ARG2 Human

Arginase Type II Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ARG2 Human

ARG2 Human, also known as Arginase 2, is an enzyme encoded by the ARG2 gene in humans. It is a mitochondrial enzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea, playing a crucial role in various physiological and pathological processes . Unlike Arginase 1, which is primarily expressed in the liver, Arginase 2 is found in extra-hepatic tissues such as the kidney, brain, and immune cells .

Structure and Function

Arginase 2 is a binuclear manganese-dependent metalloenzyme with a molecular weight of approximately 36 kDa. It forms a trimeric active molecule, similar to Arginase 1, despite being located in different cellular compartments . The enzyme's structure has been elucidated through crystallography, revealing insights into its catalytic mechanism .

CharacteristicsDescription
Enzyme TypeBinuclear manganese metalloenzyme
Molecular WeightApproximately 36 kDa
Subcellular LocationMitochondria
Tissue DistributionKidney, brain, immune cells, prostate

Physiological Roles

Arginase 2 is involved in several physiological processes, including nitric oxide and polyamine metabolism. Its role in regulating nitric oxide synthesis is significant, as it affects vascular health and sexual arousal . Additionally, Arginase 2 influences inflammatory responses, particularly in lung and bronchial epithelial diseases .

Intervertebral Disc Degeneration (IDD)

Recent studies have shown that Arginase 2 plays a role in intervertebral disc degeneration. Overexpression of Arginase 2 promotes apoptosis, inflammation, and extracellular matrix degradation in nucleus pulposus cells, suggesting it as a potential therapeutic target for IDD .

Metabolic Disorders

Hepatic overexpression of Arginase 2 has been linked to reduced fat accumulation and improved metabolic profiles in diabetic models. This suggests a protective role against hepatic and peripheral fat accumulation .

Immune Response

Arginase 2-specific T cells have been identified, which can recognize and react to activated Tregs and cancer cells expressing Arginase 2. This indicates potential applications in cancer immunotherapy .

Research Findings

Study FocusKey Findings
IDDARG2 overexpression exacerbates ECM degradation and apoptosis in NPCs .
Metabolic DisordersHepatic ARG2 overexpression reduces fat accumulation and improves metabolic health .
Immune ResponseARG2-specific T cells can target cancer cells and activated Tregs .

Product Specs

Introduction
ARG2 is involved in regulating arginine metabolism outside the urea cycle and suppressing nitric oxide synthesis. By regulating L-arginine availability to NO synthase in extrahepatic tissues, ARG2 plays a role in male and female sexual arousal. This makes it a potential target for treating sexual arousal disorders in both sexes, given NO synthase's presence in penile and clitoral corpus cavernosum smooth muscle and the vagina.
Description
Recombinant ARG2, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 353 amino acids (residues 23-354) and has a molecular weight of 38.3 kDa. A 21 amino acid His-Tag is fused to the N-terminus of the ARG2 protein. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The ARG2 human solution is supplied in a buffer of 20mM Tris-HCl at pH 8.0 with 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
SDS-PAGE analysis confirms a purity exceeding 95.0%.
Synonyms
Kidney Arginase, Non-Hepatic Arginase, EC 3.5.3.1, Arginase-2, Type II arginase, Kidney-type arginase, Arginase-2 mitochondrial.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MVHSVAVIGA PFSQGQKRKG VEHGPAAIRE AGLMKRLSSL GCHLKDFGDL SFTPVPKDDL YNNLIVNPRS VGLANQELAE VVSRAVSDGY SCVTLGGDHS LAIGTISGHA RHCPDLCVVW VDAHADINTP LTTSSGNLHG QPVSFLLREL QDKVPQLPGF SWIKPCISSA SIVYIGLRDV DPPEHFILKN YDIQYFSMRD IDRLGIQKVM ERTFDLLIGK RQRPIHLSFD IDAFDPTLAP ATFTPVVGGL TYREGMYIAE EIHNTGLLSA LDLVEVNPQL ATSEEEAKTT ANLAVDVIAS SFGQTREGGH IVYDQLPTPS SPDESENQAR VRI.

Q&A

What is the basic structure and enzymatic function of human ARG2?

Human Arginase 2 (ARG2) is a binuclear manganese-dependent metalloenzyme with a molecular weight of approximately 36-40 kDa. It forms a trimeric active molecule that catalyzes the conversion of L-arginine into L-ornithine and urea. The functional protein spans amino acids Val23-Ile354 (Accession # P78540) and contains critical manganese ions in its active site that are essential for its catalytic activity . ARG2 differs from its isoform ARG1, sharing only 58% sequence identity despite catalyzing the same reaction. The trimeric quaternary structure is critical for enzyme function, and structural analysis reveals specific binding sites for manganese ions that coordinate the hydrolysis reaction .

How does ARG2 differ from ARG1 in terms of cellular localization and function?

While ARG1 is primarily cytosolic and highly expressed in the liver as part of the urea cycle, ARG2 is predominantly localized in the mitochondria and is expressed in various tissues including kidneys, brain, small intestine, and immune cells. This mitochondrial localization allows ARG2 to function in arginine metabolism independently of the urea cycle. The distinct tissue distribution suggests ARG2 plays specific roles in local arginine metabolism and regulation of L-arginine availability within specific cellular microenvironments. ARG2's unique expression pattern makes it particularly relevant in contexts such as tumor microenvironments and inflammatory conditions, where it can contribute to immunosuppression through localized arginine depletion .

What are the key post-translational modifications of ARG2 that affect its activity?

Several post-translational modifications regulate ARG2 activity and stability. Phosphorylation at specific serine and threonine residues can modulate enzyme activity, while S-nitrosylation can inhibit catalytic function. The enzyme's activity is also dependent on the availability of manganese ions, which must be incorporated during protein folding for optimal enzymatic function. Additionally, proper formation of the trimeric structure is essential for maintaining ARG2 in its most active conformation. Researchers studying ARG2 should consider these modifications when designing experiments, as cellular conditions that affect these modifications may significantly impact experimental outcomes and interpretation of results.

What are the best methods for detecting ARG2 expression in tissue samples and cell lines?

Multiple complementary approaches should be employed for robust ARG2 detection. Western blotting using specific antibodies such as Anti-Human ARG2 Monoclonal Antibody (e.g., MAB10602) can effectively detect ARG2 at approximately 40 kDa under reducing conditions . For immunohistochemistry, specific protocols for fixation and antigen retrieval are crucial for accurate detection in tissue samples. qRT-PCR can be used to quantify ARG2 mRNA levels, providing insight into transcriptional regulation. The choice between these methods should be guided by the specific research question—protein detection methods are preferable for assessing functional enzyme levels, while mRNA quantification is useful for studying transcriptional regulation. When analyzing human samples, researchers should account for potential variations in ARG2 expression related to pathological conditions .

How can ARG2 enzymatic activity be accurately measured in experimental settings?

ARG2 enzymatic activity can be measured through several approaches. The standard method involves quantifying urea production via colorimetric assays, where L-arginine is converted to L-ornithine and urea. Alternatively, researchers can measure the consumption of L-arginine or production of L-ornithine using HPLC or mass spectrometry for more precise quantification. When conducting these assays, it's critical to optimize reaction conditions, including pH (typically 9.0-9.5), temperature (37°C), and manganese concentration (1-5 mM MnCl₂) to ensure maximum activity. Sample preparation must include careful consideration of potential inhibitors present in the biological sample. For specific inhibition studies, researchers can employ competitive inhibitors like N^ω^-hydroxy-L-arginine or antibody-based inhibitors like C0021158 that act through non-competitive mechanisms .

What techniques are available for modulating ARG2 expression or activity in experimental models?

Several approaches can be employed for modulating ARG2 in experimental systems. For gene silencing, siRNA transfection has proven effective, with researchers selecting the siRNA with highest silencing efficiency through preliminary validation . For stable knockdown, shRNA constructs or CRISPR-Cas9 gene editing can generate long-term ARG2-deficient models. Overexpression can be achieved using ARG2 overexpression plasmids with appropriate promoters for the target cell type . For pharmacological inhibition, specific ARG2 antibodies like C0021158 offer complete enzymatic inhibition through non-competitive mechanisms . Small-molecule inhibitors should be evaluated for ARG2 vs. ARG1 selectivity. When designing these experiments, it's essential to include appropriate controls and validate the modulation efficiency at both mRNA and protein levels. The experimental timeline should consider the stability of the intervention and potential cellular adaptation responses.

How does ARG2 contribute to immunosuppression in tumor microenvironments?

ARG2 creates immunosuppressive conditions in tumor microenvironments through several mechanisms. Primarily, increased ARG2 expression depletes local L-arginine levels, which directly impairs T-cell proliferation, cytokine production, and receptor expression . This arginine depletion leads to downregulation of the CD3ζ chain of the T-cell receptor complex, reducing T-cell responsiveness to tumor antigens. Additionally, the byproducts of ARG2 activity contribute to an immunosuppressive milieu by promoting myeloid-derived suppressor cell (MDSC) functions. The net effect renders tumors effectively "invisible" to the host's immune system . Experimentally, this can be demonstrated by co-culturing ARG2-expressing cells with T-cells and measuring proliferation markers, cytokine production, and activation markers. Inhibition studies using ARG2-specific antibodies like C0021158 provide valuable insights, as they can restore T-cell proliferation in vitro by blocking ARG2's enzymatic function .

What is the relationship between ARG2 expression and intervertebral disc degeneration (IDD)?

ARG2 plays a significant role in intervertebral disc degeneration through multiple pathological mechanisms. Studies have demonstrated that ARG2 expression is significantly elevated in degenerated intervertebral discs compared to normal discs, with a positive correlation between expression levels and degeneration severity . Mechanistically, ARG2 activates the NF-κB pathway, which subsequently increases expression of matrix-degrading enzymes like MMP3 and MMP13 while decreasing production of critical extracellular matrix components such as type II collagen and aggrecan . ARG2 also promotes nucleus pulposus cell (NPC) senescence, apoptosis, oxidative stress, and inflammatory responses. Experimental evidence shows that ARG2 silencing using siRNA prevents these detrimental effects and ameliorates ECM degradation in NPCs challenged with IL-1β. Conversely, ARG2 overexpression exacerbates these pathological processes, confirming its mechanistic role in IDD pathogenesis .

How does ARG2 interact with the NF-κB pathway in inflammatory conditions?

ARG2 exhibits a complex and bidirectional relationship with the NF-κB pathway in inflammatory settings. Research indicates that ARG2 activates NF-κB signaling, which plays a critical role in inflammatory responses, senescence, and stress reactions . When ARG2 is overexpressed, significant activation of NF-κB occurs, leading to upregulation of pro-inflammatory cytokines (TNFα, IL-6) and matrix-degrading enzymes . Conversely, silencing ARG2 with siRNA inhibits NF-κB activation, reducing inflammatory markers and protecting against tissue degradation. Experimental data from nucleus pulposus cells shows that ARG2 silencing significantly reduces NF-κB nuclear translocation and phosphorylation of p65, a key component of the NF-κB complex. The molecular mechanism likely involves ARG2-mediated alterations in cellular redox status and mitochondrial function, which subsequently affects IκB kinase activity and NF-κB nuclear translocation .

What mechanisms explain the non-competitive inhibition of ARG2 by antibodies like C0021158?

The non-competitive inhibition of ARG2 by antibodies like C0021158 involves sophisticated allosteric mechanisms. Structural analysis through co-crystallization of the C0021158 Fab with trimeric ARG2 revealed that the antibody binds at a site distant from the enzyme's substrate binding cleft . Upon binding, C0021158 induces substantial conformational changes in ARG2, including complete rearrangement of a surface-exposed loop and formation of a new short helix structure at the Fab-ARG2 interface. These structural alterations propagate to the enzyme's active site, where Arg39 reorients inward, creating steric hindrance that impedes L-arginine binding . Additionally, this conformational change likely alters the pKₐ of the catalytic histidine residue at position 160, further attenuating enzymatic function. Isothermal calorimetry experiments with an L-arginine mimetic confirm that substrate binding is significantly impaired when the antibody is bound. This mechanism explains how C0021158 inhibits ARG2 independently of L-arginine concentrations, maintaining effectiveness even in high-substrate environments .

How can researchers address the challenge of distinguishing between ARG1 and ARG2 effects in complex biological systems?

Distinguishing between ARG1 and ARG2 effects requires a multi-faceted approach. First, researchers should employ isoform-specific antibodies and primers that target unique regions of each protein. The use of highly specific inhibitors or neutralizing antibodies like C0021158, which shows selectivity for ARG2, can help differentiate functional contributions . In genetic approaches, selective knockdown using validated siRNAs targeting unique mRNA regions can provide isoform-specific suppression . For tissue-specific studies, researchers should consider the predominant distribution patterns—ARG1 in hepatocytes and ARG2 in renal tissues, vascular endothelium, and specific immune cell populations. In cellular models, compartmentalization studies can help distinguish mitochondrial ARG2 from cytosolic ARG1. When analyzing complex tissues, single-cell approaches or cell sorting followed by isoform-specific detection can resolve cell-specific expression patterns. Finally, considering differential regulation of the two isoforms under various stimuli can provide additional experimental separation strategies.

What are the methodological considerations for studying ARG2 in the context of hypoxic microenvironments?

Studying ARG2 in hypoxic conditions presents unique methodological challenges. Researchers must establish reliable hypoxic conditions, ideally using controlled oxygen chambers that maintain stable O₂ levels (typically 0.5-2%) while allowing sample manipulation. HIF-1α stabilization should be verified as a hypoxia marker. Since ARG2 is mitochondrially localized, mitochondrial function assessments under hypoxia are crucial, measuring parameters like membrane potential and ROS production. Protein extraction protocols must be modified to protect oxygen-sensitive post-translational modifications, conducting procedures under nitrogen atmosphere when possible. When measuring enzymatic activity, researchers should account for pH changes under hypoxia and standardize reaction conditions. The interplay between ARG2 and nitric oxide synthase pathways becomes particularly important under hypoxia, necessitating simultaneous measurement of both pathways. Time-course studies are essential as hypoxic regulation of ARG2 may involve both acute and chronic adaptive responses with distinct mechanisms and functional consequences.

What experimental controls are essential when studying ARG2 inhibition strategies?

When investigating ARG2 inhibition strategies, several critical controls must be implemented for rigorous experimental design. For antibody-based inhibitors like C0021158, appropriate isotype controls are essential to rule out non-specific effects . When using siRNA approaches, non-targeting siRNA controls with similar chemical modifications must be included, and researchers should validate knockdown efficiency at both mRNA and protein levels . Rescue experiments through ARG2 overexpression can confirm the specificity of observed phenotypes. For functional readouts, parallel assessment of ARG1 activity helps exclude compensatory mechanisms. When studying downstream effects like T-cell function or NF-κB activation, pathway-specific positive and negative controls (e.g., NF-κB inhibitors) help establish causality. Time-course experiments are crucial, as inhibition efficacy may vary temporally. Finally, concentration-response studies should be conducted to establish optimal inhibitor concentrations, with parallel cytotoxicity assessments to exclude non-specific effects.

How should researchers design experiments to study ARG2's role in cellular senescence and apoptosis?

Studying ARG2's role in cellular senescence and apoptosis requires careful experimental design. For senescence assessment, researchers should use multiple complementary markers including SA-β-galactosidase activity, p21 and p16 expression levels , senescence-associated heterochromatin foci (SAHF), and the senescence-associated secretory phenotype (SASP) profile. Time-course experiments are essential as senescence develops progressively. For apoptosis studies, researchers should combine multiple detection methods including Annexin V/PI staining, TUNEL assays, and assessment of key apoptotic proteins like Bcl2, Bax, and cleaved caspases . In both contexts, genetic manipulation of ARG2 through siRNA knockdown or overexpression provides mechanistic insights . Researchers should include physiologically relevant stressors that mimic disease conditions, such as IL-1β treatment to simulate inflammatory environments . Rescue experiments using L-arginine supplementation can determine whether ARG2's effects are mediated through arginine depletion or alternative mechanisms. Finally, pathway inhibition studies targeting NF-κB can elucidate the relationship between ARG2 and these cellular processes.

What are the key considerations for translating ARG2 research findings to potential therapeutic applications?

Translating ARG2 research to therapeutics requires addressing several critical considerations. First, isoform selectivity is paramount—therapeutic agents must specifically target ARG2 over ARG1 to avoid disrupting hepatic urea cycle function. Antibody-based approaches like C0021158 offer promising selectivity . Second, cellular penetration and subcellular targeting present challenges due to ARG2's mitochondrial localization, necessitating delivery systems capable of reaching this compartment. Third, researchers must establish precise therapeutic windows, as complete ARG2 inhibition may disrupt normal arginine metabolism in non-target tissues. Disease-specific considerations are crucial: in cancer immunotherapy, ARG2 inhibition should be evaluated in combination with existing immune checkpoint inhibitors; in inflammatory conditions like IDD, researchers should assess both preventative and therapeutic efficacy . The potential for compensatory mechanisms, including ARG1 upregulation or alternative arginine metabolic pathways, must be thoroughly investigated. Finally, translational studies should include appropriate biomarkers to monitor target engagement and functional outcomes, such as arginine/ornithine ratios in accessible biofluids or T-cell function parameters in immunomodulatory applications.

Product Science Overview

Introduction

Arginase Type II (ARG2) is one of the two isoforms of the enzyme arginase, which plays a crucial role in the urea cycle by catalyzing the hydrolysis of L-arginine into L-ornithine and urea. While Arginase Type I is predominantly found in the liver, Arginase Type II is primarily located in extra-hepatic tissues, such as the kidney, and is localized in the mitochondria .

Structure and Function

Arginase Type II is a binuclear manganese-dependent metalloenzyme. It forms a trimeric active molecule with a molecular mass of approximately 36 kDa . The enzyme’s primary function is to regulate the levels of L-arginine, which is a precursor for the synthesis of nitric oxide, polyamines, proline, glutamate, creatine, and agmatine . By converting L-arginine into L-ornithine and urea, Arginase Type II plays a significant role in cellular metabolism and the regulation of nitric oxide synthesis .

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 critical biological processes, including nitric oxide and polyamine metabolism . Nitric oxide is a vital signaling molecule involved in various physiological processes, such as vasodilation, neurotransmission, and immune response. Polyamines are essential for cell growth and differentiation .

Clinical Significance

Arginase Type II has been implicated in various pathological conditions. For instance, it has been associated with the regulation of immune responses and inflammation. Studies have shown that Arginase Type II can influence macrophage function and is linked to chronic inflammation, insulin resistance, and atherosclerosis . Additionally, the enzyme’s role in nitric oxide metabolism suggests its potential involvement in cardiovascular diseases and other conditions related to nitric oxide dysregulation .

Recombinant Human Arginase Type II

Recombinant human Arginase Type II is produced using E. coli expression systems. The recombinant protein typically includes an N-terminal Met and a 6-His tag for purification purposes . The specific activity of the recombinant enzyme is measured by the production of urea during the hydrolysis of arginine, with a specific activity greater than 60,000 pmol/min/μg . The recombinant protein is supplied as a 0.2 μm filtered solution in HEPES and NaCl and is stable for up to six months when stored at -20 to -70 °C .

Applications

Recombinant human Arginase Type II is used in various research applications, including studies on amino acid metabolism, nitric oxide synthesis, and polyamine metabolism. It is also utilized in investigating the enzyme’s role in pathological conditions such as cancer, cardiovascular diseases, and immune responses .

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.