ARG2 Antibody

Arginase Type II, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to ARG2 Antibody

ARG2 (Arginase 2) antibodies are therapeutic agents designed to inhibit the enzymatic activity of ARG2, a mitochondrial metalloenzyme that converts L-arginine into L-ornithine and urea. Overexpression of ARG2 in tumor microenvironments depletes extracellular arginine, suppressing T-cell proliferation and promoting immune evasion in cancers such as pancreatic ductal adenocarcinoma, bowel cancer, and acute myeloid leukemia (AML) . ARG2 antibodies aim to restore arginine availability, reactivating antitumor immunity.

Mechanism of Action

ARG2 antibodies inhibit enzymatic activity through distinct mechanisms:

  • Non-competitive inhibition: Lead antibodies like C0021158 bind to epitopes outside the substrate-binding domain, inducing structural changes that block arginine access to the active site .

  • Conformational disruption: X-ray crystallography revealed that antibody binding alters ARG2’s trimeric structure, preventing manganese coordination essential for catalysis .

  • Specificity: C0021158 shows no cross-reactivity with ARG1 (a urea cycle enzyme), minimizing off-target effects .

AntibodyMechanismTarget SpecificityKey Advantage
C0021158Non-competitive inhibitionARG2 onlyHigh affinity (173 pM)
PA1805Polyclonal bindingARG2 (mitochondrial)Western blot validation
A-10Epitope recognitionARG2 (C-terminal)Detects multiple species

Enhanced T-Cell Activity

  • In vitro restoration: C0021158 fully reverses ARG2-mediated inhibition of CD8+ T-cell proliferation at 157 nM EC₅₀ .

  • Synergy with immunotherapy: ARG2-deficient CD8+ T cells exhibit superior tumor control when combined with PD-1 blockade .

Tumor Microenvironment Targeting

  • ARG2-specific T cells: CD8+ T cells recognize an HLA-B8-presented ARG2 epitope, targeting ARG2-expressing cancer cells and regulatory T cells (Tregs) .

  • AML suppression: ARG2 antibodies counteract arginine depletion caused by AML blasts, restoring immune function .

Therapeutic Potential in Oncology

ARG2 antibodies represent a novel strategy to address "immune cold" tumors by:

  1. Restoring arginine: Replenishing L-arginine levels to support T-cell metabolism and cytotoxic activity .

  2. Combination therapies: Synergizing with chemotherapy or checkpoint inhibitors (e.g., PD-1 blockade) .

  3. Reducing Treg activity: Eliminating immunosuppressive Tregs expressing high ARG2 .

Comparison with Other Antibodies

AntibodyTypeApplicationAffinitySelectivity
C0021158Human monoclonalTherapeuticHigh (173 pM)ARG2-specific
PA1805PolyclonalResearch (WB/IHC)ModerateMitochondrial ARG2
A-10Mouse monoclonalResearch (WB/IP/IF)ModerateBroad species
ab228700Rabbit polyclonalResearch (WB/IF/IHC)ModerateHuman/rodent

Challenges and Future Directions

  • Clinical translation: Partnering with pharmaceutical companies to advance C0021158 into trials .

  • Safety profile: Ensuring no hepatotoxicity due to ARG1 sparing .

  • Biomarker development: Identifying ARG2-expressing tumors to stratify patients .

Product Specs

Introduction
ARG2 plays a crucial role in regulating arginine metabolism outside the urea cycle and suppressing nitric oxide synthesis. In tissues beyond the liver, arginase influences the availability of L-arginine for NO synthase. Given the presence of NO synthase in the penile and clitoral corpus cavernosum smooth muscle and the vagina, ARG2 is implicated in both male and female sexual arousal. This makes it a potential therapeutic target for sexual arousal disorders in both sexes.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The solution is provided at a concentration of 1mg/ml and contains PBS at a pH of 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For storage up to 1 month, maintain the product at 4°C. For extended storage periods, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
The ARG2 antibody has undergone rigorous testing in various applications, including ELISA, Western blot analysis, flow cytometry, and ICC/IF, to confirm its specificity and reactivity. However, it is essential to optimize the antibody concentration for each specific application to achieve the best results.
Synonyms
Kidney Arginase, Non-Hepatic Arginase, EC 3.5.3.1, Arginase-2, Type II arginase, Kidney-type arginase, Arginase-2 mitochondrial, Arginase Type II, ARG2.
Purification Method
ARG2 antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT7F8AT.
Immunogen
Anti-human ARG2 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human ARG2 protein 23-354 amino acids purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is ARG2 and how does it differ structurally and functionally from ARG1?

ARG2 (Arginase 2) is a mitochondrial enzyme that catalyzes the hydrolysis of L-arginine into L-ornithine and urea, playing crucial roles in the urea cycle and nitric oxide regulation. The enzyme contributes significantly to L-arginine homeostasis and the biosynthesis of polyamines, which are essential for cell proliferation and differentiation . Unlike its paralogue ARG1 (Arginase 1), which is primarily expressed in the liver, ARG2 is predominantly found in extrahepatic tissues, particularly the kidney and small intestine .

The structural differences between these paralogues are notable. The human ARG2 gene is located on chromosome 14q24.1 and encodes a larger 354 amino acid protein, whereas ARG1 is located on chromosome 6q23 and encodes a 322 amino acid protein . A key distinguishing feature of ARG2 is its amino-terminal mitochondrial localization sequence, which directs the enzyme to the mitochondria, creating a unique subcellular distribution compared to the cytosolic localization of ARG1 .

Functionally, both enzymes catalyze the same reaction but serve different physiological roles due to their distinct tissue distribution patterns. ARG2 significantly modulates immune responses and vascular function by competing with nitric oxide synthase for L-arginine, thereby influencing nitric oxide production, endothelial function, and blood pressure regulation . This competition mechanism represents a critical point of intervention for therapeutic strategies targeting ARG2.

What types of ARG2 antibodies are available for research applications and how should they be selected?

Several types of ARG2 antibodies have been developed for research applications, each with specific characteristics suitable for different experimental needs. The Santa Cruz Biotechnology catalog offers the Arg2 Antibody (C-8), a mouse monoclonal IgG1 kappa light chain antibody specifically designed to target human ARG2, which was raised against amino acids 291-354 of human ARG2 . Another option is the Arg2 Antibody (A-10), a mouse monoclonal IgA kappa light chain antibody that targets an epitope between amino acids 304-335 near the C-terminus of human ARG2 .

For more specialized applications, researchers have developed inhibitory antibodies like C0021158, a high-affinity antibody generated through innovative affinity maturation techniques, which demonstrates complete inhibition of ARG2 enzymatic function . This particular antibody has shown promise in restoring T-cell proliferation in cancer research contexts .

When selecting an appropriate ARG2 antibody, researchers should consider several factors: the specific application (western blotting, immunoprecipitation, immunofluorescence, etc.), species reactivity requirements, antibody format (monoclonal vs. polyclonal), and whether inhibitory properties are needed for functional studies . The reactivity profile of each antibody should be examined carefully, as some antibodies like ARG2 (A-10) demonstrate cross-species reactivity with mouse, rat, human, canine, bovine, and porcine ARG2 .

For studies requiring highly specific inhibition of ARG2 without affecting ARG1, antibodies like C0021158 that have been specifically developed to avoid cross-reactivity with ARG1 would be most appropriate . Additionally, researchers should consider the binding epitope, as this can significantly impact the functional effects of the antibody on ARG2 activity.

What detection methods and experimental approaches are most effective when working with ARG2 antibodies?

ARG2 antibodies can be utilized across multiple detection platforms, with optimization strategies varying based on the specific application. For western blotting (WB), ARG2 antibodies like the C-8 and A-10 variants demonstrate high specificity and can effectively detect the 38-40 kDa ARG2 protein . When performing WB, researchers should optimize protein extraction methods, particularly given ARG2's mitochondrial localization, which may require specific isolation protocols to ensure sufficient yield.

Immunoprecipitation (IP) applications using ARG2 antibodies provide valuable tools for studying protein-protein interactions and post-translational modifications. Both C-8 and A-10 antibodies have demonstrated efficacy in IP experiments . For optimal results, researchers should consider using magnetic beads conjugated with protein A/G for IgG antibodies like C-8, while protein L-based approaches may be more appropriate for IgA antibodies like A-10.

Immunofluorescence (IF) and immunohistochemistry with paraffin-embedded sections (IHCP) require careful consideration of fixation methods, as overfixation may mask the ARG2 epitope and reduce antibody binding . Antigen retrieval steps, particularly heat-induced epitope retrieval in citrate buffer (pH 6.0), often improve staining results with ARG2 antibodies. For IF studies, researchers should consider the mitochondrial localization of ARG2 and may benefit from co-staining with mitochondrial markers to confirm appropriate subcellular localization.

Enzyme-linked immunosorbent assay (ELISA) approaches can be employed for quantitative analysis of ARG2 expression levels . Sandwich ELISA formats using two non-competing ARG2 antibodies targeting different epitopes can enhance specificity and sensitivity. For inhibition studies, specialized assays measuring ARG2 enzymatic activity in the presence of inhibitory antibodies like C0021158 have been developed, with IC50 values serving as important metrics for comparative analysis .

How do ARG2 inhibitory antibodies function in cancer research and what advantages do they offer over small molecule inhibitors?

ARG2 inhibitory antibodies represent a significant advancement in cancer immunotherapy research due to their ability to reverse the immunosuppressive effects of ARG2 overexpression in tumors. In various cancer types, including pancreatic ductal adenocarcinoma, bowel cancer, and acute myeloid leukemia, elevated ARG2 levels deplete local L-arginine concentrations in the tumor microenvironment . This depletion critically impairs T-cell function, as L-arginine is essential for immune cells to mount effective anti-tumor responses, thereby creating an immunosuppressive environment that allows tumors to evade immune surveillance .

The inhibitory antibody C0021158, developed through sophisticated affinity maturation techniques, demonstrates complete inhibition of ARG2 enzymatic function with an IC50 of 18.5 ± 5.1 nM in its IgG format . This inhibition effectively restores T-cell proliferation capacity, potentially reinstating immune-mediated tumor control . The antibody exhibits high specificity for ARG2, with no detectable binding to human ARG1 as assessed by bio-layer interferometry, addressing a critical requirement for targeted therapeutic intervention .

Compared to small molecule inhibitors, ARG2-specific therapeutic antibodies offer several advantages. Antibodies typically possess longer half-lives and better bioavailability, resulting in improved pharmacokinetic profiles . Additionally, while many small molecule inhibitors often fail to discriminate between ARG1 and ARG2 due to their highly conserved active sites, antibodies can be designed to target regions of sequence divergence outside the substrate binding domain, achieving paralog specificity and mitigating off-target, ARG1-mediated effects .

The mechanism of ARG2 inhibition by antibodies like C0021158 appears to be non-competitive, as revealed by crystallography studies showing that three Fab molecules bind per ARG2 trimer to form a highly symmetric complex . This binding mode does not directly block the active site but instead induces conformational changes that disrupt enzyme function, potentially offering advantages over competitive small molecule inhibitors in specific research contexts.

What structural insights have been revealed through crystallography studies of ARG2-antibody complexes?

Crystallography studies have provided remarkable insights into the molecular basis of ARG2 inhibition by antibodies, revealing structural features critical for understanding inhibition mechanisms and guiding further antibody development. X-ray crystallography analyses of affinity-matured inhibitory Fabs (C0021158 and C0021181) bound to full-length, trimeric ARG2 have yielded atomic resolution diffraction data sets that illuminate key structural aspects of these interactions .

The crystal structures revealed that ARG2 forms a homotrimer, with each subunit containing a binuclear manganese cluster essential for catalytic activity . When bound to inhibitory antibodies, the ARG2 trimer accommodates three Fab molecules to form a highly symmetric complex . This binding arrangement is significant because it indicates a stoichiometric relationship of one Fab per ARG2 monomer, suggesting a coordinated inhibition mechanism across the entire enzymatic complex.

One of the most striking findings from these structural studies is the extensive reorientation of the binding paratope that occurs in affinity-matured antibodies compared to their parent counterparts . This reorientation facilitates substantial increases in contact surface area and shape complementarity to the antigen, directly contributing to the enhanced binding affinity and inhibitory potency observed in these optimized antibodies . The structural data revealed that while some complementarity-determining regions (CDRs) like VLCDR1 and VHCDR3 remained highly conserved during affinity maturation, suggesting critical epitope interactions that cannot be modified without compromising binding, other CDRs underwent significant sequence changes .

The structures also provided evidence for a non-competitive inhibition mechanism, showing that the antibodies do not directly block the enzyme's active site . Instead, they appear to induce conformational changes in ARG2 that disrupt its catalytic function through allosteric effects. This structural insight explains the observed inhibition kinetics, including the shift from apparent negative cooperativity in the parent antibody (C0020187) to the steeper inhibitory profile seen with C0021158, suggesting a substantial change in binding mode following affinity maturation .

How have affinity maturation techniques advanced the development of ARG2 inhibitory antibodies?

A groundbreaking method combining antibody chain shuffling with a staggered-extension process was employed to produce unbiased libraries that recombined beneficial mutations from all six complementarity-determining regions (CDRs) simultaneously . This approach allowed for the exploration of a vastly greater sequence space than conventional methods. Researchers leveraged the immense display capacity of ribosome display to accommodate the extensive sequence diversity required for these diverse library builds .

Further diversity was introduced through pool maturation, enabling the simultaneous optimization of multiple lead candidates. This strategy led to antibodies with remarkable improvements in both binding properties and inhibition potency . The C0021158 antibody, a product of this affinity maturation process, exhibited a binding affinity of 173 pM, representing an approximately 50-fold improvement compared to its parent antibody, C0020187 . This enhanced affinity translated directly to improved functional performance, with C0021158 fully inhibiting recombinant human ARG2 with an IC50 of 18.5 ± 5.1 nM in IgG format .

The extensive sequence changes resulting from this sophisticated approach manifested as striking structural adaptations in the affinity-matured antibodies when bound to ARG2 . Crystal structures revealed a large reorientation of the binding paratope that facilitated increases in contact surface and shape complementarity to the antigen . Interestingly, some CDRs like VLCDR1 and VHCDR3 remained unchanged during affinity maturation, suggesting critical epitope interactions intolerant to modification, while other CDRs underwent substantial sequence alterations .

What strategies can researchers employ to validate ARG2 antibody specificity and avoid cross-reactivity with ARG1?

Validating ARG2 antibody specificity is a critical step in experimental design, particularly given the structural similarity between ARG2 and its paralog ARG1. Several complementary approaches can be implemented to ensure antibody specificity. Western blotting using lysates from tissues known to differentially express ARG1 and ARG2 serves as an initial validation step. Liver tissue, which predominantly expresses ARG1, should be compared against kidney or small intestine samples that primarily express ARG2 . The antibody should detect the appropriate band size (38-40 kDa for ARG2) in ARG2-expressing tissues while showing minimal cross-reactivity with ARG1.

For more definitive validation, recombinant protein analysis using purified human ARG1 and ARG2 proteins can directly assess cross-reactivity. Techniques such as ELISA or surface plasmon resonance (SPR) can quantitatively measure binding affinities to both proteins, with a highly specific ARG2 antibody demonstrating strong affinity for ARG2 and negligible binding to ARG1 . The C0021158 antibody, for example, showed no detectable binding to human ARG1 when assessed by bio-layer interferometry, confirming its excellent specificity .

Genetic approaches offer another robust validation strategy. Testing antibody reactivity in ARG2 knockout cell lines or tissues provides compelling evidence of specificity. Similarly, siRNA-mediated knockdown of ARG2 should result in reduced antibody signal if the antibody is truly specific for ARG2. For complete validation, researchers can perform ARG2 overexpression experiments in cell lines with low endogenous ARG2 expression and confirm increased antibody signal.

Epitope mapping represents an advanced approach to understanding antibody specificity. Knowing which region of ARG2 the antibody targets helps predict the likelihood of cross-reactivity with ARG1. Antibodies targeting highly conserved regions, particularly near the active site, are more likely to cross-react than those binding to divergent regions outside the substrate binding domain . For instance, the A-10 antibody targets an epitope between amino acids 304-335 near the C-terminus of human ARG2, while the C-8 antibody was raised against amino acids 291-354 of human ARG2 .

How can researchers effectively measure ARG2 enzymatic inhibition by antibodies in experimental systems?

Measuring ARG2 enzymatic inhibition by antibodies requires carefully designed assays that accurately quantify enzyme activity under various conditions. The colorimetric urea assay represents a foundational approach for assessing ARG2 activity. This method quantifies urea production, a direct product of arginine hydrolysis by ARG2 . When implementing this assay, researchers should establish a standard curve using known urea concentrations, ensure linear enzyme kinetics by optimizing enzyme concentrations and reaction times, and include appropriate controls including enzyme-only and antibody-only reactions.

For more sophisticated analyses, researchers can employ isothermal titration calorimetry (ITC) to characterize the thermodynamics of antibody-ARG2 interactions and correlate binding parameters with inhibition potency. This technique provides valuable insights into binding affinity (KD), enthalpy changes (ΔH), and binding stoichiometry, offering a comprehensive biophysical profile of the interaction . When using ITC, careful attention should be paid to buffer matching between samples and titrant solutions to minimize background heat signals.

Cell-based inhibition assays provide crucial information about antibody performance in more complex biological systems. Researchers can measure ARG2 activity in cell lines overexpressing ARG2 in the presence of various antibody concentrations. The IC50 values derived from such dose-response curves serve as important metrics for comparing inhibitory potency across different antibodies . The C0021158 antibody, for instance, demonstrated full inhibition of recombinant human ARG2 with an IC50 of 18.5 ± 5.1 nM in its IgG format .

T-cell proliferation rescue assays represent particularly relevant functional readouts for cancer immunology applications. Since ARG2-mediated depletion of L-arginine impairs T-cell proliferation, measuring the ability of inhibitory antibodies to restore T-cell proliferative capacity provides a direct assessment of functional efficacy . These assays typically co-culture T-cells with ARG2-expressing cells in the presence or absence of inhibitory antibodies, with T-cell proliferation quantified via methods such as CFSE dilution or thymidine incorporation.

What are the key experimental considerations when studying ARG2-mediated immunosuppression in tumor microenvironments?

Studying ARG2-mediated immunosuppression in tumor microenvironments requires thoughtful experimental design that accounts for the complex interplay between cancer cells, immune cells, and metabolic factors. Sample selection and preparation represent crucial initial considerations. Researchers should compare ARG2 expression across different tumor types, focusing on those known to overexpress ARG2 such as pancreatic ductal adenocarcinoma, bowel cancer, and acute myeloid leukemia . Fresh tumor samples are preferable to preserve ARG2 enzymatic activity, though flash-frozen tissues can also be suitable for many analyses.

Comprehensive profiling of ARG2 expression within the tumor microenvironment is essential. Multiplexed immunohistochemistry or immunofluorescence using ARG2 antibodies alongside markers for various cell types can identify which cells within the tumor microenvironment express ARG2 . This approach helps determine whether ARG2 originates primarily from tumor cells or infiltrating immune cells, providing critical contextual information for interpreting experimental results.

L-arginine quantification in the tumor microenvironment serves as a direct measure of ARG2 activity. High-performance liquid chromatography (HPLC) or mass spectrometry can accurately measure L-arginine concentrations in tumor tissue extracts or interstitial fluid . Researchers should correlate these measurements with ARG2 expression levels and T-cell functionality markers to establish relationships between ARG2 activity, arginine depletion, and immune suppression.

Ex vivo functional assays provide valuable insights into ARG2-mediated immune suppression. Tumor-infiltrating lymphocytes (TILs) can be isolated from tumor samples and assessed for proliferation capacity and effector functions in the presence or absence of ARG2 inhibitory antibodies like C0021158 . A successful ARG2 inhibitor should restore T-cell proliferation and enhance cytokine production in conditions where ARG2-mediated arginine depletion would otherwise suppress these functions.

For in vivo studies, syngeneic mouse models with tumors engineered to overexpress ARG2 can be treated with murine versions of ARG2 inhibitory antibodies. Researchers should monitor not only tumor growth but also changes in tumor-infiltrating immune cell populations, particularly T-cell activation status and effector functions. Additionally, measuring intratumoral L-arginine concentrations before and after antibody treatment provides direct evidence of the inhibitor's impact on the metabolic environment.

What emerging applications are being developed for ARG2 antibodies beyond cancer immunotherapy?

While cancer immunotherapy represents a primary focus for ARG2 antibody research, emerging applications are expanding into other disease contexts where ARG2 dysregulation plays a pathological role. Cardiovascular disease research increasingly recognizes ARG2's significance in endothelial dysfunction and atherosclerosis development . ARG2 competes with endothelial nitric oxide synthase (eNOS) for their common substrate L-arginine, thereby reducing nitric oxide production, which is crucial for vascular relaxation and endothelial health . ARG2 inhibitory antibodies could potentially restore proper nitric oxide signaling in vascular tissues, representing a novel therapeutic approach for hypertension and atherosclerosis.

Neurodegenerative disorders present another promising application area. Research has implicated ARG2 dysregulation in neurodegenerative processes through mechanisms involving disrupted arginine metabolism and nitric oxide signaling in neuronal tissues . Antibodies that selectively inhibit ARG2 could help restore metabolic balance in affected neurons and potentially slow disease progression. Additionally, their high specificity would avoid interfering with ARG1, which serves important functions in ammonia detoxification through the urea cycle.

Inflammatory and autoimmune conditions may also benefit from ARG2 antibody interventions. ARG2 expression in myeloid cells can modulate inflammatory responses and contribute to immunosuppressive phenotypes in certain disease contexts . Inhibitory antibodies targeting ARG2 could potentially reprogram these cells toward more immunostimulatory functions, offering novel strategies for treating conditions characterized by inappropriate immune suppression.

Metabolic disorders represent an emerging area where ARG2 antibodies might provide both research insights and therapeutic potential. ARG2's role in arginine metabolism connects to broader metabolic pathways affecting energy balance and cellular homeostasis . Researchers are investigating how ARG2 inhibition might influence metabolic parameters in conditions like obesity and diabetes, with preliminary evidence suggesting beneficial effects on insulin sensitivity and glucose metabolism in preclinical models.

How might combinatorial approaches using ARG2 antibodies with other immunomodulatory agents enhance therapeutic efficacy?

Combinatorial approaches using ARG2 inhibitory antibodies alongside other immunomodulatory agents represent a promising strategy to overcome the multifaceted immune evasion mechanisms employed by tumors. Combining ARG2 antibodies with immune checkpoint inhibitors (ICIs) targeting PD-1, PD-L1, or CTLA-4 could synergistically enhance anti-tumor immunity . While ICIs release intrinsic T-cell inhibitory signals, ARG2 inhibitors address the metabolic constraints imposed by arginine depletion, potentially converting "cold" tumors unresponsive to ICIs into "hot" immunologically responsive tumors through complementary mechanisms.

ARG2 antibodies might also be effectively paired with adoptive cell therapies such as CAR-T cells or TIL therapy . The immunosuppressive tumor microenvironment, partly maintained through ARG2-mediated arginine depletion, often limits the efficacy of these cellular therapies. Administering ARG2 inhibitory antibodies could create a more favorable metabolic environment for these engineered or expanded T cells, enhancing their persistence, proliferation, and effector functions within the tumor.

Combinations with cancer vaccines represent another rational approach. Cancer vaccines aim to prime T-cell responses against tumor antigens, but these responses may be undermined by the immunosuppressive mechanisms active within tumors, including ARG2-mediated arginine depletion . Administering ARG2 inhibitory antibodies alongside cancer vaccines could ensure that vaccine-induced T cells retain their functionality when encountering the tumor, potentially improving clinical outcomes.

Targeting multiple metabolic immune checkpoints simultaneously may yield particularly powerful synergies. Beyond ARG2, tumors exploit other metabolic mechanisms to suppress T-cell functions, including indoleamine 2,3-dioxygenase (IDO), CD73/adenosine signaling, and lactate production . Combinations of inhibitors targeting these multiple metabolic pathways could comprehensively remodel the tumor microenvironment to support robust anti-tumor immunity. Preliminary research suggests that such combinatorial approaches might overcome resistance mechanisms that develop against single-agent therapies.

What novel structural modifications to ARG2 antibodies might enhance their research and therapeutic potential?

Novel structural modifications to ARG2 antibodies present exciting opportunities to enhance their research applications and therapeutic potential. Antibody fragments, including Fab, F(ab')2, and single-chain variable fragments (scFvs), offer advantages in certain research contexts due to their smaller size, potentially enabling better tissue penetration and reduced immunogenicity . These fragments might access epitopes on ARG2 that are sterically hindered when using full-size antibodies, potentially revealing new inhibitory mechanisms. The crystal structures of ARG2-Fab complexes have already provided valuable insights into inhibition mechanisms, and further structural variants could expand this knowledge base .

Bispecific antibodies that simultaneously target ARG2 and another relevant target represent an innovative approach for enhancing functionality. For instance, a bispecific antibody targeting both ARG2 and a tumor-associated antigen could localize ARG2 inhibition specifically to the tumor microenvironment, potentially reducing off-target effects . Alternatively, bispecific constructs targeting ARG2 and immune cell receptors like CD3 could directly bring T cells into proximity with ARG2-expressing cells, combining ARG2 inhibition with immune cell recruitment.

Antibody-drug conjugates (ADCs) leveraging ARG2 antibodies as targeting moieties could deliver cytotoxic payloads specifically to ARG2-overexpressing cells within tumors. This approach would transform ARG2 antibodies from purely inhibitory agents to targeted delivery vehicles, potentially expanding their therapeutic applications. The high specificity of antibodies like C0021158 for ARG2 makes them excellent candidates for such applications, as they would minimize off-target delivery .

Engineered antibodies with enhanced tissue penetration properties could address the challenge of accessing ARG2 within solid tumors. Modifications such as deglycosylation or specific framework region alterations that reduce antibody size while maintaining specificity and affinity could improve biodistribution profiles . Additionally, pH-sensitive antibodies designed to release from their target under the acidic conditions found in tumors could enhance tissue penetration through a "bind, release, and diffuse" mechanism.

Product Science Overview

Introduction

Arginase Type II, also known as arginase 2 or ARG2, is an enzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. This enzyme is one of the two isoforms of arginase found in mammals, with the other being arginase type I. Arginase type II is primarily located in the mitochondria and is expressed in extra-hepatic tissues, especially the kidney .

Structure and Function

Arginase type II is encoded by the ARG2 gene. Unlike arginase type I, which is predominantly found in the liver, arginase type II is distributed in various tissues, including the kidney, brain, and prostate . The enzyme plays a crucial role in the urea cycle, which is essential for the detoxification of ammonia in the body. Additionally, arginase type II is involved in the regulation of nitric oxide and polyamine metabolism .

Physiological Role

The physiological role of arginase type II is not as well understood as that of arginase type I. However, it is believed to be involved in several important biological processes. For instance, arginase type II has been shown to promote macrophage inflammatory responses through mitochondrial reactive oxygen species, contributing to insulin resistance and atherogenesis . It is also implicated in the regulation of immune responses and cellular senescence .

Mouse Anti Human Arginase Type II Antibodies

Mouse anti human arginase type II antibodies are monoclonal antibodies developed to specifically target and bind to human arginase type II. These antibodies are widely used in various scientific applications, including Western blotting, immunohistochemistry, immunocytochemistry, enzyme-linked immunosorbent assay (ELISA), and flow cytometry . They are valuable tools for researchers studying the expression, localization, and function of arginase type II in different tissues and under various physiological and pathological conditions.

Applications in Research

The use of mouse anti human arginase type II antibodies has facilitated numerous studies aimed at understanding the role of arginase type II in health and disease. For example, research has shown that silencing arginase type II in human monocytes decreases their adhesion to endothelial cells and reduces the production of proinflammatory mediators . Additionally, studies using arginase type II-deficient mice have demonstrated the enzyme’s involvement in macrophage inflammation, glucose metabolism, and atherosclerosis .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.