Mouse IgG2b Isotype Control (12B9) | PA007130
$150.00 – $800.00
Recombinant mouse IgG2b isotype control and mutants good for in vitro and in vivo studies. Low or no specific binding to mouse samples tested. More choices of recombinant mouse IgG2b isotype controls, including targets, mutants, tags, and conjagates.
- Details & Specifications
- References
- More Offers
Catalog No. | PA007130 |
---|---|
Product Name | Mouse IgG2b Isotype Control (12B9) | PA007130 |
Supplier Name | Syd Labs, Inc. |
Brand Name | Syd Labs |
Synonyms | Recombinant Mouse IgG2b Isotype Antibody |
Summary | The In Vivo Grade Recombinant Mouse IgG2b Isotype Control Antibody was produced in HEK 293 cells. |
Clone | 12B9 |
Isotype | mouse IgG2b, kappa |
Applications | an isotype-matched negative control for mouse IgG2b isotype antibody used in ELISA, Western Blot (WB), Flow Cytometry (Flow), Immunoprecipitation (IP), Immunohistochemistry (Paraffin) (IHC (P)), Immunohistochemistry (Frozen) (IHC (F)), and in vivo animal model research. |
Immunogen | N/A. |
Form Of Antibody | 0.2 μM filtered solution of 1x PBS. |
Endotoxin | Less than 1 EU/mg of protein as determined by LAL method. |
Purity | >95% by SDS-PAGE under reducing conditions. |
Shipping | Recombinant Mouse IgG2b Isotype Control Antibody is shipped with ice pack. Upon receipt, store it immediately at the temperature recommended below. |
Stability & Storage | Use a manual defrost freezer and avoid repeated freeze-thaw cycles. 1 month from date of receipt, 2 to 8°C as supplied. 3 months from date of receipt, -20°C to -70°C as supplied. |
Order Offline | Phone: 1-617-401-8149 Fax: 1-617-606-5022 Email: message@sydlabs.com Or leave a message with a formal purchase order (PO) Or credit card. |
Description
PA007130: Recombinant Mouse IgG2b Isotype Control Antibody (Clone: 12B9), In Vivo Grade
The Mouse IgG2b Isotype Control Antibody (Clone: 12B9) is a high-purity monoclonal antibody engineered to function as a negative control in immunological experiments, such as flow cytometry and immunohistochemistry. This antibody is meticulously produced to deliver consistent, reliable performance, enabling researchers to distinguish non-specific binding and background signals from true experimental outcomes. Its high quality ensures precision, making it an essential tool for validating results in a variety of research settings.
Sourced from the renowned Clone: 12B9, this antibody is widely respected in the scientific community for its exceptional specificity and minimal cross-reactivity. Provided by a trusted biotechnology supplier with over two decades of expertise, the Mouse IgG2b Isotype Control Antibody undergoes stringent quality control processes to guarantee purity and functionality across batches. This reliability empowers researchers to confidently use it in critical applications, ensuring their data reflects accurate biological interactions without interference.
Ideal for use in flow cytometry, immunohistochemistry, and other immunological assays, this antibody enhances experimental accuracy and reproducibility. Syd Labs’s in vivo grade recombinant mouse IgG2b isotype control antibody (mIgG2b isotype control) was produced in HEK 293 cells. Backed by Syd Labs’s commitment to excellence and comprehensive technical support, the Mouse IgG2b Isotype Control Antibody (Clone: 12B9) is a valuable asset for any laboratory aiming to achieve robust and dependable research outcomes. Contact us today to learn more about availability and how this product can support your scientific endeavors.
References for Recombinant Mouse IgG2b Isotype Control (12B9):
1、Antibody effector functions are associated with protection from respiratory syncytial virus
Yannic C Bartsch,et al.Cell. 2022.PMCID: 36513064
“Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.
Keywords: Ad26; Fc effector functions; Fc glycosylation; IgA; RSV; antibodies; human challenge study; mAbs; vaccination.”
2、The gut microbiota posttranslationally modifies IgA1 in autoimmune glomerulonephritis
Patrick J Gleeson,et al.Sci Transl Med. 2024.PMCID: 38536935
“Mechanisms underlying the disruption of self-tolerance in acquired autoimmunity remain unclear. Immunoglobulin A (IgA) nephropathy is an acquired autoimmune disease where deglycosylated IgA1 (IgA subclass 1) auto-antigens are recognized by IgG auto-antibodies, forming immune complexes that are deposited in the kidneys, leading to glomerulonephritis. In the intestinal microbiota of patients with IgA nephropathy, there was increased relative abundance of mucin-degrading bacteria, including Akkermansia muciniphila. IgA1 was deglycosylated by A. muciniphila both in vitro and in the intestinal lumen of mice. This generated neo-epitopes that were recognized by autoreactive IgG from the sera of patients with IgA nephropathy. Mice expressing human IgA1 and the human Fc α receptor I (α1KI-CD89tg) that underwent intestinal colonization by A. muciniphila developed an aggravated IgA nephropathy phenotype. After deglycosylation of IgA1 by A. muciniphila in the mouse gut lumen, IgA1 crossed the intestinal epithelium into the circulation by retrotranscytosis and became deposited in the glomeruli of mouse kidneys. Human α-defensins-a risk locus for IgA nephropathy-inhibited growth of A. muciniphila in vitro. A negative correlation observed between stool concentration of α-defensin 6 and quantity of A. muciniphila in the guts of control participants was lost in patients with IgA nephropathy. This study demonstrates that gut microbiota dysbiosis contributes to generation of auto-antigens in patients with IgA nephropathy and in a mouse model of this disease.”
3、Mouse IgG2a Isotype Therapeutic Antibodies Elicit Superior Tumor Growth Control Compared with mIgG1 or mIgE
Natasa Vukovic,et al.Cancer Res Commun. 2023.PMCID: 36968226
“In the last decades, antibody-based tumor therapy has fundamentally improved the efficacy of treatment for patients with cancer. Currently, almost all tumor antigen-targeting antibodies approved for clinical application are of IgG1 Fc isotype. Similarly, the mouse homolog mIgG2a is the most commonly used in tumor mouse models. However, in mice, the efficacy of antibody-based tumor therapy is largely restricted to a prophylactic application. Direct isotype comparison studies in mice in a therapeutic setting are scarce. In this study, we assessed the efficacy of mouse tumor-targeting antibodies of different isotypes in a therapeutic setting using a highly systematic approach. To this end, we engineered and expressed antibodies of the same specificity but different isotypes, targeting the artificial tumor antigen CD90.1/Thy1.1 expressed by B16 melanoma cells. Our experiments revealed that in a therapeutic setting mIgG2a was superior to both mIgE and mIgG1 in controlling tumor growth. Furthermore, the observed mIgG2a antitumor effect was entirely Fc mediated as the protection was lost when an Fc-silenced mIgG2a isotype (LALA-PG mutations) was used. These data confirm mIgG2a superiority in a therapeutic tumor model.
Significance: Direct comparisons of different antibody isotypes of the same specificity in cancer settings are still scarce. Here, it is shown that mIgG2a has a greater effect compared with mIgG1 and mIgE in controlling tumor growth in a therapeutic setting.”
4、Intranasal vaccination with lipid-conjugated immunogens promotes antigen transmucosal uptake to drive mucosal and systemic immunity
Brittany L Hartwell,et al.Sci Transl Med. 2022.PMCID: 35857825
“To combat the HIV epidemic and emerging threats such as SARS-CoV-2, immunization strategies are needed that elicit protection at mucosal portals of pathogen entry. Immunization directly through airway surfaces is effective in driving mucosal immunity, but poor vaccine uptake across the mucus and epithelial lining is a limitation. The major blood protein albumin is constitutively transcytosed bidirectionally across the airway epithelium through interactions with neonatal Fc receptors (FcRn). Exploiting this biology, here, we demonstrate a strategy of “albumin hitchhiking” to promote mucosal immunity using an intranasal vaccine consisting of protein immunogens modified with an amphiphilic albumin-binding polymer-lipid tail, forming amph-proteins. Amph-proteins persisted in the nasal mucosa of mice and nonhuman primates and exhibited increased uptake into the tissue in an FcRn-dependent manner, leading to enhanced germinal center responses in nasal-associated lymphoid tissue. Intranasal immunization with amph-conjugated HIV Env gp120 or SARS-CoV-2 receptor binding domain (RBD) proteins elicited 100- to 1000-fold higher antigen-specific IgG and IgA titers in the serum, upper and lower respiratory mucosa, and distal genitourinary mucosae of mice compared to unmodified protein. Amph-RBD immunization induced high titers of SARS-CoV-2-neutralizing antibodies in serum, nasal washes, and bronchoalveolar lavage. Furthermore, intranasal amph-protein immunization in rhesus macaques elicited 10-fold higher antigen-specific IgG and IgA responses in the serum and nasal mucosa compared to unmodified protein, supporting the translational potential of this approach. These results suggest that using amph-protein vaccines to deliver antigen across mucosal epithelia is a promising strategy to promote mucosal immunity against HIV, SARS-CoV-2, and other infectious diseases.”
5、A bivalent Epstein-Barr virus vaccine induces neutralizing antibodies that block infection and confer immunity in humanized mice
Chih-Jen Wei,et al.Sci Transl Med. 2022.PMCID: 35507671
“Epstein-Barr virus (EBV) is the major cause of infectious mononucleosis and is associated with several human cancers and, more recently, multiple sclerosis. Despite its prevalence and health impact, there are currently no vaccines or treatments. Four viral glycoproteins (gp), gp350 and gH/gL/gp42, mediate entry into the major sites of viral replication, B cells, and epithelial cells. Here, we designed a nanoparticle vaccine displaying these proteins and showed that it elicits potent neutralizing antibodies that protect against infection in vivo. We designed single-chain gH/gL and gH/gL/gp42 proteins that were each fused to bacterial ferritin to form a self-assembling nanoparticle. Structural analysis revealed that single-chain gH/gL and gH/gL/gp42 adopted a similar conformation to the wild-type proteins, and the protein spikes were observed by electron microscopy. Single-chain gH/gL or gH/gL/gp42 nanoparticle vaccines were constructed to ensure product homogeneity needed for clinical development. These vaccines elicited neutralizing antibodies in mice, ferrets, and nonhuman primates that inhibited EBV entry into both B cells and epithelial cells. When mixed with a previously reported gp350 nanoparticle vaccine, gp350D123, no immune competition was observed. To confirm its efficacy in vivo, humanized mice were challenged with EBV after passive transfer of IgG from mice vaccinated with control, gH/gL/gp42+gp350D123, or gH/gL+gp350D123 nanoparticles. Although all control animals were infected, only one mouse in each vaccine group that received immune IgG had detectable transient viremia. Furthermore, no EBV lymphomas were detected in immune animals. This bivalent EBV nanoparticle vaccine represents a promising candidate to prevent EBV infection and EBV-related malignancies in humans.”
6、Nasal vaccination of triple-RBD scaffold protein with flagellin elicits long-term protection against SARS-CoV-2 variants including JN.1
Xian Li,et al.Signal Transduct Target Ther. 2024.PMCID: 38678055
“Developing a mucosal vaccine against SARS-CoV-2 is critical for combatting the epidemic. Here, we investigated long-term immune responses and protection against SARS-CoV-2 for the intranasal vaccination of a triple receptor-binding domain (RBD) scaffold protein (3R-NC) adjuvanted with a flagellin protein (KFD) (3R-NC + KFDi.n). In mice, the vaccination elicited RBD-specific broad-neutralizing antibody responses in both serum and mucosal sites sustained at high level over a year. This long-lasting humoral immunity was correlated with the presence of long-lived RBD-specific IgG- and IgA-producing plasma cells, alongside the Th17 and Tfh17-biased T-cell responses driven by the KFD adjuvant. Based upon these preclinical findings, an open labeled clinical trial was conducted in individuals who had been primed with the inactivated SARS-CoV-2 (IAV) vaccine. With a favorable safety profile, the 3R-NC + KFDi.n boost elicited enduring broad-neutralizing IgG in plasma and IgA in salivary secretions. To meet the challenge of frequently emerged variants, we further designed an updated triple-RBD scaffold protein with mutated RBD combinations, which can induce adaptable antibody responses to neutralize the newly emerging variants, including JN.1. Our findings highlight the potential of the KFD-adjuvanted triple-RBD scaffold protein is a promising prototype for the development of a mucosal vaccine against SARS-CoV-2 infection.”
7、A spike-based mRNA vaccine that induces durable and broad protection against porcine deltacoronavirus in piglets
Jizong Li,et al.J Virol. 2024.PMCID: 39158273
“Coronaviruses (CoVs) are important pathogens for humans and other vertebrates, causing severe respiratory and intestinal infections that have become a threat to public health because of the potential for interspecies transmission between animals and humans. Therefore, the development of safe, effective vaccines remains a top priority for the control of CoV infection. The unique immunological characteristics of vaccines featuring messenger RNA (mRNA) present an advantageous tool for coronavirus vaccine development. Here, we designed two lipid nanoparticle (LNP)-encapsulated mRNA (mRNA-LNP) vaccines: one encoding full-length spike (S) protein and the other encoding the spike ectodomain (Se) from porcine deltacoronavirus (PDCoV). Fourteen days after primary immunization, both mRNA vaccines induced high levels of immunoglobulin G and neutralizing antibodies in mice, with the S vaccine showing better performance than the Se vaccine. Passive immune protection of the S mRNA vaccine in suckling piglets was confirmed by the induction of robust PDCoV-specific humoral and cellular immune responses. The S mRNA vaccine also showed better protective effects than the inactivated vaccine. Our results suggest that the novel PDCoV-S mRNA-LNP vaccine may have the potential to combat PDCoV infection.
Importance: As an emerging porcine enteropathogenic coronavirus, porcine deltacoronavirus (PDCoV) has the potential for cross-species transmission, attracting extensive attention. Messenger RNA (mRNA) vaccines are a promising option for combating emerging and re-emerging infectious diseases, as evidenced by the demonstrated efficacy of the COVID-19 mRNA vaccine. Here, we first demonstrated that PDCoV-S mRNA-lipid nanoparticle (LNP) vaccines could induce potent humoral and cellular immune responses in mice. An evaluation of passive immune protection of S mRNA vaccines in suckling piglets confirmed that the protective effect of mRNA vaccine was better than that of inactivated vaccine. This study suggests that the PDCoV-S mRNA-LNP vaccine may serve as a potential and novel vaccine candidate for combating PDCoV infection.
Keywords: PDCoV; lipid nanoparticle; mRNA vaccine; spike protein; suckling piglets.”
8、Modeling MOG Antibody-Associated Disorder and Neuromyelitis Optica Spectrum Disorder in Animal Models: Visual System Manifestations
Jana Remlinger,et al.Neurol Neuroimmunol Neuroinflamm. 2023.PMCID: 37429715
“Background and objectives: Mechanisms of visual impairment in aquaporin 4 antibody (AQP4-IgG) seropositive neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody (MOG-IgG)-associated disorder (MOGAD) are incompletely understood. The respective impact of optic nerve demyelination and primary and secondary retinal neurodegeneration are yet to be investigated in animal models.
Methods: Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6Jrj mice, and monoclonal MOG-IgG (8-18C5, murine), recombinant AQP4-IgG (rAb-53, human), or isotype-matched control IgG (Iso-IgG, human) was administered 10 days postimmunization. Mobility impairment was scored daily. Visual acuity by optomotor reflex and ganglion cell complex thickness (GCC, 3 innermost retinal layers) by optical coherence tomography (OCT) were longitudinally assessed. Histopathology of optic nerve and retina was investigated during presymptomatic, acute, and chronic disease phases for immune cells, demyelination, complement deposition, natural killer (NK) cell, AQP4, and astrocyte involvement, retinal ganglion cells (RGCs), and Müller cell activation. Groups were compared by nonparametric tests with a p value <0.05 indicating statistical significance.
Results: Visual acuity decreased from baseline to chronic phase in MOG-IgG (mean ± standard error of the mean: 0.54 ± 0.01 to 0.46 ± 0.02 cycles/degree, p < 0.05) and AQP4-IgG EAE (0.54 ± 0.01 to 0.43 ± 0.02, cycles/degree, p < 0.05). Immune cell infiltration of optic nerves started in presymptomatic AQP4-IgG, but not in MOG-IgG EAE (5.85 ± 2.26 vs 0.13 ± 0.10 macrophages/region of interest [ROI] and 1.88 ± 0.63 vs 0.15 ± 0.06 T cells/ROI, both p < 0.05). Few NK cells, no complement deposition, and stable glial fibrillary acid protein and AQP4 fluorescence intensity characterized all EAE optic nerves. Lower GCC thickness (Spearman correlation coefficient r = -0.44, p < 0.05) and RGC counts (r = -0.47, p < 0.05) correlated with higher mobility impairment. RGCs decreased from presymptomatic to chronic disease phase in MOG-IgG (1,705 ± 51 vs 1,412 ± 45, p < 0.05) and AQP4-IgG EAE (1,758 ± 14 vs 1,526 ± 48, p < 0.01). Müller cell activation was not observed in either model.
Discussion: In a multimodal longitudinal characterization of visual outcome in animal models of MOGAD and NMOSD, differential retinal injury and optic nerve involvement were not conclusively clarified. Yet optic nerve inflammation was earlier in AQP4-IgG-associated pathophysiology. Retinal atrophy determined by GCC thickness (OCT) and RGC counts correlating with mobility impairment in the chronic phase of MOG-IgG and AQP4-IgG EAE may serve as a generalizable marker of neurodegeneration.”
9、Liver macrophages and sinusoidal endothelial cells execute vaccine-elicited capture of invasive bacteria
Juanjuan Wang,et al.Sci Transl Med. 2023.PMCID: 38117903
“Vaccination has substantially reduced the morbidity and mortality of bacterial diseases, but mechanisms of vaccine-elicited pathogen clearance remain largely undefined. We report that vaccine-elicited immunity against invasive bacteria mainly operates in the liver. In contrast to the current paradigm that migrating phagocytes execute vaccine-elicited immunity against blood-borne pathogens, we found that invasive bacteria are captured and killed in the liver of vaccinated host via various immune mechanisms that depend on the protective potency of the vaccine. Vaccines with relatively lower degrees of protection only activated liver-resident macrophage Kupffer cells (KCs) by inducing pathogen-binding immunoglobulin M (IgM) or low amounts of IgG. IgG-coated pathogens were directly captured by KCs via multiple IgG receptors FcγRs, whereas IgM-opsonized bacteria were indirectly bound to KCs via complement receptors of immunoglobulin superfamily (CRIg) and complement receptor 3 (CR3) after complement C3 activation at the bacterial surface. Conversely, the more potent vaccines engaged both KCs and liver sinusoidal endothelial cells by inducing higher titers of functional IgG antibodies. Endothelial cells (ECs) captured densely IgG-opsonized pathogens by the low-affinity IgG receptor FcγRIIB in a “zipper-like” manner and achieved bacterial killing predominantly in the extracellular milieu via an undefined mechanism. KC- and endothelial cell-based capture of antibody-opsonized bacteria also occurred in FcγR-humanized mice. These vaccine protection mechanisms in the liver not only provide a comprehensive explanation for vaccine-/antibody-boosted immunity against invasive bacteria but also may serve as in vivo functional readouts of vaccine efficacy.”
10、An FcRn-targeted mucosal vaccine against SARS-CoV-2 infection and transmission
Weizhong Li,et al.Nat Commun. 2023.PMCID: 37932271
“SARS-CoV-2 is primarily transmitted through droplets and airborne aerosols, and in order to prevent infection and reduce viral spread vaccines should elicit protective immunity in the airways. The neonatal Fc receptor (FcRn) transfers IgG across epithelial barriers and can enhance mucosal delivery of antigens. Here we explore FcRn-mediated respiratory delivery of SARS-CoV-2 spike (S). A monomeric IgG Fc was fused to a stabilized spike; the resulting S-Fc bound to S-specific antibodies and FcRn. Intranasal immunization of mice with S-Fc and CpG significantly induced antibody responses compared to the vaccination with S alone or PBS. Furthermore, we intranasally immunized mice or hamsters with S-Fc. A significant reduction of virus replication in nasal turbinate, lung, and brain was observed following nasal challenges with SARS-CoV-2 and its variants. Intranasal immunization also significantly reduced viral airborne transmission in hamsters. Nasal IgA, neutralizing antibodies, lung-resident memory T cells, and bone-marrow S-specific plasma cells mediated protection. Hence, FcRn delivers an S-Fc antigen effectively into the airway and induces protection against SARS-CoV-2 infection and transmission.”
Other in vivo grade Recombinant Mouse IgG2b Isotype Control Antibodies and Mutants:
Recombinant MOPC-21 Mouse IgG2b Isotype Control Antibodies and Mutants, In vivo Grade
Recombinant HyHEL-10 Mouse IgG2b Isotype Control Antibodies and Mutants, In vivo Grade
Recombinant HEL D1.3 Mouse IgG2b Isotype Control Antibodies and Mutants, In vivo Grade
Recombinant C1.18.4 Mouse IgG2b Isotype Control Antibodies and Mutants, In vivo Grade
Recombinant B1-8 Mouse IgG2b Isotype Control Antibodies and Mutants, In vivo Grade
Other in vivo grade Recombinant IgG Isotype Control Antibodies and Mutants:
Recombinant Human IgG1 Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Human IgG2 Isotype Control Antibody, In vivo Grade
Recombinant Human IgG3 Isotype Control Antibody, In vivo Grade
Recombinant Human IgG4-S228P Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG1 Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG2a Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG2b Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG2c Isotype Control Antibody and Mutants, In vivo Grade
Recombinant Mouse IgG3 Isotype Control Antibody, In vivo Grade
Recombinant Rat IgG1 Isotype Control Antibody, In vivo Grade
Recombinant Rat IgG2a Isotype Control Antibody, In vivo Grade
Recombinant Rat IgG2b Isotype Control Antibody, In vivo Grade
Recombinant Rat IgG2c Isotype Control Antibody, In vivo Grade
Recombinant Hamster IgG1 Isotype Control Antibody, In vivo Grade
Recombinant Hamster IgG2 Isotype Control Antibody, In vivo Grade
In vivo Grade Recombinant IgG Fc Proteins:
Recombinant Human IgG1 Fc Protein (hIgG1), In vivo Grade
Recombinant Human IgG2 Fc Protein (hIgG2), In vivo Grade
Recombinant Human IgG4 Fc Protein (hIgG4), In vivo Grade
Recombinant Mouse IgG1 Fc Protein (mIgG1), In vivo Grade
Recombinant Mouse IgG2a Fc Protein (mIgG2a), In vivo Grade
Recombinant Mouse IgG2b Fc Protein (mIgG2b), In vivo Grade
Recombinant Rat IgG2a Fc Protein (rtIgG2a), In vivo Grade
Recombinant Rat IgG2b Fc Protein (rtIgG2b), In vivo Grade
Recombinant Llama IgG2b Fc Protein (lIgG2b), In vivo Grade
Recombinant Rabbit IgG Fc Protein (rIgG), In vivo Grade
Fc ELISA Kits and Reagents:
Human Fc ELISA Kit
Mouse Fc ELISA Kit
Human Fc ELISA Reagent Kit
Mouse Fc ELISA Reagent Kit
Mouse IgG2b Isotype Control from: In Vivo Grade Recombinant Mouse IgG2b Isotype Control Antibody: PA007130 Syd Labs