MDSC and their subsets were isolated from spleens of tumor-bearing or control mice using cell sorting on FACSAria cell sorter (Becton Dickinson). The purity of cell populations was >99%. As responder cells we used total spleen cells from OT-1 mice. CD8+ T cells from these mice have a TCR that recognize OVA-derived peptide SIINFEKL. The number of IFN-γ producing cells in response to stimulation to the specific or control peptides (10 μg/ml) was evaluated in an ELISPOT assay and performed as described earlier (28 (link)). The numbers of spots were counted in triplicates and calculated using an automatic ELISPOT counter (Cellular Technology, Ltd). Cell proliferation induced by antigen specific or CD3 (0.5 μg/ml)/CD28 (5 μg/ml) stimulation was evaluated using 3H-thymidine incorporation as described previously (29 (link)). For experiments which examined the effect of NO, arginase, or ROS inhibitor, L-NMMA (0.5 mM; Calbiochem, San Diego, CA), nor-NOHA (0.5 mM; Calbiochem, San Diego, CA), or catalase (1000 U/mL; Sigma-Aldrich) were added at the beginning of the culture.
>
Chemicals & Drugs
>
Amino Acid
>
Arginase
Arginase
Arginase is an enzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea.
It plays a key role in the urea cycle, as well as in regulating nitric oxide production and cell proliferation.
Arginase has been implicated in various pathological conditions, including inflammation, cancer, and neurological disorders.
Understanding the functions and regulation of arginase is crucial for developing effective therapies targeting these diseases.
PubCompare.ai's AI-powered platform can enhance your arginase research by helping you easily locate relevant protocols from literature, preprints, and patents, while its AI-driven comparisons identify the best protocols and products to improve reproducibility and accuracy in your arginase studies.
It plays a key role in the urea cycle, as well as in regulating nitric oxide production and cell proliferation.
Arginase has been implicated in various pathological conditions, including inflammation, cancer, and neurological disorders.
Understanding the functions and regulation of arginase is crucial for developing effective therapies targeting these diseases.
PubCompare.ai's AI-powered platform can enhance your arginase research by helping you easily locate relevant protocols from literature, preprints, and patents, while its AI-driven comparisons identify the best protocols and products to improve reproducibility and accuracy in your arginase studies.
Most cited protocols related to «Arginase»
Antigens
Arginase
Biological Assay
Catalase
CD8-Positive T-Lymphocytes
Cell Proliferation
Cells
Enzyme-Linked Immunospot Assay
Exanthema
Interferon Type II
Mus
Myeloid-Derived Suppressor Cells
omega-N-Methylarginine
Peptides
Population Group
Spleen
Splenic Neoplasms
Thymidine
J774 cells (1x106/ml) or peritoneal cells (1x106/ml) were treated as previously described above. Arginase activity was measured in cell lysates as described previously 16 (link), 39 (link). Briefly, cells were lysed with 50 µl of 0.1% Triton X-100 containing protease inhibitors. This mixture was stirred for 30 min and then 50 μl of 10 mM MnCl2 with 50 mM Tris-HCl we added to activate the enzyme for 10 min at 56 ◦C. Arginine hydrolysis was initiated by the addition of 25 µl of 0.5 M L-arginine, pH 9.7, at 37 ◦C for 45 min. The reaction was stopped with a mixture of acids, and the urea concentration was measured at 540 nm after the addition of 25 μl of α-isonitrosopropiophenone (dissolved in 100% ethanol) followed by heating at 95 ◦C for 45 min. The results are expressed as Arginase Index (fold increase of arginase activity in samples above basal).
Acids
Arginase
Arginine
Cells
Enzymes
Ethanol
Hydrolysis
manganese chloride
Peritoneum
SERPINA1 protein, human
Triton X-100
Tromethamine
Urea
Hemoglobin and white blood cell counts were measured by a counter (T890; Beckman Coulter), and routine biochemistry, acid-base parameters, and lactate were measured using a bedside biochemical analyzer (i-STAT-1; i-STAT Corp.). Parasite counts were determined by Giemsa-stained thick and thin fields and were cross-checked by an experienced microscopist. Plasma was separated within 30 min of collection by centrifugation and stored at −70°C. Amino acids were extracted from 50 μl of plasma after the addition of 50 μl of internal standard (norleucine) and 200 μl of cold ethanol. Deproteinized plasma was derivitized with AccQFluor reagent (Waters), and amino acids were measured by HPLC (Shimadzu) using a method modified from van Wandelen and Cohen (55 ). Plasma concentrations of the endothelial activation markers soluble ICAM-1 and E-selectin were assayed by ELISA (R&D Systems). To quantitate total parasite biomass, plasma HRP2 was measured by ELISA, as previously described (56 (link)). Purified HRP2 was provided by D. Sullivan (Johns Hopkins University, Baltimore, MD). Plasma haptoglobin and LDH were measure by ELISA and a calorimetric assay, respectively (Roche Diagnostics). Plasma arginase activity was measured using a radiometric assay, as previously described, and reported as micromole/milliliter/hour (16 (link)).
Acids
Amino Acids
Arginase
Biological Assay
Calorimetry
Centrifugation
Cold Temperature
Diagnosis
Endothelium
Enzyme-Linked Immunosorbent Assay
Ethanol
Haptoglobins
Hemoglobin
High-Performance Liquid Chromatographies
Intercellular Adhesion Molecule-1
Lactates
Leukocyte Count
Norleucine
Parasites
Plasma
Radiometry
SELE protein, human
Human PBMCs were isolated from heparinised venous blood samples by density gradient centrifugation method using Histopaque (Sigma). Briefly, the heparinised blood was layered on LSM medium gently in the ratio of 1∶1 and subjected to centrifugation at 100 g for 30 minutes. The white layer representing PBMCs was aspirated out gently and transferred aseptically into sterile centrifuge tubes. The suspension of cells was then washed and cultured in sterile DMEM supplemented with 20 mM of L-Glutamine (ICN), 10% of autologus plasma/FBS and antibiotics (1 ml penicillin and streptomycin/100 ml of medium) (Sigma).The no. of cells was adjusted 0.5×106 cells/well in 24 well plates. After 8–10 hrs incubation at 37°C non adherent cells were removed by flushing with sterile DMEM and the adherent cells were stimulated with LPS, FAg or chtx, LPS along with FAg and LPS along with chtx at 10 µg/ml concentration for 48 hrs. after which the supernatants were removed and used for cytokine estimation. The adherent cells were removed and analyzed for intracellular Arginase activity by calorimetric assay as described below.
For study of recycling of the receptor, the PBMCs were resuspended in DMEM containing 0.1% BSA and incubated at 37°C for 4 hrs. Then cells were incubated with biotinylated FAg at 4°C for 30 minutes followed by staining with streptavidin-FITC and analyzed by FACS.
For study of recycling of the receptor, the PBMCs were resuspended in DMEM containing 0.1% BSA and incubated at 37°C for 4 hrs. Then cells were incubated with biotinylated FAg at 4°C for 30 minutes followed by staining with streptavidin-FITC and analyzed by FACS.
Full text: Click here
Antibiotics
Arginase
Biological Assay
BLOOD
Calorimetry
Cells
Centrifugation
Centrifugation, Density Gradient
Charybdotoxin
Cytokine
Fluorescein-5-isothiocyanate
Glutamine
histopaque
Homo sapiens
Penicillins
Plasma
Protoplasm
Sterility, Reproductive
Streptavidin
Streptomycin
Veins
Experiments were conducted in male D2.B6-Ins2Akita/MatbJ and their wild-type littermate mice (Dilute Brown Agouti [DBA]/2J background; The Jackson Laboratory, stock number 007562) starting at 5 weeks of age (2 weeks of diabetes) until 14 weeks of age (11 weeks of diabetes). Ins2Akita mice, recommended by the Animal Models of Diabetes Complications Consortium as an optimal model of DN (18 (link),19 (link)), developed hyperglycemia at 3 weeks of age. Additional experiments were conducted in male 6-week-old DBA/2J mice (The Jackson Laboratory, stock number 000671) and in male arginase-2–deficient (Arg2−/−) mice on C57BL/6J background (provided by B. Lee, Baylor College of Medicine, Houston, TX) using multiple low doses of vehicle (lactated Ringer’s solution) or STZ (Sigma, St. Louis, MO; 50 mg/kg body wt dissolved in lactated Ringer’s solution) via intraperitoneal injection. All animal studies were approved by the Penn State University College of Medicine Institutional Animal Care and Use Committee.
Animal Model
Animals
Arginase
Complications of Diabetes Mellitus
Cuniculus
Diabetes Mellitus
Human Body
Hyperglycemia
Injections, Intraperitoneal
Institutional Animal Care and Use Committees
Lactated Ringer's Solution
Males
Mice, House
Mice, Inbred C57BL
Pharmaceutical Preparations
Technique, Dilution
Most recents protocols related to «Arginase»
An Asp release assay (36 (link)) was used to detect Asp release from β-Asp-X dipeptides: Each 100 µL reaction contained 100 mM HEPES pH 8.2, 20 mM KCl, 5 mM α-ketoglutarate, 500 nM purified enzyme, 0.3 U malate dehydrogenase, 1 mM NADH, 2.4 U aspartate aminotransferase, and 1 mM dipeptide substrate. For other Arg-containing substrates, Arg release was monitored by using a free Arg detection kit (K-LARGE, Neogen, USA): The 135 µL reactions contained 15 µL buffer solution, 10 µL NADPH solution, 1 µL GIDH suspension, 2.5 µL urease solution, 1 µL arginase suspension, 1 mM substrate, and 500 nM enzyme. For both assays, reaction progression was monitored by following 340 nm transmittance in 96-well plates. Data were collected using a SpectraMax Paradigm (Molecular Devices, USA) and analyzed using GraphPad Prism (GraphPad, USA). β-Asp-Arg dipeptides were purified as previously described (26 (link)). β-Asp-Ala and α-Asp-Arg were purchased from Bachem (Switzerland), N2-acetyl arginine, β-Asp-Lys, and β-Asp-Leu from Toronto Research Chemicals (Canada), β-Asp-Asp from AchemBlock (USA), and N2-succinyl arginine from BLD Pharmatech (USA).
acetyl arginine
alpha-Ketoglutaric Acid
Arginase
Arginine
Aspartate Transaminase
aspartyl-aspartic acid
aspartyllysine
Biological Assay
Buffers
Dipeptides
Disease Progression
Enzymes
HEPES
Malate Dehydrogenase
Medical Devices
NADH
NADP
prisma
Urease
The crystallographic structure of Plasmodium falciparum arginase in the liganded complex with co-crystalized ABH (2(S)-amino-6-boronohexanoic acid) and determined using X-ray diffraction at 2.14 Å resolution was downloaded from the PDBe repository (PDBe code: 3mmr). Apart from six crystalic water molecules in the active site AC3 and AC4 all remaining heteroatoms (including ABH molecule) were eradicated prior docking in AutoDock Vina 1.2.0 program. Initially, the ligand/enzyme structures were prepared in the pdbqt file format with the calculated Gasteiger charges. The grid box (size 15 × 15 × 15 Å) was centered on the central atom of ABH analogue. In AutoDock Vina, docking simulations different poses (default nine) were generated progressively from a single conformer (an energy-optimized molecule). The resulting molecular conformations and orientations with the preferred torsion angles and the rotatable bonds were then evaluated by the united-atom (UA) scoring function. Schrödinger Maestro graphical viewers and Protein-Ligand Interaction Profiler (PLIP) were employed to illustrate the foreseen 2D/3D binding modes, respectively.
Full text: Click here
Amino Acids
Arginase
Crystallography
Enzymes
Ligands
Mental Orientation
Plasmodium falciparum
Proteins
X-Ray Diffraction
Arginase activity was determined in aortic tissue homogenates using the Arginase Activity Assay Kit (MAK112-1KT, Sigma-Aldrich).
Aorta segments isolated in PBS solution during the sacrifice were cleaned with saline (0.9% NaCl) prepared using sterile water with DEPC, under an illuminated dissecting loupe (Wild M3C, Leica) in cold light. This procedure was followed by the lysis of the cells with a prepared mixture (containing 100 µL of Tris-HCL 10 mM (pH 7.4), 1 µL of protease inhibitor (pepstatin and leupeptin) and 1 µL Triton X-100 0.4% w/v), and its homogenization using autoclaved tubes and a T 10 basic Ultra-Turrax (IKA Processing Equipment).
The quantification of arginase activity was based on the reaction of the enzyme present in the samples and the substrate buffer (8 µL of arginine buffer and 2 µL of Mn solution), to form urea and ornithine as products, after two hours of incubation at 37 °C. The urea reagent (100 µL of reagent A and 100 µL of reagent B) was added to the obtained urea, giving a colored compound, followed by a room temperature incubation of one hour, stopping the reaction. A control (blank) reaction was run in parallel, containing the same amount of sample, urea reagent and substrate buffer. The amount of colored compound was read at 430 nm in a spectrophotometer (Spectramax plus 384, Molecular Devices, San Jose, CA, USA). The data obtained was exported into a computer program (Softmax pro 6.2.2, Molecular Devices) connected to the device, and then copied into an Excel document.
Aorta segments isolated in PBS solution during the sacrifice were cleaned with saline (0.9% NaCl) prepared using sterile water with DEPC, under an illuminated dissecting loupe (Wild M3C, Leica) in cold light. This procedure was followed by the lysis of the cells with a prepared mixture (containing 100 µL of Tris-HCL 10 mM (pH 7.4), 1 µL of protease inhibitor (pepstatin and leupeptin) and 1 µL Triton X-100 0.4% w/v), and its homogenization using autoclaved tubes and a T 10 basic Ultra-Turrax (IKA Processing Equipment).
The quantification of arginase activity was based on the reaction of the enzyme present in the samples and the substrate buffer (8 µL of arginine buffer and 2 µL of Mn solution), to form urea and ornithine as products, after two hours of incubation at 37 °C. The urea reagent (100 µL of reagent A and 100 µL of reagent B) was added to the obtained urea, giving a colored compound, followed by a room temperature incubation of one hour, stopping the reaction. A control (blank) reaction was run in parallel, containing the same amount of sample, urea reagent and substrate buffer. The amount of colored compound was read at 430 nm in a spectrophotometer (Spectramax plus 384, Molecular Devices, San Jose, CA, USA). The data obtained was exported into a computer program (Softmax pro 6.2.2, Molecular Devices) connected to the device, and then copied into an Excel document.
Full text: Click here
Aorta
Arginase
Arginine
Biological Assay
Buffers
Cells
Cold Temperature
Enzymes
leupeptin
Light
Medical Devices
Normal Saline
Ornithine
pepstatin
Saline Solution
SERPINA1 protein, human
Sterility, Reproductive
Tissues
Triton X-100
Tromethamine
Urea
Thoracic aorta rings from G6PD-Tg and WT mice were isolated from young and old groups. The artery was cleaned with saline (0.9% NaCl) prepared with sterile water treated with diethylpyrocarbonate (DEPC), under an illuminated dissecting loupe (Wild M3C, Leica Microsystems Inc. Buffalo Grove, IL USA) in cold light.
The rings were mounted in an organ bath system to measure the isometric tension and filled with 5 mL of modified Krebs-Henseleit physiological solution composed of (in mM): NaCl 115; KCl 4.6; MgCl2, 6H2O 1,2; CaCl2 2.5; NaHCO3 25; glucose 11.1 and EDTA disodium 0.01. This solution was equilibrated with a gaseous mixture (95% O2 and 5% CO2) that provides a pH of 7.3–7.4. The temperature of the solution was maintained at 37 °C. The optimal passive tension used was 1 g to perform the vascular studies. Vasodilation to acetylcholine (10−6 M) [51 (link)] was studied in aortic rings previously contracted with noradrenaline (3 × 10−7 M–3 × 10−6 M), in the absence and presence of L-arginine (10−3 M) [52 (link)] plus nor-NOHA acetate (10−5 M), an arginase inhibitor for 30 min [20 (link),53 (link)]. A high concentration of acetylcholine (10−6 M) is used to stimulate maximal NO release from endothelial cells. A decreased relaxation to this dose means maximal capacity of NO production.
The rings were mounted in an organ bath system to measure the isometric tension and filled with 5 mL of modified Krebs-Henseleit physiological solution composed of (in mM): NaCl 115; KCl 4.6; MgCl2, 6H2O 1,2; CaCl2 2.5; NaHCO3 25; glucose 11.1 and EDTA disodium 0.01. This solution was equilibrated with a gaseous mixture (95% O2 and 5% CO2) that provides a pH of 7.3–7.4. The temperature of the solution was maintained at 37 °C. The optimal passive tension used was 1 g to perform the vascular studies. Vasodilation to acetylcholine (10−6 M) [51 (link)] was studied in aortic rings previously contracted with noradrenaline (3 × 10−7 M–3 × 10−6 M), in the absence and presence of L-arginine (10−3 M) [52 (link)] plus nor-NOHA acetate (10−5 M), an arginase inhibitor for 30 min [20 (link),53 (link)]. A high concentration of acetylcholine (10−6 M) is used to stimulate maximal NO release from endothelial cells. A decreased relaxation to this dose means maximal capacity of NO production.
Full text: Click here
Acetate
Acetylcholine
Aorta
Arginase
Arginine
Arteries
Bath
Bicarbonate, Sodium
Blood Vessel
Buffaloes
Cold Temperature
Diethyl Pyrocarbonate
Edetic Acid
Endothelial Cells
Gases
Glucose
Glucosephosphate Dehydrogenase
Krebs-Henseleit solution
Light
Magnesium Chloride
Mus
Norepinephrine
Normal Saline
physiology
Saline Solution
Sodium Chloride
Sterility, Reproductive
Thoracic Aorta
Vasodilation
In molecular docking studies with structure-based virtual screening and docking we used various bioinformatics tools, such as AutoDock vina [23 (link),24 (link)], BIOVIA Discovery, Studio 2020 pipeline (Accelrys Inc., San Diego, CA, USA, http://www.accelrys.com (accessed on 16 December 2022), and LigPlot [25 ,26 (link)]. All the 3D structures of the target proteins pyruvate kinase (PDB: 1 PKL), glyceraldehyde-3-phosphate dehydrogenase (PDB: 1A7K), triose phosphate isomerase (PDB: 1AMK), aldolase (PDB: 1EPX), phosphoglucose isomerase (PDB: 1Q50), transketolase (PDB: 1R9J), arginase (PDB: 4ITY) and cysteine peptidases A (PDB: 2C34) of L. mexicana were downloaded from the Protein Data Bank RCSB (PDB). The structures of compounds identified from the subfraction from M. alceifolia were obtained from PubChem and optimized using MMFF94 as a force field and conjugate gradient. The compounds were energy minimized, and the lowest energy conformation of each compound was used for docking studies within the allosteric domains of the evaluated proteins. Finally, the scoring function was determined [27 (link)].
Full text: Click here
Arginase
Cysteine Proteases
Fructosediphosphate Aldolase
Glucosephosphate Isomerase
Glyceraldehyde-3-Phosphate Dehydrogenases
Molecular Structure
Protein Domain
Protein Kinases
Pyruvates
Transketolase
Triose-Phosphate Isomerase
Top products related to «Arginase»
Sourced in United States, Germany
The Arginase Activity Assay Kit is a laboratory tool designed to measure the enzymatic activity of arginase, a metalloenzyme involved in the urea cycle. The kit provides the necessary reagents and protocols to quantify arginase levels in biological samples, enabling researchers to study its role in various physiological and pathological processes.
Sourced in United States
The QuantiChrom Arginase Assay Kit is a colorimetric assay that measures the activity of the enzyme arginase in biological samples. The kit provides a simple, direct, and high-throughput procedure to quantify arginase levels. The assay is based on the enzymatic conversion of L-arginine to L-ornithine and urea, which is then detected using a chromogenic reagent.
Sourced in United States, Germany, Poland, France, United Kingdom, Italy, Sao Tome and Principe, Switzerland, Spain, Croatia, Australia, China, Brazil
L-arginine is an amino acid that plays a crucial role in various physiological processes. It serves as a substrate for the production of nitric oxide, which is essential for maintaining healthy blood flow and cardiovascular function. This lab equipment product can be utilized for research and scientific applications related to the study of L-arginine and its associated biological functions.
Sourced in Taiwan, Province of China, United Kingdom
The Arginase assay kit is a laboratory tool used to measure the activity of the enzyme arginase. Arginase is an enzyme involved in the urea cycle, which is a metabolic pathway that breaks down the amino acid arginine. The assay kit provides a quantitative method to determine the level of arginase activity in biological samples.
Sourced in United States, Germany, United Kingdom, Italy, China, Japan, France, Canada, Sao Tome and Principe, Switzerland, Macao, Poland, Spain, Australia, India, Belgium, Israel, Sweden, Ireland, Denmark, Brazil, Portugal, Panama, Netherlands, Hungary, Czechia, Austria, Norway, Slovakia, Singapore, Argentina, Mexico, Senegal
Triton X-100 is a non-ionic surfactant commonly used in various laboratory applications. It functions as a detergent and solubilizing agent, facilitating the solubilization and extraction of proteins and other biomolecules from biological samples.
Sourced in United States, Germany, China, Japan, United Kingdom, Canada, France, Italy, Australia, Spain, Switzerland, Belgium, Denmark, Netherlands, India, Ireland, Lithuania, Singapore, Sweden, Norway, Austria, Brazil, Argentina, Hungary, Sao Tome and Principe, New Zealand, Hong Kong, Cameroon, Philippines
TRIzol is a monophasic solution of phenol and guanidine isothiocyanate that is used for the isolation of total RNA from various biological samples. It is a reagent designed to facilitate the disruption of cells and the subsequent isolation of RNA.
Sourced in United States
α-isonitrosopropiophenone is a laboratory chemical used in various analytical and research applications. It is a white crystalline solid with a specific chemical formula and structure. This compound is commonly used as a reagent or intermediate in scientific experiments and procedures, but a detailed description of its core function is not available while maintaining an unbiased and factual approach.
Sourced in United States, Switzerland, Germany, China, United Kingdom, France, Canada, Japan, Italy, Australia, Austria, Sweden, Spain, Cameroon, India, Macao, Belgium, Israel
Protease inhibitor cocktail is a laboratory reagent used to inhibit the activity of proteases, which are enzymes that break down proteins. It is commonly used in protein extraction and purification procedures to prevent protein degradation.
Sourced in United States, Germany, China, United Kingdom, Sao Tome and Principe, Macao, Italy, Japan, Canada, France, Switzerland, Israel, Australia, Spain, India, Ireland, Brazil, Poland, Netherlands, Sweden, Denmark, Hungary, Austria, Mongolia
The LPS laboratory equipment is a high-precision device used for various applications in scientific research and laboratory settings. It is designed to accurately measure and monitor specific parameters essential for various experimental procedures. The core function of the LPS is to provide reliable and consistent data collection, ensuring the integrity of research results. No further details or interpretations can be provided while maintaining an unbiased and factual approach.
Sourced in Germany, United States, United Kingdom, Netherlands, Spain, Japan, Canada, France, China, Australia, Italy, Switzerland, Sweden, Belgium, Denmark, India, Jamaica, Singapore, Poland, Lithuania, Brazil, New Zealand, Austria, Hong Kong, Portugal, Romania, Cameroon, Norway
The RNeasy Mini Kit is a laboratory equipment designed for the purification of total RNA from a variety of sample types, including animal cells, tissues, and other biological materials. The kit utilizes a silica-based membrane technology to selectively bind and isolate RNA molecules, allowing for efficient extraction and recovery of high-quality RNA.
More about "Arginase"
Arginase is a crucial enzyme that plays a vital role in the urea cycle, regulating nitric oxide production, and cell proliferation.
This enzyme catalyzes the hydrolysis of L-arginine, a semi-essential amino acid, into L-ornithine and urea.
Understanding the functions and regulation of arginase is crucial for developing effective therapies targeting various pathological conditions, such as inflammation, cancer, and neurological disorders.
Arginase can be measured using specialized assay kits, such as the QuantiChrom Arginase Assay Kit and the Arginase Activity Assay Kit.
These kits utilize colorimetric or fluorometric methods to quantify arginase activity in biological samples.
L-arginine, the substrate for arginase, is also an important compound in these assays.
To study arginase, researchers often employ techniques like cell culture, protein extraction, and enzymatic activity measurement.
Lysis buffers containing Triton X-100 and protease inhibitor cocktails are commonly used to extract and preserve arginase from cells.
RNA extraction methods, such as the RNeasy Mini Kit and TRIzol, can be utilized to analyze arginase gene expression.
Arginase plays a key role in various physiological and pathological processes.
For instance, it is involved in the urea cycle, which is responsible for the excretion of nitrogenous waste products.
Additionally, arginase regulates nitric oxide (NO) production by competing with nitric oxide synthase (NOS) for the common substrate, L-arginine.
This interplay between arginase and NOS can have significant implications in conditions like inflammation, where nitric oxide production is altered.
Furthermore, arginase has been implicated in the development and progression of certain cancers, as it can influence cell proliferation and tumor angiogenesis.
Researchers are actively investigating the potential of arginase as a therapeutic target in cancer and other diseases.
By leveraging the insights provided by PubCompare.ai's AI-powered platform, researchers can enhance their arginase studies by easily accessing relevant protocols from literature, preprints, and patents.
The platform's AI-driven comparisons can also help identify the best protocols and products, improving the reproducibility and accuracy of arginase research.
This enzyme catalyzes the hydrolysis of L-arginine, a semi-essential amino acid, into L-ornithine and urea.
Understanding the functions and regulation of arginase is crucial for developing effective therapies targeting various pathological conditions, such as inflammation, cancer, and neurological disorders.
Arginase can be measured using specialized assay kits, such as the QuantiChrom Arginase Assay Kit and the Arginase Activity Assay Kit.
These kits utilize colorimetric or fluorometric methods to quantify arginase activity in biological samples.
L-arginine, the substrate for arginase, is also an important compound in these assays.
To study arginase, researchers often employ techniques like cell culture, protein extraction, and enzymatic activity measurement.
Lysis buffers containing Triton X-100 and protease inhibitor cocktails are commonly used to extract and preserve arginase from cells.
RNA extraction methods, such as the RNeasy Mini Kit and TRIzol, can be utilized to analyze arginase gene expression.
Arginase plays a key role in various physiological and pathological processes.
For instance, it is involved in the urea cycle, which is responsible for the excretion of nitrogenous waste products.
Additionally, arginase regulates nitric oxide (NO) production by competing with nitric oxide synthase (NOS) for the common substrate, L-arginine.
This interplay between arginase and NOS can have significant implications in conditions like inflammation, where nitric oxide production is altered.
Furthermore, arginase has been implicated in the development and progression of certain cancers, as it can influence cell proliferation and tumor angiogenesis.
Researchers are actively investigating the potential of arginase as a therapeutic target in cancer and other diseases.
By leveraging the insights provided by PubCompare.ai's AI-powered platform, researchers can enhance their arginase studies by easily accessing relevant protocols from literature, preprints, and patents.
The platform's AI-driven comparisons can also help identify the best protocols and products, improving the reproducibility and accuracy of arginase research.