodium channels like SNS/PN3 and rH1, but with distinct differences, such as a longer linker joining S3 and S4 of D4, which may contribute to faster recovery from inactivation.

These references do not specifically discuss "clone nan" in terms of its citation metrics or impacts in broader scientific literature. Instead, they focus on the molecular and functional characteristics of a specific sodium channel referred to as "NaN."

Dosing regimens of antibody clones and drugs in mouse models vary by multiple factors, including the specific clone, the mouse strain, disease model, desired biological effect, and experimental objectives. There is no universal dosing schedule, and optimization often depends on both the antibody/drug properties and the intended outcome of the experiment.

Key factors influencing dosing regimens:

  • Clone-specific properties: Each antibody clone (e.g., anti-PD-1, anti-CD4) has recommended dose ranges and schedules based on prior efficacy and safety studies.
  • Mouse strain/disease model: Different mouse genetic backgrounds and disease contexts (such as cancer, infection, autoimmunity) can require alterations in dose to achieve desired pharmacodynamic or immunological effects.
  • Route of administration: Most commonly, antibodies are delivered intraperitoneally, but alternative routes (such as intratumoral) are sometimes used depending on the study goal.

Examples of clone-specific dosing regimens (for commonly used monoclonal antibodies in mice):

Antibody Clone (Target)Dose per MouseFrequency & DurationTypical Disease Models
RMP1-14 (PD-1)200–500 μgEvery 3–4 daysCancer, infection
10F.9G2 (PD-L1)100–250 μg2–3 times/weekCancer, infection
29F.1A12 (PD-1)100–200 μg3 times at 3-day intervalsCancer
9H10 (CTLA-4)100–200 μgEvery ~3 daysCancer
GK1.5 (CD4)200–250 μg2–3 times/weekImmunodepletion
2.43 (CD8)250 μg2–3 times/weekImmunodepletion
RB6-8C5 (Gr-1/neutrophils)200–250 μgEvery 2–3 daysNeutrophil depletion

Source for detailed regimens above:

Regimen variation due to strain or model:

  • Doses might be adjusted for immunocompromised versus wild-type mice, or between syngeneic versus xenograft tumor models.
  • Chronic versus acute disease models may use different intervals or cumulative doses.
  • Strains (e.g., C57BL/6J, BALB/c) can have differing sensitivities to both antibodies and drugs, affecting optimal dose.
  • Route changes (e.g., intratumoral versus intraperitoneal) may be used for local versus systemic effects.

Principles for regimen selection:

  • Researchers generally start with literature- or manufacturer-recommended doses and frequency.
  • Pilot experiments often refine these regimens for the specific experimental setup.
  • Dosing regimens are chosen to balance efficacy (e.g., complete target engagement or depletion) with safety/tolerability (avoiding toxicity or off-target immunosuppression).

Isotype controls and irrelevant clones:

  • Doses for isotype controls or non-binding clones should match those of the test clone and can also vary by study aim or mouse background.

Summary: Dosing regimens of antibody clones in mice are highly variable and are tailored to the specific biological question, the clone being used, the mouse strain, the disease context, and experimental design. Always consult the literature for the precise clone, mouse model, and context to select or adapt an evidence-based regimen.

References & Citations

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