Anti-LCMV nucleoprotein – Purified in vivo PLATINUM™ Functional Grade

Anti-LCMV nucleoprotein – Purified in vivo PLATINUM™ Functional Grade

Product No.: L331

- -
- -
Clone
VL-4
Target
LCMV Nucleoprotein
Formats AvailableView All
Product Type
Hybridoma Monoclonal Antibody
Alternate Names
Protein N, LCMV Nucleocapsid Protein
Isotype
Rat IgG2a κ
Applications
ELISA
,
FA
,
FC
,
IF

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
LCMV
Host Species
Rat
Recommended Isotype Controls
Recommended Isotype Controls
Recommended Dilution Buffer
Immunogen
Lymphocytic choriomeningitis virus (LCMV)
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 0.5 EU / ml as determines by the LAL method
Purity
≥98% 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.
State of Matter
Liquid
Product Preparation
Functional grade preclinical antibodies are manufactured in an animal free facility using only in vitro protein free 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.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s Purified Functional PLATINUM<sup>TM</sup> antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
This antibody 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.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
IF
N
FA
FC
Focus Formation Assay
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
VL-4 activity is directed against LCMV nucleoprotein, staining LCMV-infected cells internally.
VL-4 does not react with influenza-, vaccinia-, or vesicular stomatitis-infected cells.
Background
Lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogenic arenavirus with worldwide distribution 1,2. Arenaviruses cause human infection through mucosal exposure to aerosols or by direct contact with abraded skin of infected rodents 1. LCMV infection may carry health risks for humans who are immunocompromised or pregnant 2.

LCMV infection in mice can be acute or persistent depending on age, immunocompetence, genetic background, route of infection, strain, and dosage 1. Due to the versatility of outcome, LCMV mouse models are extensively used to examine basic questions of immunology and virology, including: virus-induced immunopathological disease, MHC restriction, T cell and B cell regulation, T cell-mediated killing, and immune T cell therapy in clearing viral infection 1,2. LCMV mouse models were used to identify PD-1 as a critical regulator of T cell exhaustion 2 and have also been used to study the synergy between PD-1 blockade and IL-2 cytokine in cancer immunotherapy 3.

LCMV is an enveloped virus with a bi-segmented negative-stranded, ambisense RNA genome 1,2. LCMV has a non-cytolytic life cycle restricted to the cell cytoplasm. Cell entry is cholesterol-dependent but clathrin-, dynamin-, caveolin-, ARF6-, flotillin-, and actin-independent and occurs via receptor-mediated endocytosis utilizing alpha-dystroglycan as the main extracellular matrix protein receptor 1. Most of the disease caused by LCMV is mediated by the host T cell response 2.

Arenavirus nucleoprotein (NP) is the most abundant viral protein component in virions as well as in infected cells 4. NP encapsidates the viral genomic RNA and is part of the viral ribonucleoprotein complex that directs viral RNA replication and gene transcription in the cytoplasm of infected cells. NP also counteracts host type I interferon response during infection via a functional 3’–5’ exonuclease domain in its C-terminal region 5,6. The same domain also interacts with LCMV Z, as well as Lassa Virus Z, but different residues are involved 4. This NP-Z interaction is a novel target for antiviral drug development.

VL-4 antibody was generated by immunizing a (Louvain X DA) F1 rat with LCMV strain WE and fusing the resulting spleen cells with the YM3 myeloma cell line 7.
Antigen Distribution
LCMV nucleoprotein encapsidates the viral genome RNA and is present in infected cells and virions.
UniProt.org
Research Area
Infectious Disease
.
IVD Raw Material

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.

There is no specific information in the search results about the common in vivo applications of clone VL-4 in mice. However, a similar antibody-related term, VL-4, is mentioned in the context of staining LCMV (Lymphocytic Choriomeningitis Virus) nucleoprotein, which is used to identify LCMV-infected cells. This suggests that VL-4 might be used in research related to viral infections, but without further details, it is unclear if this specifically applies to clone VL-4 or if there is confusion with another antibody.

If you are looking for information on antibodies or clones used in mice for in vivo studies, you might be interested in other antibodies like PS/2 for VLA-4 neutralization or GK1.5 for CD4+ T cell depletion. These antibodies are used in various immunological studies.

If clone VL-4 is mixed up with another entity or if there's a specific application not covered, additional context or clarification might be needed to provide a more accurate answer.

In the literature, the VL-4 antibody, which targets Lymphocytic Choriomeningitis virus (LCMV) nucleoprotein, is often used in isolation for specific applications like ELISA, flow cytometry, and immunofluorescence . However, when considering other commonly used antibodies or proteins in similar contexts, several options emerge:

  1. VHHs (Single-Domain Antibodies): These are highly versatile and used in various therapeutic applications, including cancer, infectious diseases, and inflammatory conditions. They are often used due to their high stability and ability to target specific antigens .

  2. Anti-EGFR Antibodies: Used in cancer treatment for blocking epidermal growth factor receptor interactions, these can be in monovalent or bivalent formats to enhance efficacy .

  3. Anti-TNFα Antibodies: These are used for treating conditions like rheumatoid arthritis by blocking tumor necrosis factor-alpha interactions .

  4. Anti-VLA-4 Antibodies: Targeting Very Late Antigen 4, these are used in treating autoimmune diseases like multiple sclerosis and rheumatoid arthritis by inhibiting leukocyte migration .

These antibodies and proteins are not necessarily used directly with VL-4 but are significant in immunological research and therapeutic applications.

For specific applications involving VL-4, such as studying LCMV infections, researchers might use other antibodies targeting viral or cellular components relevant to the study context, but these are not directly mentioned in the available literature.

The key findings related to clone VL-4 are not explicitly detailed in the current indexed scientific literature. None of the provided search results specifically reference "clone VL-4" in the context of widely tracked scientific topics such as immunology, virology, molecular biology, or experimental methods. There are results describing other clones and their applications (such as CD4 T cell clones, CD155 antibodies, or Fv-4 gene retroviral clones), but "VL-4" does not appear among these.

  • If you are referencing a specific monoclonal antibody, genetic clone, or cell line termed "VL-4," the indexed literature does not provide findings for it directly.
  • It is possible "VL-4" may be referenced in a niche study not included in major open-access repositories indexed by this search.

If you intended a different clone identifier or a specific research field, please provide additional context or clarification so results can be targeted more precisely.

The dosing regimens for clone VL-4 in mouse models are not directly specified in the provided search results, and no specific guide or protocol was found referencing the use of "VL-4" in this context. However, extensive information is available for analogous in vivo antibody dosing in mouse models (e.g., anti-PD-1, anti-PD-L1, anti-CTLA-4 clones), which may provide insight into general best practices for injectable monoclonal antibodies in mice.

General Guidelines for Monoclonal Antibody Dosing in Mice

  • Dose Range: Typical monoclonal antibodies used for immune checkpoint blockade in mice (such as RMP1-14 for PD-1, 10F.9G2 for PD-L1, or 9D9/9H10 for CTLA-4) are administered at doses of 100–250 μg per mouse per injection.
  • Administration Route: The standard route is intraperitoneal injection.
  • Dosing Frequency: Most immunotherapy antibodies are given every 2–4 days, with 2–3 times per week being common for sustained blockade.
  • Duration: Treatment duration varies by study design, but repeated dosing over 2–3 weeks is typical in tumor models.
  • Drug Clearance: Dose selection may also need to account for target-mediated drug disposition or the development of anti-drug antibodies (ADAs), which can accelerate clearance and reduce efficacy.
  • Customization: Dosing may be adjusted based on the antibody’s pharmacokinetics, target expression, and model-specific requirements (e.g., tumor burden, immune status).

Factors Influencing Dosing Regimens

  • Target Density: High target expression may require higher or more frequent dosing.
  • Antibody Affinity: Higher affinity may allow for lower doses or less frequent administration.
  • Effector Function: Engineered antibodies lacking Fc effector function (like clone 80, an anti-PD-L1 surrogate) are dosed similarly to those with effector function, but clinical relevance may differ.
  • Model Purpose: Infection, autoimmunity, and cancer models may have different dosing needs; oncology models often use more aggressive regimens to maximize therapeutic effect.
  • Combination Therapies: When combined with other agents, dose and schedule may be modified to minimize toxicity while maintaining efficacy.

Example Regimens for Reference

Clone/AgentTypical Dose (μg/mouse)RouteFrequencyApplication
RMP1-14 (anti-PD-1)200–500IntraperitonealEvery 3–4 daysCancer immunotherapy
10F.9G2 (anti-PD-L1)100–250Intraperitoneal2–3x/weekCancer, infection models
9D9 (anti-CTLA-4)100–250IntraperitonealEvery 3 daysCancer immunotherapy

Summary

While specific dosing details for clone VL-4 are not provided in the available literature, researchers typically administer monoclonal antibodies like VL-4 at doses of 100–250 μg per mouse, delivered intraperitoneally, 2–3 times per week, adjusting as needed based on pharmacokinetic data and model-specific requirements. It is essential to consult the literature for the specific antibody clone and model, perform pilot pharmacokinetic studies, and monitor for signs of toxicity or loss of efficacy due to ADA development. If VL-4 is a novel or less commonly used clone, detailed characterization (binding, blocking, PK/PD) should precede large-scale in vivo studies.

References & Citations

1. Grande-Pérez A, Martin V, Moreno H, et al. Curr Top Microbiol Immunol. 392:231-276. 2016.
2. Dangi T, Chung YR, Palacio N, et al. Curr Protoc Immunol. 130(1):e99. 2020.
3. Hashimoto M, Araki K, Cardenas MA, et al. Nature. 610(7930):173-181. 2022.
4. Ortiz-Riaño E, Cheng BY, de la Torre JC, et al. J Virol. 85(24):13038-13048. 2011.
5. Martínez-Sobrido L, Zúñiga EI, Rosario D, et al. J Virol. 80: 9192–9199. 2006.
6. Borrow P, Martinez-Sobrido L, de la Torre JC. Viruses 2: 2443–2480. 2010.
7. Battegay M, Cooper S, Althage A, et al. J Virol Methods. 33(1-2):191-198. 1991.
8. Seiler P, Kalinke U, Rülicke T, et al. J Virol. 72(3):2253-2258. 1998.
9. Straub T, Schweier O, Bruns M, et al. Eur J Immunol. 43(9):2338-2348. 2013.
Indirect Elisa Protocol
FA
Flow Cytometry
IF

Certificate of Analysis

- -
- -

Formats Available

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