Welcome to LookChem.com Sign In|Join Free

CAS

  • or

526-95-4

Post Buying Request

526-95-4 Suppliers

Recommended suppliersmore

  • Product
  • FOB Price
  • Min.Order
  • Supply Ability
  • Supplier
  • Contact Supplier

526-95-4 Usage

Description

Different sources of media describe the Description of 526-95-4 differently. You can refer to the following data:
1. Gluconic acid (also known as gluconate) is an organic compound occurring widely in nature arising from the glucose oxidation. It is naturally found in fruit, honey and wine. It can also be used as a food additive to regulate acidity and a cleaning agent in alkaline solution. Its calcium salt, calcium gluconate can be used to treat burns from hydrofluoric acid and avoid necrosis of deep tissues as well as treating the verapamil poisoning and hypocalcemia in hospitalized patient. Some salts of gluconate can also be used to treat malaria (quinidine gluconate) and anemia (ferrous gluconate). In microbiology, gluconate is a common carbon source that can be supplemented to the medium for cell growth.
2. Gluconic acid is an organic compound with molecular formula C6H12O7 and condensed structural formula HOCH2(CHOH)4COOH. It is one of the 16 stereoisomers of 2,3,4,5,6-penta hydroxy hexanoic acid. In aqueous solution at neutral pH, gluconic acid forms the gluconate ion. The salts of gluconic acid are known as "gluconates". Gluconic acid, gluconate salts, and gluconate esters occur widely in nature because such species arise from the oxidation of glucose. Some drugs are injected in the form of gluconates.

References

[1]Mandelstam, J. "The repression of constitutive β-galactosidase in Escherichia coli by glucose and other carbon sources." Biochemical Journal82.3 (1962): 489. [2]Ghotbi, Mohammad Yeganeh, et al. "LDH-intercalated d-gluconate: Generation of a new food additive-inorganic nanohybrid compound." Journal of Physics and Chemistry of Solids 70.6 (2009): 948-954. [3]Perkins, C. M. "Serious verapamil poisoning: treatment with intravenous calcium gluconate." British medical journal 2.6145 (1978): 1127. [4]Broner, Cynthia W., et al. "A prospective, randomized, double-blind comparison of calcium chloride and calcium gluconate therapies for hypocalcemia in critically ill children." The Journal of pediatrics 117.6 (1990): 986-989. [5]Miller, K. D., A. E. Greenberg, and C. C. Campbell. "Treatment of severe malaria in the United States with a continuous infusion of quinidine gluconate and exchange transfusion. " New England Journal of Medicine321.2(1989):65-70. [6]Gillespie RS(Driscoll Children's Hospital, Corpus Christi, TX 78411, USA. kidneys@rrmail.com), and J. M. Symons. "Sodium ferric gluconate for post-transplant anemia in pediatric and young adult renal transplant recipients." Pediatric Transplantation 9.1(2005):43–46.

Chemical Properties

Different sources of media describe the Chemical Properties of 526-95-4 differently. You can refer to the following data:
1. clear yellow to brownish-yellow solution
2. d-Gluconic acid is an acid sugar composed of white crystals with a milk-acidic taste. In aqueous solutions, it is in equilibrium with gamma- and delta-gluconolactones. It is prepared by enzymatic oxidation of glucose and strains of the microorganisms used to supply the enzyme action are nonpathogenic and nontoxicogenic to man or other animals. This substance is used as a component of bottle rinsing formulations, at levels not to exceed good manufacturing practice.An FDA letter to a trade association revoking affirmation of general recognition of safety in dietary supplements was dated April 9, 1970.

Physical properties

The chemical structure of gluconic acid consists of a six-carbon chain with five hydroxyl groups terminating in a carboxylic acid group. In aqueous solution, gluconic acid exists in equilibrium with the cyclic ester glucono delta-lactone.

Occurrence

Gluconic acid occurs naturally in fruit, honey, kombucha tea, and wine. As a food additive ( E574 ), it is an acidity regulator. It is also used in cleaning products where it dissolves mineral deposits especially in alkaline solution. The gluconate anion chelates Ca2+,Fe2+, Al3+, and other metals. In 1929 Horace Terhune Herrick developed a process for producing the salt by fermentation. Calcium gluconate, in the form of a gel, is used to treat burns from hydrofluoric acid; calcium gluconate injections may be used for more severe cases to avoid necrosis of deep tissues. Quinine gluconate is a salt between gluconic acid and quinine, which is used for intramuscular injection in the treatment of malaria. Zinc gluconate injections are used to neuter male dogs. Iron gluconate injections have been proposed in the past to treat anemia.

Uses

Different sources of media describe the Uses of 526-95-4 differently. You can refer to the following data:
1. A chemical used in glycolytic pathway studies.
2. Gluconic Acid is an acidulant that is a mild organic acid which is the hydrolyzed form of glucono-delta-lactone. it is prepared by the fermentation of dextrose, whereby the physiological d-form is produced. it is soluble in water with a solubility of 100 g/100 ml at 20°c. it has a mild taste and at 1% has a ph of 2.8. it functions as an antioxidant and enhances the function of other antioxidants. in beverages, syrups, and wine, it can eliminate calcium turbidities. it is used as a leavening component in cake mixes, and as an acid component in dry-mix desserts and dry beverage mixes.
3. Gluconic acid occurs naturally in fruit, honey, and wine. As a food additive it is an acidity regulator. It is also used in cleaning products.

Definition

Different sources of media describe the Definition of 526-95-4 differently. You can refer to the following data:
1. A soluble crystalline organic acid made by the oxidation of glucose (using specific molds). It is used in paint strippers.
2. gluconic acid: An opticallyactive hydroxycarboxylic acid,CH2(OH)(CHOH)4COOH. It is the carboxylicacid corresponding to the aldosesugar glucose, and can be madeby the action of certain moulds.

Biotechnological Production

Currently, gluconic acid is commercially produced by submerged fed-batch cultivations of Aspergillus niger using glucose as substrate. A. niger produces citric acid and gluconic acid growing on glucose. The product concentration and yields of the product depend on the fermentation conditions. For optimal gluconic acid production, high glucose concentrations (110–250 g.L-1), low concentrations of nitrogen and phosphorus in the medium, a limitation of metal ion concentrations, a pH value in the range of 4.5–6.5, and high aeration rates for the oxygen supply are needed. Much research has been carried out to find new ways for cheaper production. Different microorganisms have been studied (e.g. G. oxydans, Z. mobilis, A. methanolicous, and P. fluorescence. Moreover, new microbial strains have been developed by mutagenesis or genetic engineering. Additionally, the fermentation process and recovery have been optimized. New inexpensive substrates (e.g. cornstarch, grape or banana must, figs, and cheese whey) have been tested. One example of a new and efficient production process of gluconic acid is the cultivation of Aureobasidium pullulans growing on glucose. Using a continuous process with biomass retention by crossover filtration, a product concentration of 375 g.L-1, a yield of 0.83 g of gluconic acid per gram of glucose, and a productivity of 17 g.L-1.h-1 could be achieved at a residence time of 22 h. In this process, 100 % of the glucose is converted. This process might be interesting for industrial applications. In continuous gluconic acid production with immobilized mycelia of A. niger, product concentrations of 120–140 g.L-1 have been achieved.

Flammability and Explosibility

Nonflammable

Check Digit Verification of cas no

The CAS Registry Mumber 526-95-4 includes 6 digits separated into 3 groups by hyphens. The first part of the number,starting from the left, has 3 digits, 5,2 and 6 respectively; the second part has 2 digits, 9 and 5 respectively.
Calculate Digit Verification of CAS Registry Number 526-95:
(5*5)+(4*2)+(3*6)+(2*9)+(1*5)=74
74 % 10 = 4
So 526-95-4 is a valid CAS Registry Number.
InChI:InChI=1/C6H12O7/c7-1-2(8)3(9)4(10)5(11)6(12)13/h2-5,7-11H,1H2,(H,12,13)/p-1/t2-,3-,4+,5-/m1/s1

526-95-4 Well-known Company Product Price

  • Brand
  • (Code)Product description
  • CAS number
  • Packaging
  • Price
  • Detail
  • Alfa Aesar

  • (A16806)  D-Gluconic acid, 50% aq. soln.   

  • 526-95-4

  • 500ml

  • 248.0CNY

  • Detail
  • Alfa Aesar

  • (A16806)  D-Gluconic acid, 50% aq. soln.   

  • 526-95-4

  • 2500ml

  • 452.0CNY

  • Detail
  • Aldrich

  • (G1951)  D-Gluconicacidsolution  49-53 wt. % in H2O

  • 526-95-4

  • G1951-25G

  • 284.31CNY

  • Detail
  • Aldrich

  • (G1951)  D-Gluconicacidsolution  49-53 wt. % in H2O

  • 526-95-4

  • G1951-1KG

  • 374.40CNY

  • Detail
  • Aldrich

  • (G1951)  D-Gluconicacidsolution  49-53 wt. % in H2O

  • 526-95-4

  • G1951-4KG

  • 1,075.23CNY

  • Detail

526-95-4SDS

SAFETY DATA SHEETS

According to Globally Harmonized System of Classification and Labelling of Chemicals (GHS) - Sixth revised edition

Version: 1.0

Creation Date: Aug 12, 2017

Revision Date: Aug 12, 2017

1.Identification

1.1 GHS Product identifier

Product name D-gluconic acid

1.2 Other means of identification

Product number -
Other names Gluconic acid

1.3 Recommended use of the chemical and restrictions on use

Identified uses For industry use only. Chelating Agents;Processing Aids and Additives
Uses advised against no data available

1.4 Supplier's details

1.5 Emergency phone number

Emergency phone number -
Service hours Monday to Friday, 9am-5pm (Standard time zone: UTC/GMT +8 hours).

More Details:526-95-4 SDS

526-95-4Synthetic route

β-D-glucose
492-61-5

β-D-glucose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With water; oxygen at 83.84℃; under 3750.38 Torr; for 6h; Autoclave;99%
With 4-methanesulfonyl-2,2,6,6-tetramethyl-1-piperidinyloxy radical; manganese(II) nitrate; oxygen In water at 80℃; under 7500.75 Torr; for 18h; Reagent/catalyst; Pressure; Temperature;82.4%
With Au/Al2O3; dihydrogen peroxide In water at 120℃; for 0.166667h; Sealed tube; Microwave irradiation;76%
D-glucose
50-99-7

D-glucose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium carbonate In water at 30℃; for 40h; Wavelength; Irradiation;99%
With oxygen; sodium carbonate In water at 24.84℃; under 750.075 Torr; for 2h; pH=< 9; Catalytic behavior; Reagent/catalyst;99%
With 5% Pd/C; water; oxygen; sodium carbonate at 20℃; under 760.051 Torr; for 2h; Reagent/catalyst;98%
alpha-D-glucopyranose
492-62-6

alpha-D-glucopyranose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With dihydrogen peroxide; iron(II) sulfate In water at 22.5℃; for 0.25h; Temperature; Reagent/catalyst; Irradiation; Green chemistry;97%
With carbon dioxide; bromine; barium carbonate ueber das δ-Lacton;
With chromium (VI); pyridinium p-toluenesulfonate In acidic aq. solution at 30℃; Kinetics; Oxidation;
Cellobiose
13360-52-6

Cellobiose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With water; oxygen at 144.84℃; under 3750.38 Torr; for 3h; Autoclave;97%
alpha-D-glucopyranose
492-62-6

alpha-D-glucopyranose

A

D-Fructose
57-48-7

D-Fructose

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With dihydrogen peroxide; iron(II) sulfate In water at 70℃; for 0.25h; Temperature; Reagent/catalyst; Green chemistry;A 7%
B 84%
D-glucose
50-99-7

D-glucose

A

D-Fructose
57-48-7

D-Fructose

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With oxygen In water at 50℃; under 750.075 Torr; for 12h; Catalytic behavior; Time; Autoclave; Green chemistry;A 8%
B 83%
LACTOSE
5965-66-2

LACTOSE

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With Aspergillus oryzae β-galactosidase immobilized onto amino-terminated Ni0.5Zn0.5-Fe2O4 magnetic nanoparticles In aq. phosphate buffer at 43.5℃; for 72h; pH=6.7; Reagent/catalyst; Concentration; Enzymatic reaction;79.57%
Cellobiose
13360-52-6

Cellobiose

A

glycolic Acid
79-14-1

glycolic Acid

B

gluconic acid
526-95-4

gluconic acid

C

succinic acid
110-15-6

succinic acid

D

oxalic acid
144-62-7

oxalic acid

E

acetic acid
64-19-7

acetic acid

Conditions
ConditionsYield
With carbon nanotube supported gold nanoparticles (0.5 wt%); water; oxygen at 145℃; under 7500.75 Torr; for 3h;A n/a
B 70%
C n/a
D n/a
E n/a
LACTOSE
5965-66-2

LACTOSE

A

gluconic acid
526-95-4

gluconic acid

B

C18H32O17

C18H32O17

Conditions
ConditionsYield
With Aspergillus oryzae β-galactosidase immobilized onto amino-terminated Ni0.5Zn0.5-Fe2O4 magnetic nanoparticles In aq. phosphate buffer at 43.5℃; for 72h; pH=6.7; Reagent/catalyst; Concentration; Enzymatic reaction;A 68.33%
B 27.4%
Cellobiose
13360-52-6

Cellobiose

A

glycolic Acid
79-14-1

glycolic Acid

B

D-glucose
50-99-7

D-glucose

C

gluconic acid
526-95-4

gluconic acid

D

succinic acid
110-15-6

succinic acid

E

oxalic acid
144-62-7

oxalic acid

F

acetic acid
64-19-7

acetic acid

Conditions
ConditionsYield
With carbon nanotube supported gold nanoparticles (0.5 wt%); water; oxygen at 145℃; under 3750.38 Torr; for 3h;A n/a
B n/a
C 68%
D n/a
E n/a
F n/a
D-Mannose
3458-28-4

D-Mannose

A

mannonic acid
642-99-9

mannonic acid

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium anthraquinone-2-sulfonate; calcium chloride In sodium hydroxide at 50℃;A 57%
B 3%
With sulfuric acid; manganese(III) ion In water at 32℃; Kinetics; Activation energy; Further Variations:; pH-values; Reagents; Solvents; Temperatures; Oxidation;
D-glucose
50-99-7

D-glucose

A

mannonic acid
642-99-9

mannonic acid

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium anthraquinone-2-sulfonate; calcium chloride In sodium hydroxide at 50℃;A 57%
B 3%
With sulfuric acid; manganese(III) ion In water at 32℃; Kinetics; Activation energy; Further Variations:; pH-values; Reagents; Solvents; Temperatures; Oxidation;
D-glucose
50-99-7

D-glucose

A

formic acid
64-18-6

formic acid

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With iron(III) chloride at 110℃; for 4h; Concentration; Sealed tube;A 15.6%
B 52.3%
at 160℃; under 15001.5 Torr; for 2h; Acidic aq. solution; Inert atmosphere; Electrolysis; Autoclave;
With oxygen; titanium(IV) oxide In water for 3h; Catalytic behavior; UV-irradiation;
D-glucose
50-99-7

D-glucose

A

formic acid
64-18-6

formic acid

B

gluconic acid
526-95-4

gluconic acid

C

acetic acid
64-19-7

acetic acid

Conditions
ConditionsYield
With iron(III) chloride at 120℃; for 1.5h; Temperature; Concentration;A 15.4%
B 51.3%
C 6.5%
With iron(III) chloride at 110℃; for 2h; Concentration; Sealed tube;A 12.3%
B 48.7%
C 10.8%
With iron(III) chloride at 110℃; for 2h; Concentration; Sealed tube;A 8.2%
B 29.8%
C 8.7%
With iron(III) chloride at 100℃; for 4h;A 9.2%
B 13.3%
C 27.2%
With iron(III) chloride In water at 110℃; for 4h; Concentration; Temperature; Time;A n/a
B 52.3 %Chromat.
C n/a
D-glucose
50-99-7

D-glucose

chloroamine-T
127-65-1

chloroamine-T

B

D-threonic acid
20246-26-8

D-threonic acid

C

D-erythronic acid
488-16-4

D-erythronic acid

D

arabinoic acid
488-30-2

arabinoic acid

E

D-ribonic acid
642-98-8

D-ribonic acid

F

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
In water at 35℃; Kinetics; Thermodynamic data; Mechanism; Ea; ΔH excit.; ΔG excit.; ΔS excit; add. of NaOH, var. solv. D2O;A 3%
B n/a
C n/a
D 35%
E 21%
F 5%
D-glucose
50-99-7

D-glucose

A

formic acid
64-18-6

formic acid

B

gluconic acid
526-95-4

gluconic acid

C

D-glucaric acid
87-73-0

D-glucaric acid

Conditions
ConditionsYield
With dihydrogen peroxide; 3-butyl-1-methyl-1H-imidazol-3-ium hexafluorophosphate In water at 60℃; for 18h;A 7%
B 35%
C 17%
With oxygen; potassium hydrogen phthalate In water for 3h; pH=4.01; Catalytic behavior; Kinetics; Mechanism; Reagent/catalyst; pH-value; UV-irradiation;
D-glucose
50-99-7

D-glucose

A

gluconic acid
526-95-4

gluconic acid

B

D-glucaric acid
87-73-0

D-glucaric acid

Conditions
ConditionsYield
With oxygen; sodium hydroxide In water at 60℃; under 7500.75 Torr; for 24h; Catalytic behavior; Temperature; Pressure; Reagent/catalyst; Autoclave;A 31%
B 18%
With MoO5; dihydrogen peroxide; 3-butyl-1-methyl-1H-imidazol-3-ium hexafluorophosphate In water at 60℃; for 18h;A 27%
B 13%
With ammonium vanadate In sulfuric acid at 90℃; for 3h; Kinetics; Mechanism; Equilibrium constant; activation energy; different concentrations of H2SO4;
With oxygen; sodium nitrite In sulfolane; perchloric acid at 59.85℃; under 750.06 Torr; Kinetics; Further Variations:; Temperatures; Pressures;
With Fe-doped TiO2-supported zeolite; air In water; acetonitrile at 30℃; under 760.051 Torr; for 1.5h; Reagent/catalyst; Solvent; UV-irradiation;
D-glucose
50-99-7

D-glucose

A

D-Fructose
57-48-7

D-Fructose

B

mannitol
69-65-8

mannitol

C

D-sorbitol
50-70-4

D-sorbitol

D

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With hydrogen In water at 90℃; under 12001.2 Torr; for 6h; Time; Green chemistry;A 14%
B 19.5%
C 26%
D 5.5%
D-Glucose
2280-44-6

D-Glucose

A

formic acid
64-18-6

formic acid

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With dihydrogen peroxide In water at 160℃; for 0.25h; pH=5.4;A 9.18%
B 15.03%
With dihydrogen peroxide In water at 160℃; for 1h; pH=5.4;A 10.65%
B 7.5%
D-Glucose
2280-44-6

D-Glucose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With dihydrogen peroxide In water at 200℃; for 0.25h; pH=5.4;11.69%
With oxygen; Pd (SB12)-activated carbon In water at 56℃; Product distribution; normal pressure; other catalyst;
With sodium hexachloroplatinate; hydroxide In water at 29.9℃; Kinetics; Mechanism; Thermodynamic data; ΔH(excit.); ΔS(excit.); ΔG(excit.); different hydroxide ion, sodium hexachloroplatinate, sodium chloride and substrate concentrations and temperatures;
brucine
357-57-3

brucine

mannonic acid
642-99-9

mannonic acid

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
das Lacton ragiert;
tetrachloromethane
56-23-5

tetrachloromethane

D-gluconamide
3118-85-2

D-gluconamide

gluconic acid
526-95-4

gluconic acid

D-Fructose
57-48-7

D-Fructose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium hydroxide; platinum
With copper(II) hydroxide; sodium hydroxide
D-Mannose
3458-28-4

D-Mannose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With copper(II) hydroxide; sodium hydroxide
D-glucose
50-99-7

D-glucose

A

D-sorbitol
50-70-4

D-sorbitol

B

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium hydroxide; nickel
With sodium hydroxide; platinum
With sodium hydroxide; sodium sulfate; sodium bromide In water at 60℃; paired electrolysis with Pb sheet cathode and dimension stable anode, current density of 50 mA cm-2, Qr is 110percent;
D-mannono-1,4-lactone
26301-79-1

D-mannono-1,4-lactone

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With barium dihydroxide at 95℃; 100 h;
D-glucaro-6,3-lactone
2782-04-9

D-glucaro-6,3-lactone

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
With sodium amalgam
Sucrose
57-50-1

Sucrose

gluconic acid
526-95-4

gluconic acid

Conditions
ConditionsYield
durch verschiedene Essigbakterien;
durch Aspergillus niger;
mit Hilfe von Penicillium crustaceum;
(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-methoxy-chromen-4-one

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-methoxy-chromen-4-one

gluconic acid
526-95-4

gluconic acid

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-methoxy-chromen-4-one gluconate

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-methoxy-chromen-4-one gluconate

Conditions
ConditionsYield
In methanol; water100%
In methanol; water100%
(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methylpyrrolidin-3-yl)-7-(2-methoxyethoxymethoxy)-chromen-4-one

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methylpyrrolidin-3-yl)-7-(2-methoxyethoxymethoxy)-chromen-4-one

gluconic acid
526-95-4

gluconic acid

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-(2-methoxyethoxymethoxy)-chromen-4-one gluconate

(+)-trans-2-(2-chlorophenyl)-5-hydroxy-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-7-(2-methoxyethoxymethoxy)-chromen-4-one gluconate

Conditions
ConditionsYield
In methanol at 20℃; for 0.5h;100%
In methanol at 20℃; for 0.5h;100%
(+)-trans-2-(2-chlorophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one

(+)-trans-2-(2-chlorophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one

gluconic acid
526-95-4

gluconic acid

(+)-trans-2-(2-chlorophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one gluconate

(+)-trans-2-(2-chlorophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one gluconate

Conditions
ConditionsYield
In methanol100%
In methanol100%
gluconic acid
526-95-4

gluconic acid

rasagiline
136236-51-6

rasagiline

R-(+)-N-propargyl-1-aminoindan gluconate
1204184-71-3

R-(+)-N-propargyl-1-aminoindan gluconate

Conditions
ConditionsYield
In ethanol; water for 2h; Product distribution / selectivity;100%
In water; isopropyl alcohol at 40℃; for 1h;
gluconic acid
526-95-4

gluconic acid

tetra-n-butylphosphonium hydroxide
14518-69-5

tetra-n-butylphosphonium hydroxide

tetrabutylphosphonium gluconate

tetrabutylphosphonium gluconate

Conditions
ConditionsYield
In water for 24h;98%
gluconic acid
526-95-4

gluconic acid

A

carbon dioxide
124-38-9

carbon dioxide

B

hydrogen
1333-74-0

hydrogen

Conditions
ConditionsYield
With water at 20℃; pH=4.5; Quantum yield; UV-irradiation; Inert atmosphere;A n/a
B 97%
gluconic acid
526-95-4

gluconic acid

betaine
107-43-7

betaine

betainium D-gluconate

betainium D-gluconate

Conditions
ConditionsYield
In ethanol at 25℃; for 0.5h;97%
gluconic acid
526-95-4

gluconic acid

H-Gpn-OH
60142-96-3

H-Gpn-OH

C9H17NO2*C6H12O7

C9H17NO2*C6H12O7

Conditions
ConditionsYield
In neat (no solvent) for 0.5h; Green chemistry;95.2%
gluconic acid
526-95-4

gluconic acid

2-diethylamino-N-(2,6-dimethylphenyl)-acetamide
137-58-6

2-diethylamino-N-(2,6-dimethylphenyl)-acetamide

2-((2,6-dimethylphenyl)amino)-N,N-diethyl-2-oxoethan-1-aminium (2R,3S,4R,5R)-2,3,4,5,6-pentahydroxyhexanoate

2-((2,6-dimethylphenyl)amino)-N,N-diethyl-2-oxoethan-1-aminium (2R,3S,4R,5R)-2,3,4,5,6-pentahydroxyhexanoate

Conditions
ConditionsYield
In ethanol for 5h; pH=2 - 7;95%
gluconic acid
526-95-4

gluconic acid

tetra(n-butyl)ammonium hydroxide
2052-49-5

tetra(n-butyl)ammonium hydroxide

tetrabutylammonium gluconate

tetrabutylammonium gluconate

Conditions
ConditionsYield
In water for 24h;95%
(+)-trans-2-(2-chloro-4-nitrophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one

(+)-trans-2-(2-chloro-4-nitrophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one

gluconic acid
526-95-4

gluconic acid

(+)-trans-2-(2-chloro-4-nitrophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one gluconate

(+)-trans-2-(2-chloro-4-nitrophenyl)-8-(2-hydroxymethyl-1-methyl-pyrrolidin-3-yl)-5,7-dimethoxy-chromen-4-one gluconate

Conditions
ConditionsYield
In methanol93.9%
In methanol93.9%
gluconic acid
526-95-4

gluconic acid

D-Glucono-1,5-lactone
90-80-2

D-Glucono-1,5-lactone

Conditions
ConditionsYield
In 1,4-dioxane; water for 0.183333h; microwave irradiation;92%
at 50℃; under 20 Torr;
at 70 - 80℃;
gluconic acid
526-95-4

gluconic acid

carboxymethyl-dodecyl-dimethyl-ammonium betaine
683-10-3

carboxymethyl-dodecyl-dimethyl-ammonium betaine

C16H34NO2(1+)*C6H11O7(1-)

C16H34NO2(1+)*C6H11O7(1-)

Conditions
ConditionsYield
In ethanol at 25℃; for 0.5h;92%
gluconic acid
526-95-4

gluconic acid

C8H17NO2

C8H17NO2

C8H18NO2(1+)*C6H11O7(1-)

C8H18NO2(1+)*C6H11O7(1-)

Conditions
ConditionsYield
In ethanol at 25℃; for 0.5h;91%
gluconic acid
526-95-4

gluconic acid

mercury(II) oxide

mercury(II) oxide

C12H22Hg2O14

C12H22Hg2O14

Conditions
ConditionsYield
In water at 50 - 55℃;85%
gluconic acid
526-95-4

gluconic acid

1,4-diaminobutane
110-60-1

1,4-diaminobutane

N-(4-aminobutyl)-D-gluconamide
230305-46-1

N-(4-aminobutyl)-D-gluconamide

Conditions
ConditionsYield
With O-(3,4-dihydro-4-oxo-1,2,3-benzotriazin-3-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate In N,N-dimethyl-formamide for 4h;83.8%
gluconic acid
526-95-4

gluconic acid

[N-(2-{[2-(dimethylamino)ethyl](methyl)amino}-4-methoxy-5-{[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino}phenyl)propen-2-amide]
1421373-65-0

[N-(2-{[2-(dimethylamino)ethyl](methyl)amino}-4-methoxy-5-{[4-(1-methyl-1H-indol-3-yl)pyrimidin-2-yl]amino}phenyl)propen-2-amide]

AZD9291 gluconate

AZD9291 gluconate

Conditions
ConditionsYield
In water; acetone at 20℃; for 0.5h;77%
methanol
67-56-1

methanol

gluconic acid
526-95-4

gluconic acid

3,4-dichlorobenzaldehyde
6287-38-3

3,4-dichlorobenzaldehyde

2,4-(3,4-dichloro)benzylidene methy-D-gluconate

2,4-(3,4-dichloro)benzylidene methy-D-gluconate

Conditions
ConditionsYield
With hydrogenchloride In water at 20℃; for 36h;76%
With hydrogenchloride In water at 20℃; for 24h;76%
gluconic acid
526-95-4

gluconic acid

3,4-dichlorobenzaldehyde
6287-38-3

3,4-dichlorobenzaldehyde

C14H16Cl2O7

C14H16Cl2O7

Conditions
ConditionsYield
Stage #1: gluconic acid With hydrogenchloride In methanol; water for 0.5h;
Stage #2: 3,4-dichlorobenzaldehyde In methanol; water
76%
methanol
67-56-1

methanol

gluconic acid
526-95-4

gluconic acid

3,4-dichlorobenzaldehyde
6287-38-3

3,4-dichlorobenzaldehyde

C14H16Cl2O7

C14H16Cl2O7

Conditions
ConditionsYield
With hydrogenchloride In water at 20℃; for 24h;76%
gluconic acid
526-95-4

gluconic acid

(8S)-2,3-dimethyl-8-(2-methylphenyl)-3,6,7,8-tetrahydrochromeno[7,8-d]imidazole-5-carboxamide
1092938-91-4

(8S)-2,3-dimethyl-8-(2-methylphenyl)-3,6,7,8-tetrahydrochromeno[7,8-d]imidazole-5-carboxamide

(8S)-N,N,1,2-tetramethyl-8-(2-methylphenyl)-3,6,7,8-tetrahydrochromeno[7,8-d]imidazole-5-carboxamide glucuronate (1:1.48)

(8S)-N,N,1,2-tetramethyl-8-(2-methylphenyl)-3,6,7,8-tetrahydrochromeno[7,8-d]imidazole-5-carboxamide glucuronate (1:1.48)

Conditions
ConditionsYield
In Isopropyl acetate at 55℃; for 5h;75%
gluconic acid
526-95-4

gluconic acid

tegaserod
1044642-88-7

tegaserod

3-(5-methoxy-1H-indol-3-ylmethylene)-N-pentylcarbazimidamide D-gluconate

3-(5-methoxy-1H-indol-3-ylmethylene)-N-pentylcarbazimidamide D-gluconate

Conditions
ConditionsYield
In water; isopropyl alcohol at 20 - 70℃;74.8%

526-95-4Relevant articles and documents

The reduction of Cr(VI) to Cr(III) by the α and β anomers of D-glucose in dimethyl sulfoxide. A comparative kinetic and mechanistic study

Signorella, Sandra,Lafarga, Rubén,Daier, Verónica,Sala, Luis F.

, p. 127 - 135 (2000)

The reduction of Cr(VI) by α-D-glucose and β-D-glucose was studied in dimethyl sulfoxide in the presence of pyridinium p-toluensulfonate, a medium where mutarotation is slower than the redox reaction. The two anomers reduce Cr(VI) by formation of an intermediate Cr(VI) ester precursor of the slow redox step. The equilibrium constant for the formation of the intermediate chromic ester and the rate of the redox steps are different for each anomer. α-D-Glucose forms the Cr(VI)-Glc ester with a higher equilibrium constant than β-D-glucose, but the electron transfer within this complex is slower than for the β anomer. The difference is attributed to the better chelating ability of the 1,2-cis-diolate moiety of the α anomer. The Cr(V) species, generated in the reaction mixture, reacts with the two anomers at a rate comparable with that of Cr(VI). The EPR spectra show that the α anomer forms several linkage isomers of the five-coordinate Cr(V) bis-chelate, while β-D- glucose affords a mixture of six-coordinate Cr(V) mono-chelate and five- coordinate Cr(V) bis-chelate. The conversion of the Cr(V) mono- to bis- chelate is discussed in terms of the ability of the 1,2-cis- versus 1,2- trans-diolate moieties of the glucose anomers to bind Cr(V). (C) 2000 Elsevier Science Ltd.

Glucose biosensor based on new carbon nanotube-gold-titania nano-composites modified glassy carbon electrode

Wu, Yue-Li,Li, Qi-Wei,Zhang, Xiao-Lu,Chen, Xiao,Wang, Xue-Mei

, p. 1087 - 1090 (2013)

In this paper, a novel biosensor was prepared by immobilizing glucose oxidase (GOx) on carbon nanotube-gold-titania nanocomposites (CNT/Au/TiO 2) modified glassy carbon electrode (GCE). SEM was initially used to investigate the surface morphology of CNT/Au/TiO2 nanocomposites modified GCE, indicating the formation of the nano-porous structure which could readily facilitate the attachment of GOx on the electrode surface. Cyclic voltammogram (CV) and electrochemical impedance spectrum (EIS) were further utilized to explore relevant electrochemical activity on CNT/Au/TiO2 nanocomposites modified GCE. The observations demonstrated that the immobilized GOx could efficiently execute its bioelectrocatalytic activity for the oxidation of glucose. The biosensor exhibited a wider linearity range from 0.1 mmol L-1 to 8 mmol L-1 glucose with a detection limit of 0.077 mmol L-1.

Sol-gel encapsulated glucose oxidase arrays based on a pH sensitive fluorescent dye

Yildirim, Nimet,Odaci, Dilek,Ozturk, Gulsiye,Alp, Serap,Ergun, Yavuz,Dornbusch, Kay,Feller, Karl-Heinz,Timur, Suna

, p. 144 - 148 (2011)

Optical glucose oxidase (GOx) arrays based on pH sensitive fluorescent dye (2-(4-tolyl)-4-[4-(1,4,7,10-tetraoxa-13-azacyclopentadecyl)benzylidene] -5-oxazolone) (CPO) has been constructed. The arrays were prepared by spotting of CPO and GOx together with tetraethoxysilane (TEOS)/Chitosan (CHIT) mixture via a microarrayer. After optimization studies, analytical characterization of enzyme arrays were carried out. The fluorescence intensity of the system was linearly correlated to glucose concentration in the range of 1.0-30.0 mM (in potassium phosphate buffer; 2.5 mM at pH 7.0). Furthermore, the developed arrays were used to analyze glucose in some beverages and HPLC was used as a reference method for independent glucose analysis.

Label-free Si quantum dots as photoluminescence probes for glucose detection

Yi, Yinhui,Deng, Jianhui,Zhang, Youyu,Li, Haitao,Yao, Shouzhuo

, p. 612 - 614 (2013)

Si quantum dots have been demonstrated to be environmentally friendly photoluminescence probes and their fluorescence was quenched by H 2O2 that was produced from the glucose oxidase-catalyzed oxidation of glucose. This strategy could be used to detect glucose with high sensitivity and selectivity.

Diverse applications of TMB-based sensing probes

Huang, Li,Cao, Ying-Juan,Sun, Xiang-Ying,Liu, Bin,Shen, Jiang-Shan

, p. 5667 - 5676 (2018)

Extending the research on 3,3′,5,5′-tetramethylbenzidine (TMB) and its derivatives in analytical chemistry is important, considering that TMB is widely used as an enzyme catalytic substrate. In this work, two TMB derivatives, TMBS and TMBB, were synthesized via a facile and one-step condensation reaction between the -NH2 group of TMB and the -CHO group of salicylaldehyde or benzaldehyde. Because at low pH the two Schiff base compounds can release TMB which can emit strong fluorescence, the probes could show dual-modal signal responses, fluorescence and UV-vis absorption, towards the pH. Practical applications of pH sensing in Chinese rice vinegar and lemon juice samples were successfully demonstrated. On the basis of these findings, a catalytic chromogenic reaction was developed to monitor the pH with the naked eye, too. Furthermore, considering the chemical equilibrium reaction between CO2 and H2O and that glucose oxidase (GOD) can catalyse the dehydrogenation and oxidation reaction of β-d-glucose to produce gluconic acid, both of which can result in lowering the pH values of the two Schiff base systems, highly sensitive and selective dual-modal sensing systems for detecting CO2 and β-d-glucose have also been successfully established. Therefore, the two synthesized TMB derivatives can demonstrate their robust application potential.

Behavior of IrCl3 as a Homogeneous Catalyst on the Oxidation of N-Acetylglucosamine in Acid Medium and Uncatalyzed Reaction in Alkaline Medium with Bromamine-B: Exploration of Kinetic, Mechanistic and Catalytic Chemistry

Shankarlingaiah, Dakshayani,Puttaswamy

, p. 424 - 437 (2018)

Abstract: The experimental rate laws for the oxidation of N-acetylglucosamine with bromamine- B are: ? d[BAB]/dt = k/ [BAB]1 [GlcNAc]0.69 [HClO4]?0.76 [IrCl3]0.48 [BSA]?0.33 in acid medium and –d[BAB]/dt = k/?[BAB]1 [GlcNAc]1 [NaOH]0.79 in alkaline medium. The IrCl3 catalyzed reaction is thirteen fold faster than uncatalyzed reaction. Appropriate mechanisms and rate laws were deduced. Graphical Abstract: The reaction of N-acetylglucosamine with Bromamine-B in acid and alkaline medium is [Figure not available: see fulltext.].

A falling-film microreactor for enzymatic oxidation of glucose

Illner, Sabine,Hofmann, Christian,Loeb, Patrick,Kragl, Udo

, p. 1748 - 1754 (2014)

Many oxidation processes require the presence of molecular oxygen in the reaction media. Reactors are needed that provide favorable conditions for the mass transfer between the gas and the liquid phase. In this study, two recent key technologies, microreactor technology and biotechnology, were combined to present an interesting alternative to conventional methods and open up excellent possibilities to intensify chemical processes in the field of fine chemicals. An enzyme-catalyzed gas/liquid phase reaction in a falling-film microreactor (FFMR) was examined for the first time. The test reaction was the oxidation of β-D-glucose to gluconic acid catalyzed by glucose oxidase (GOx). Various factors influencing the biotransformation, such as oxygen supply, temperature, enzyme concentration, and reaction time were investigated and compared to those in conventional batch systems. The most critical factor, the volumetric mass-transfer coefficient for the efficient use of oxygen-dependent enzymes, was determined by using the integrated online detection of dissolved oxygen in all systems. The extremely large surface-to-volume ratio of the FFMR facilitated the contact between the enzyme solution and the gaseous substrate. Hence, in a continuous bubble-free FFMR system with a residence time of 25 seconds, a final conversion of up to 50 % in enzymatic oxidation was reached, whereas conversion in a conventional bubble column resulted in only 27 %. Finally, an option for scale-up was shown through an enlarged version of the FFMR. Oxygen intake: An enzyme-catalyzed gas/liquid phase reaction in a falling film microreactor (FFMR) was examined for the first time. The test reaction was the oxidation of β-D-glucose to gluconic acid catalyzed by glucose oxidase. As a result of to the large surface-to-volume ratio, extremely high oxygen transfer rates are achieved.

A sandwich-like electrochemiluminescent biosensor for the detection of concanavalin A based on a C60-reduced graphene oxide nanocomposite and glucose oxidase functionalized hollow gold nanospheres

Zhang, Juanjuan,Ruo, Yuan,Chen, Shihong,Zhong, Xia,Wu, Xiaoping

, p. 48465 - 48471 (2014)

A sensitive sandwich-like electrochemiluminescent (ECL) biosensor was designed for the detection of concanavalin A (ConA) using a C60-reduced graphene oxide (C60-rGO) nanocomposite as a platform and glucose oxidase (GOX) decorated hollow gold nanospheres (HGNSs) as a label. First, C60-rGO with a large surface area was prepared for combining with phenoxy-derivatized dextran, which served as the recognition element for interacting with ConA by biospecific carbohydrate-protein (lectin) interactions. Then, GOX decorated HGNSs (GOX-HGNSs) were linked to the electrode surface through the biospecific interaction between the intrinsic carbohydrate residues of GOX and ConA. These localized GOX and HGNSs amplified the ECL signal of luminol intensely, which was achieved by the efficient catalysis of the GOX towards the oxidation of glucose to in situ generate an improved amount of hydrogen peroxide (H2O2) as a coreactant, and the excellent catalysis of HGNSs towards the ECL reaction of luminol-H2O2. The prepared biosensor exhibited a sensitive response for the determination of ConA, ranging from 0.10 to 100 ng mL-1 with a detection limit down to 30 pg mL-1 (signal to noise = 3). With excellent stability, sensitivity, selectivity and simplicity, the prepared biosensor showed great prospects in lectin sensing or carbohydrate sensing. This journal is

Electrical contacting of flavoenzymes and NAD(P)+-dependent enzymes by reconstitution and affinity interactions on phenylboronic acid monolayers associated with Au-electrodes

Zayats, Maya,Katz, Eugenii,Willner, Itamar

, p. 14724 - 14735 (2002)

The preparation of integrated, electrically contacted, flavoenzyme and NAD(P)+-dependent enzyme-electrodes is described. The reconstitution of apo-glucose oxidase, apo-GOx, on a FAD cofactor linked to a pyrroloquinoline quinone (PQQ) phenylboronic acid monolayer yields an electrically contacted enzyme monolayer (surface coverage 2.1 × 10-12 mol cm-2) exhibiting a turnover rate of 700 s-1 (at 22 ± 2 °C). The system is characterized by microgravimetric quartz-crystal microbalance analyses, Faradaic impedance spectroscopy, rotating disk electrode experiments, and cyclic voltammetry. The performance of the enzyme-electrode for glucose sensing is described. Similarly, the electrically contacted enzyme-electrodes of NAD(P)+-dependent enzymes malate dehydrogenase, MaID, and lactate dehydrogenase, LDH, are prepared by the cross-linking of affinity complexes generated between the enzymes and the NADP+ and NAD+ cofactors linked to a pyrroloquinoline quinone phenylboronic acid monolayer, respectively. The MaID enzyme-electrode (surface coverage 1.2 × 10-12 mol cm2) exhibits a turnover rate of 190 s-1, whereas the LDH enzyme-electrode (surface coverage 7.0 × 10-12 mol cm2) reveals a turnover rate of 2.5 s-1. Chronoamperometric experiments reveal that the NAD+ cofactor is linked to the PQQ-phenylboronic acid by two different binding modes. The integration of the LDH with the two NAD+ cofactor configurations yields enzyme assemblies differing by 1 order of magnitude in their bioelectrocatalytic activities.

Low temperature synthesis of Cu2O crystals: Shape evolution and growth mechanism

Sui, Yongming,Fu, Wuyou,Yang, Haibin,Zeng, Yi,Zhang, Yanyan,Zhao, Qiang,Li, Yangen,Zhou, Xiaoming,Leng, Yan,Li, Minghui,Zou, Guangtian

, p. 99 - 108 (2010)

An interesting shape evolution of Cu2O crystals, that is, from cubes, truncated octahedra, octahedra, and finally to nanospheres was first realized in high yield by reducing the copper-citrate complex solution with glucose. X-ray powder diffraction (XRD), field-emission scanning electron microscopy (FESEM), and high-resolution transmission electron microscopy (H RTEM) techniques were employed to characterize the samples. We elucidate the important parameters (including poly (vinyl pyrrolidone) (PVP) concentration, reaction time, and reaction temperature) responsible for the shape-controlled synthesis of Cu2O crystals. The possible formation mechanism for the products with various architectures is presented, which is mainly based on the variation of the ratio (R) of the growth rates along the ( 100) and (111) direction. In addition, the effect of the low supersaturation on the formation of star-shaped samples with six symmetric branches is also taken into account. This polymer-mediated method should be readily extended to the controlled synthesis of other metal oxides and the proposed growth model could also be used to explain and direct the growth of crystals with a cubic structure

Preparation and catalytic activity of poly(N-vinyl-2-pyrrolidone)-protected Au nanoparticles for the aerobic oxidation of glucose

Zhang, Haijun,Li, Wenqi,Gu, Yajun,Zhang, Shaowei

, p. 5743 - 5751 (2014)

PVP-protected Au nanoparticles (NPs) for the aerobic oxidation of glucose were prepared by using NaBH4 reduction method. The effects of processing parameters such as Au3+ ion concentration, reaction temperature, ratio of NaBH4 or PVP to Au3+, and solvent composition on their particle sizes and catalytic activities were studied in detail and the synthesis conditions optimized. As-prepared Au NPs possessed a FCC structure, with an average size varying from about 100 to 2.6 nm depending on their preparation conditions. The size changes affected their catalytic activities in the aerobic oxidation of glucose. The Au NPs with the average size of 2.6 nm prepared under the optimal conditions showed a high instantaneous catalytic activity as well as a high long-time stability. Based on the kinetic study on the glucose oxidation over the PVP-protected Au NPs, the corresponding apparent activation energy was determined as 82 kJ mol-1. Copyright

Cook,Major

, p. 773 (1935)

A kinetic study of d-glucose oxidation by bromine in aqueous solutions

Grgur, Branimir N.,Zugic, Dragana L.,Gvozdenovic, Milica M.,Trisovic, Tomislav Lj.

, p. 1779 - 1787 (2006)

The kinetics of the oxidation of d-glucose to d-gluconic acid by bromine in aqueous solution were studied using potentiometric techniques and theoretical considerations of complex bromine-bromide-pH equilibria. The pH has a strong influence on reaction rate. At pH 9.5, the reaction is further accelerated due to the formation of hypobromite. The proposed kinetics expression for gluconic acid formation, based on the determined kinetic parameters at pH 9.24, is of the form. dc(GA)/dt = 160c2(G)c0(HOBr)c0(H+)c0(Br-).

Insights into the "free state" enzyme reaction kinetics in nanoconfinement

Wang, Chen,Ye, De-Kai,Wang, Yun-Yi,Lu, Tao,Xia, Xing-Hua

, p. 1546 - 1553 (2013)

The investigation of enzyme reaction kinetics in nanoconfined spaces mimicking the conditions in living systems is of great significance. Here, a nanofluidics chip integrated with an electrochemical detector has been designed for studying "free state" enzyme reaction kinetics in nanoconfinement. The nanofluidics chip is fabricated using the UV-ablation technique developed in our group. The enzyme and substrate solutions are simultaneously supplied from two single streams into a nanochannel through a Y-shaped junction. The laminar flow forms in the front of the nanochannel, then the two liquids fully mix at their downstream where a homogeneous enzyme reaction occurs. The "free state" enzyme reaction kinetics in nanoconfinement can thus be investigated in this laminar flow based nanofluidics device. For demonstration, glucose oxidase (GOx) is chosen as the model enzyme, which catalyzes the oxidation of beta-d-glucose. The reaction product hydrogen peroxide (H 2O2) can be electrochemically detected by a microelectrode aligning to the end of nanochannel. The steady-state electrochemical current responding to various glucose concentrations is used to evaluate the activity of the "free state" GOx under nanoconfinement conditions. The effect of liquid flow rate, enzyme concentration, and nanoconfinement on reaction kinetics has been studied in detail. Results show that the "free state" GOx activity increases significantly compared to the immobilized enzyme and bath system, and the GOx reaction rate in the nanochannel is two-fold faster than that in bulk solution, demonstrating the importance of "free state" and spatial confinement for the enzyme reaction kinetics. The present approach provides an effective method for exploiting the "free state" enzyme activity in nanospatial confinement.

KINETICS OF OXIDATION OF D-GLUCOSE BY HEXACHLOROIRIDATE(IV) AND TETRACHLOROAURATE(III)

Gupta, Kalyan Kali Sen,Gupta, Shipra Sen,Chatterejee, Uma,Tarafdar, Archana,Samanta, Tapashi,Shamra, Umashankar

, p. 81 - 88 (1983)

Kinetic data for the oxidation of D-glucose by hexachloroiridate(IV) and tetrachloroaurate(III) are reported.The reactions have been studied spectrophotometrically over a vide range of experimental conditions.Theactivation parameters have been calculated and mechanims are suggested.

Studies on the co-immobilized GOD/CAT on cross-linked chitosan microsphere modified by lysine

Zhang, Jie,Zhou, Xiaohua,Wang, Dan,Wang, Yingli,Zhou, Xing,Wang, Honghui,Li, Qiang,Tan, Shiyu

, p. 80 - 86 (2013)

Glucose oxidase (GOD) and catalase (CAT) were co-immobilized on a novel crosslinked chitosan resin modified by l-lysine. The immobilized system exhibited the best performance when the immobilization process was conducted at 4 C for 3 h with GOD/CAT activity ratio of 1:18 and pH 7.0. Under the optimum pH 4.0 and temperature 40 C, the enzyme system could achieve to the maximum conversion of glucose, which was 68.9%. Acidoresistance and thermotolerance of the enzymes were both strengthened. The Km of the co-immobilized enzyme system obtained in this study was 31.02 mM. The GOD enzyme immobilized in this system could maintain 94% of initial activity after the 12th operational cycle. 95.8% of initial activity was left after 50 weeks' storage at 4 C. GOD achieved better performance in this new immobilized enzyme system due to its effective elimination of H2O2, which showed potential applications for various commercial uses.

Glucose-specific sensing with boronic acid utilizing enzymatic oxidation

Kanekiyo, Yasumasa,Tao, Hiroaki

, p. 852 - 853 (2006)

A novel sensing system for glucose-specific detection has been established based on a combination of a boronic acid and enzymes. To overcome the inherently low biding affinity of glucose toward boronic acids, glucose is converted into gluconic acid by the enzymatic reaction using glucose oxidase (GOx), and then complexed with a fluorescent boronic acid through the α-hydroxycarboxylate moiety. According to the present strategy, glucose concentration is exclusively determined among other saccharides in aqueous solutions. Copyright

Pd(ii) nanoparticles in porous polystyrene: Factors influencing the nanoparticle size and catalytic properties

Tsvetkova, Irina B.,Matveeva, Valentina G.,Doluda, Valentin Y.,Bykov, Alexei V.,Sidorov, Alexander I.,Schennikov, Sergey V.,Sulman, Michael G.,Valetsky, Pyotr M.,Stein, Barry D.,Chen, Chun-Hsing,Sulman, Esther M.,Bronstein, Lyudmila M.

, p. 6441 - 6448 (2012)

In this paper for the first time we present a systematic study of the influence of hydrophobicity of Pd(ii) compounds, (CH3CN) 2PdCl2, (PhCN)2PdCl2, (Sty)(CH 3CN)PdCl2, and (StyPdCl2)2, on nanoparticle (NP) formation in the pores of hydrophobic micro/mesoporous hypercrosslinked polystyrene (HPS). The morphology and composition of HPS-Pd nanocomposites were studied using transmission electron microscopy, X-ray fluorescence measurements, X-ray photoelectron spectroscopy, and liquid nitrogen physisorption. The size and location of Pd compound NPs were found to depend on hydrophobicity of the Pd(ii) environment. Catalytic testing of these nanocomposites in d-glucose oxidation was carried out to illustrate the influence of nanoparticle size and environment on catalytic activity. The highest catalytic activity was achieved for (Sty)(CH3CN)PdCl 2, forming smallest NPs and allowing an optimal hydrophobicity- hydrophilicity balance with HPS. The Royal Society of Chemistry 2012.

Preparation of Au-Pd/C catalysts by adsorption of metallic species in aqueous phase for selective oxidation

Hermans, Sophie,Deffernez, Aurore,Devillers, Michel

, p. 77 - 82 (2010)

Au/C and Au-Pd/C catalysts were prepared on SX PLUS activated carbon using an adsorption method in which the precursor(s)-support interactions in aqueous solution were sought to be optimized. pH windows where maximum adsorption occurs were identified for four bimetallic cases, varying the incorporation order of the adsorbed metallic precursors and the oxidation state of the first metal introduced. All samples were characterized by XPS and SEM/EDXS, and the above-mentioned parameters were found to have an influence on the surface microstructure of the samples and on the activity of the bimetallic catalysts obtained. This preparation method leads to highly active catalysts for the selective oxidation of glucose, with the activity correlated with high surface Pd/C ratios.

Localized Supramolecular Peptide Self-Assembly Directed by Enzyme-Induced Proton Gradients

Rodon Fores, Jennifer,Martinez Mendez, Miguel Leonardo,Mao, Xiyu,Wagner, Déborah,Schmutz, Marc,Rabineau, Morgane,Lavalle, Philippe,Schaaf, Pierre,Boulmedais, Fouzia,Jierry, Lo?c

, p. 15984 - 15988 (2017)

Electrodes are ideal substrates for surface localized self-assembly processes. Spatiotemporal control over such processes is generally directed through the release of ions generated by redox reactions occurring specifically at the electrode. The so-used gradients of ions proved their effectiveness over the last decade but are in essence limited to material-based electrodes, considerably reducing the scope of applications. Herein is described a strategy to enzymatically generate proton gradients from non-conductive surfaces. In the presence of oxygen, immobilization of glucose oxidase (GOx) on a multilayer film provides a flow of protons through enzymatic oxidation of glucose by GOx. The confined acidic environment located at the solid–liquid interface allows the self-assembly of Fmoc-AA-OH (Fmoc=fluorenylmethyloxycarbonyl and A=alanine) dipeptides into β-sheet nanofibers exclusively from and near the surface. In the absence of oxygen, a multilayer nanoreactor containing GOx and horseradish peroxidase (HRP) similarly induces Fmoc-AA-OH self-assembly.

Catalytic deep eutectic solvents for highly efficient conversion of cellulose to gluconic acid with gluconic acid self-precipitation separation

Liu, Feijie,Xue, Zhimin,Zhao, Xinhui,Mou, Hongyu,He, Jing,Mu, Tiancheng

, p. 6140 - 6143 (2018)

A family of FeCl3·6H2O based catalytic deep eutectic solvents (CDESs) were formed and used for the conversion of cellulose to gluconic acid with high efficiency. More importantly, gluconic acid could be separated from the reaction system by self-precipitation.

Enzyme-immobilized hydrogels to create hypoxia for in vitro cancer cell culture

Dawes, Camron S.,Konig, Heiko,Lin, Chien-Chi

, p. 25 - 34 (2017)

Hypoxia is a critical condition governing many aspects of cellular fate processes. The most common practice in hypoxic cell culture is to maintain cells in an incubator with controlled gas inlet (i.e., hypoxic chamber). Here, we describe the design and characterization of enzyme-immobilized hydrogels to create solution hypoxia under ambient conditions for in vitro cancer cell culture. Specifically, glucose oxidase (GOX) was acrylated and co-polymerized with poly(ethylene glycol)-diacrylate (PEGDA) through photopolymerization to form GOX-immobilized PEG-based hydrogels. We first evaluated the effect of soluble GOX on inducing solution hypoxia (O2??5%) and found that both unmodified and acrylated GOX could sustain hypoxia for at least 24?h even under ambient air condition with constant oxygen diffusion from the air-liquid interface. However, soluble GOX gradually lost its ability to sustain hypoxia after 24?h due to the loss of enzyme activity over time. On the other hand, GOX-immobilized hydrogels were able to create hypoxia within the hydrogel for at least 120?h, potentially due to enhanced protein stabilization by enzyme ‘PEGylation’ and immobilization. As a proof-of-concept, this GOX-immobilized hydrogel system was used to create hypoxia for in vitro culture of Molm14 (acute myeloid leukemia (AML) cell line) and Huh7 (hepatocellular carcinoma (HCC) cell line). Cells cultured in the presence of GOX-immobilized hydrogels remained viable for at least 24?h. The expression of hypoxia associated genes, including carbonic anhydrase 9 (CA9) and lysyl oxidase (LOX), were significantly upregulated in cells cultured with GOX-immobilized hydrogels. These results have demonstrated the potential of using enzyme-immobilized hydrogels to create hypoxic environment for in vitro cancer cell culture.

Ruthenium(III) catalyzed oxidation of sugar alcohols by dichloroisocyanuric acid - A kinetic study

Lakshman Kumar,Venkata Nadh,Radhakrishnamurti

, p. 300 - 307 (2016)

Kinetics of ruthenium(III) catalyzed oxidation of biologically important sugar alcohols (myo-inositol, D-sorbitol, and D-mannitol) by dichloroisocyanuric acid was carried out in aqueous acetic acid - perchloric medium. The reactions were found to be first order in case of oxidant and ruthenium(III). Zero order was observed with the concentrations of sorbitol and mannitol whereas, a positive fractional order was found in the case of inositol concentration. An inverse fractional order was observed with perchloric acid in oxidation of three substrates. Arrhenius parameters were calculated and a plausible mechanism was proposed.

A new route to the considerable enhancement of glucose oxidase (GOx) activity: The simple assembly of a complex from CdTe quantum dots and GOx, and its glucose sensing

Cao, Lihua,Ye, Jian,Tong, Lili,Tang, Bo

, p. 9633 - 9640 (2008)

A new complex consisting of CdTe quantum dots (QDs) and glucose oxidase (GOx) has been facilely assembled to achieve considerably enhanced enzymatic activity and a wide active temperature range of GOx; these characteristics are attributed to the conformational changes of GOx during assembly. The obtained complex can be simultaneously used as a nanosensor for the detection of glucose with high sensitivity. A mechanism is put forward based on the fluorescence quenching of CdTe QDs, which is caused by the hydrogen peroxide (H 2O2) that is produced from the GOx-catalyzed oxidation of glucose. When H2O2 gets to the surface of the CdTe QDs, the electrontransfer reaction happens immediately and H2O2 is reduced to O2, which lies in electron hole traps on CdTe QDs and can be used as a good acceptor, thus forming the nonfluorescent CdTe QDs anion. The produced O2 can further participate in the catalyzed reaction of GOx, forming a cyclic electrontransfer mechanism of glucose oxidation, which is favorable for the whole reaction system. The value of the Michaelis-Menton constant of GOx is estimated to be 0.45 mML-1, which shows the considerably enhanced enzymatic activity measured by far. In addition, the GOx enzyme conjugated on the CdTe QDs possesses better thermal stability at 20-80°C and keeps the maximum activity in the wide range of 40-50°C. Moreover, the simply assembled complex as a nanosensor can sensitively determine glucose in the wide concentration range from micro- to millimolar with the detection limit of 0.10 μM, which could be used for the direct detection of low levels of glucose in biological systems. Therefore, the established method could provide an approach for the assembly of CdTe QDs with other redox enzymes, to realize enhanced enzymatic activity, and to further the design of novel nanosensors applied in biological systems in the future.

Real-time monitoring of mass-transport-related enzymatic reaction kinetics in a nanochannel-array reactor

Li, Su-Juan,Wang, Chen,Wu, Zeng-Qiang,Xu, Jing-Juan,Xia, Xing-Hua,Chen, Hong-Yuan

, p. 10186 - 10194 (2010)

To understand the fundamentals of enzymatic reactions confined in micro-/nanosystems, the construction of a small enzyme reactor coupled with an integrated real-time detection system for monitoring the kinetic information is a significant challenge. Nano-enzyme array reactors were fabricated by covalently linking enzymes to the inner channels of a porous anodic alumina (PAA) membrane. The mechanical stability of this nanodevice enables us to integrate an electrochemical detector for the real-time monitoring of the formation of the enzyme reaction product by sputtering a thin Pt film on one side of the PAA membrane. Because the enzymatic reaction is confined in a limited nanospace, the mass transport of the substrate would influence the reaction kinetics considerably. Therefore, the oxidation of glucose by dissolved oxygen catalyzed by immobilized glucose oxidase was used as a model to investigate the mass-transport-related enzymatic reaction kinetics in confined nanospaces. The activity and stability of the enzyme immobilized in the nanochannels was enhanced. In this nano-enzyme reactor, the enzymatic reaction was controlled by mass transport if the flux was low. With an increase in the flux (e.g., >50 μLmin-1), the enzymatic reaction kinetics became the rate-determining step. This change resulted in the decrease in the conversion efficiency of the nano-enzyme reactor and the apparent Michaelis-Menten constant with an increase in substrate flux. This nanodevice integrated with an electrochemical detector could help to understand the fundamentals of enzymatic reactions confined in nanospaces and provide a platform for the design of highly efficient enzyme reactors. In addition, we believe that such nanodevices will find widespread applications in biosensing, drug screening, and biochemical synthesis.

carba Nicotinamide Adenine Dinucleotide Phosphate: Robust Cofactor for Redox Biocatalysis

D?ring, Manuel,Sieber, Volker,Simon, Robert C.,Tafertshofer, Georg,Zachos, Ioannis

supporting information, p. 14701 - 14706 (2021/05/13)

Here we report a new robust nicotinamide dinucleotide phosphate cofactor analog (carba-NADP+) and its acceptance by many enzymes in the class of oxidoreductases. Replacing one ribose oxygen with a methylene group of the natural NADP+ was found to enhance stability dramatically. Decomposition experiments at moderate and high temperatures with the cofactors showed a drastic increase in half-life time at elevated temperatures since it significantly disfavors hydrolysis of the pyridinium-N?glycoside bond. Overall, more than 27 different oxidoreductases were successfully tested, and a thorough analytical characterization and comparison is given. The cofactor carba-NADP+ opens up the field of redox-biocatalysis under harsh conditions.

A Sweet H2S/H2O2Dual Release System and Specific Protein S-Persulfidation Mediated by Thioglucose/Glucose Oxidase

Li, Xiaolu,Ni, Xiang,Qian, Wei-Jun,Shen, Tun-Li,Xian, Ming

, p. 13325 - 13332 (2021/09/03)

H2S and H2O2 are two redox regulating molecules that play important roles in many physiological and pathological processes. While each of them has distinct biosynthetic pathways and signaling mechanisms, the crosstalk between these two species is also known to cause critical biological responses such as protein S-persulfidation. So far, many chemical tools for the studies of H2S and H2O2 have been developed, such as the donors and sensors for H2S and H2O2. However, these tools are normally targeting single species (e.g., only H2S or only H2O2). As such, the crosstalk and synergetic effects between H2S and H2O2 have hardly been studied with those tools. In this work, we report a unique H2S/H2O2 dual donor system by employing 1-thio-β-d-glucose and glucose oxidase (GOx) as the substrates. This enzymatic system can simultaneously produce H2S and H2O2 in a slow and controllable fashion, without generating any bio-unfriendly byproducts. This system was demonstrated to cause efficient S-persulfidation on proteins. In addition, we expanded the system to thiolactose and thioglucose-disulfide; therefore, additional factors (β-galactosidase and cellular reductants) could be introduced to further control the release of H2S/H2O2. This dual release system should be useful for future research on H2S and H2O2.

Post a RFQ

Enter 15 to 2000 letters.Word count: 0 letters

Attach files(File Format: Jpeg, Jpg, Gif, Png, PDF, PPT, Zip, Rar,Word or Excel Maximum File Size: 3MB)

1

What can I do for you?
Get Best Price

Get Best Price for 526-95-4