Você está na página 1de 18

ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid

 

Ascorbic acid or L-Ascorbic acid, L-xylo-Ascorbic acid, L-threo-hex-2-enoic acid Ȗ-


Lactone is IUPAC name for Vitamin C (ULLMANN¶S Encyclopedia vol. 38).

All known disease called scurvy was known to human from ancient (Egyptians) age
occurred due to deficiency of Vitamin C. But this is not known up to 1753 when James Lind
described it to be cured by dietary mean (ULLMANN¶S Encyclopedia vol. 38).

Later on many peoples were working on Ascorbic acid. In which Szent-Gyorgyi


(1928) first isolated Ascorbic acid form Adrenal gland and from plant tissue by Herbert et al.
(1933). Chemical synthesis of L-ascorbic acid from L-xylosone was achieved. This work also
done by Haworth and Hirst (1933). They also propose the chemical structure for Ascorbic
acid, Reichstein et al. (1933). Biochemical reaction for preparation of Ascorbic from D-
Glucose was introduced by Reichstein and Grössner (1934) and this sequence is adapted for
manufacturing of Ascorbic acid industrially (Hancock and R. Viola, 2001).

This play very important role in the human as conjunctive tissue formation, ion
transportation, and cell protection against free radicals. In plants, act against reactive oxygen
species that are formed from Photosynthesis and Respiratory processes. It also linked with
cell growth, cell cycle and cofactor for many enzymes (Anderson et al. 2004).

Wide range of reactive oxygen species, such as singlet oxygen, superoxide anion and
Hydroxyl radicals which have been implicated with many chronic disorders including cancer
and cardiovascular disease.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid



   

Ascorbic acid is used by human as dietary purpose to fulfill their nutrients


requirement and fight against disease called Scurvy and some fatal disease known to be
cancer and cardiovascular disease. It also act as antioxidant in human body which give free
electron to reactive oxygen formed from hydrogen peroxide.

For this purpose we synthesize ascorbic acid on natural way by means of plant which
gives direct feed to humans. This was also synthesizing by many methods using microbes
such as bacteria and algae by biosynthesis. These microbe used as it is but yield is low so for
this to overcome genetically modified strains (×   
 ,   
 ,
 ,    are used. Using algae we also synthesize Ascorbic acid.
The more well known and commercial adapted pathway is Reichstein process. This is
chemical oriented method have a single fermentation step. Due to its energy consumption,
and required high temperature, pressure for many steps in Ascorbic acid synthesis.

To overcome this intermediate compounds are biosynthetically synthesize to increase


the yield of the ascorbic acid with low energy consumption.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid


       

3.1Microbiological fermentation

In microbiological fermentation different species are used to produce Ascorbic acid


these are Bacteria, and Algae. Different pathway has been proposed to describe how they
produce Ascorbic acid by initial starting materials.

3.1.1 Metabolic Pathway for Bacteria


At present there are six bacterial fermentation processes for the production of 2-keto-
L-gulonic acid, a direct precursor of L-ascorbic acid.
These are a) sorbitol pathway, b) L-idonic acid pathway, c) L-gulonic acid pathway, d) 2-
keto-D-gluconic acid pathway, e) 2, 5-diketo-D-gluconic acid pathway, and f) 2-keto-L-
gulonic acid pathway. The most commercially advanced methods are the oxidation of D-
sorbitol or L-sorbose to 2-keto-L-gulonic acid (2-KLG) (Shrikant et al. 2006) .
The pathway cycle for production of ascorbic acid is given below:

Figure 3.1 Bremus et al. gives different pathway cycle for production Ascorbic acid using Bacteria.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
3.1.1.1 D-Sorbitol pathway
Sorbitol is transformed by fermentation of 2-KLG (2-Keto-L-Gluconic acid) via
intermediate L-Sorbosone. This is done by many strains of bacteria namely Pseudomonas and
Acetobacter, which are catalyze the oxidation of D-Sorbitol to 2-KLG via the series of
membrane bound hydrogenase to produce L-Sorbosone. The final oxidation to 2-KLG is
catalyzed by either membrane bound or cystolic Sorbosone dehydorgenase. Using the strain
Glucano Oxydans that produce up to 60g/l. of 2-KLG from L-Sorbosone or D-Sorbitol with
60% conversion (Hancock and R. Viola, 2001).
Genetically modified strains are also used to carry out above steps. The location of
dehydrogenase required for conversion of D-Sorbitol to 2-KLG varies from strain to strain.
The transfer of D-Sorbitol pathway intermediate into cytoplasm of these strain determined by
the presence of cystolic reductase which give path to the intermediate in the pentose cycle. To
overcome this problem membrane bound dehydrogenase recombinant with Glucanobacter
Oxydans is replace this or substitute cystolic enzymes. For example Acetobacter liquifaciens
a membrane bound Sorbosone dehydrogenase is expressed in Glucanobacter Oxydans which
is membrane bound sorbitol dehydrogenase and sorbose dehydrogenase but not cystolic
dehydrogenase. A significant yield is observed from L-Sorbose (68-81%) and L-Sorbosone
(23-83%). But there is no yield improvement under fermentation condition (Shrikant et al.,
2006).

3.1.1.2 2-Keto-D-Gluconic Acid Pathway


D-Glucose is converted to 2-KLG Ô D-Gluconic acid, 2-Keto-D-Gluconic acid and
2,5-Diketo-D-Gluconic acid (2,5-DKG). Until now there is no bacterial strains capable of
efficiently catalyzing the complete conversion of D-Glcose to 2-KLG have been isolated.
This is carried out in three main steps; each step is carried out by using different
microorganisms.
( ) Transformation of D-glucose to 2-keto-D-gluconic acid: the transformation of glucose to
2-keto-D-gluconic acid is carried out by ×      and   
Some ×  strains also synthesize 2-keto-D-gluconic acid;
( ) Oxidation of 2-keto-D-gluconic acid: this oxidation iscarried out by     
and       with 2, 5-DKG as a product. In addition, ×  
can directly transform D-glucose to 2, 5-DKG;
( ) Oxidation of 2, 5-DKG acid: conversion of 2, 5-DKG into2-KLG. The mentioned strain
is of the genera Ô    and    and maximum yield was obtained from
Ô     The use of   has also been suggested.
c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
A mutant  strain to convert D-glucose into 2, 5-DKG. The whole culture broth
is then treated with sodium dodecyl sulphate to reduce cell viability, mixed with glucose and
then supplied to a mutant strain of     for conversion to 2-KLG. The whole
process achieved yields of 2-KLG of up to 85 % from D-glucose with final 2-KLG
concentrations of 10.5 g/L. bacterial strain Ô genetic engineering. Anderson et al. cloned a
cytosolic 2, 5-DKG reductase gene from     species. And expressed it in
   . The recombinant organism was capable of synthesizing 2-KLG from D-
glucose but the yields attained were very poor (1 g/L of 2-KLG from 20 g/L of D-glucose).
Recombinant    expressing cytoplasmic 2, 5-DKG reductase from    
species. could accumulate
19.8 g/L of 2-KLG with a 49 % conversion efficiency from glucose (Hancock and R. Viola
2001). Such improvements in yield were attained by optimization of the fermentation
conditions, careful selection of the promoter controlling the expression of the 2, 5-DKG
reductase gene and by the use of mutant strains of   which were unable to use 2, 5-
DKG or 2-KLG for growth.

3.1.1.3 Bioconversion Of 2-KLG to L-Ascorbic Acid (Hancock and R. Viola


2001), (Shrikant et al. 2006)

2-KLG is converted to L-ascorbic acid chemically Ô two methods.


The first involves multiple steps including:
‡ Esterification of a 2-KLG derivative under strongly acidic conditions to produce methyl 2-
keto-L-gulonate (MeKLG);
‡ Reaction of MeKLG with a base to produce a metal ascorbate salt;
‡ Treatment of metal ascorbate salt with an acidulant to obtain ascorbic acid.
The second way is a one-step method comprising acid-catalyzed cyclization of KLG.
Both methods are commercially undesirable due to the requirement for multiple chemical
steps for first method, or the use of large amounts of gaseous hydrogen chloride or
requirement for very expensive process equipment for second method.
To overcome this situation hydrolase enzyme is used to convert ester of 2-KLG to L-
Ascorbic acid. In similar way lactonases is extracted from       
 and   
 are capable to convert 2-KLG to L-Ascorbic acid.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid

3.1.2 Metabolic Pathway for Algae

The pathway for the production of ascorbic acid from D-Glucose using Algae is shown
below.

Figure 3.2 Bremus et al. gives (2006) Pathway cycle for production of Ascorbic acid using Algae.

Many attempts are employed to use microalgae for the direct production of L-
Ascorbic acid from inexpensive feedstock. One-step fermentation process for the production
of L-Ascorbic acid using the heterotrophic green microalga     produce
low quantity up to 40 mg/l L-Ascorbic acid.. After continuous chemical mutagenesis and
fermentation optimization, an improved amount of up to 2 g/l L-Ascorbic acid is obtained.
Accumulation of L-Ascorbic acid in the fermentation medium was obtained by the use of the
colorless microalgae    . Using this alga it is shown that a pH reduction
could stabilize L-Ascorbic acid in the fermentation reactor, so most of the L-Ascorbic acid
became harvestable from the medium.

For strain-improvement  used formany mutant strains with increased and
reduced abilities to accumulate L-Ascorbic acid. These mutants are used to identify the
pathway for L-Ascorbic acid biosynthesis that involves mannose-containing intermediates.
Similarly in plants, l-galactono-1, 4,-lactone is produced from d-glucose through GDP-d-
mannose, GDP-l-galactose and l-galactose. Finally l-galactono-1,4,-lactone is converted into
l-AA by l-GL-DH. This is shown by Wheeler et al. (1998) in higher plants.

c
Institute Of Chemical Engineering, Mumbai c
 
cccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccccM t li  t
i i
 t    ti   i i

M t li t l t R i l i t l
]

t l ti i i l i i l :

   
i
    R il 
i !  "t l   ti  # i i i "l t

t t t t i t i i l t i ti l t
l t l
ti t i l
t i t i l t i i ll i t i t i
l t i i t:
i ti t C l ;
i ti tC C ;
i i ti tC ;
t i ti t C C
i M t l t i t t i i ti t i t
M t li M i i li l t l
l tt ti it l t it t t li l t
i li M t i t C i M t M
i t l t MM M t i M

c
  
 tit t  C il 
i i
 M  i c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
P and GTP is catalysed by GDPD- Man pyrophosphorylase (GMP). GDP-D Man is converted
to GDP-L-Gal by a reversible double epimerization, catalysed by GDP-D-Man-3,5-epimerase
(GME) that was first identified in Chlorella pea This enzyme has recently been purified and
cloned from A. thaliana and purified from the alga Prototheca (Running et al. 2004). As well
as being involved in ascorbate synthesis, GDP-D-Man and GDP-L-Gal are substrates for
polysaccharide synthesis and protein glycoslyation. The steps subsequent to GDP-L-Gal are
likely to be dedicated to ascorbate synthesis. GDP-L-Gal is initially broken down to L-Gal 1-
P, which is subsequently hydrolysed to L-Gal. Enzymes catalysing these steps have been
recently purified and characterized. GDP-L-Gal is converted to L-Gal 1-P and GDP by a
novel and highly specific phosphate-dependent GDP-L-Gal phosphorylase. The released L-
Gal is then oxidized in two steps, first by a cytosolic NAD-dependent L-Gal dehydrogenase
(L-GalDH) at C1 to form L-galactono-1,4-lactone (L-GalL) (Wheeler et al. 1998) and then by
L-GalL dehydrogenase (L-GalLDH) at C2/C3 resulting in the production of ascorbate.
The antisense suppression of LGalDH and L-GalLDH decreases ascorbate concentration
Although the D-Man/L-Gal pathway appears to be the predominant pathway, there is some
suggestion that other biosynthetic pathways via uronic acid intermediates contribute to the
ascorbate content of plant tissues and that these may be developmentally regulated.
The authors proposed that the hydrolysis of GDP-L-gulose would result in the production of
L-gulose which could be converted to L-gulono-1,4-lactone by L-GalDH and subsequently
into ascorbate by the L-gulono-1,4-lactone dehydrogenase activity known to exist in plants.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid

3.3 Metabolic Pathway for Animals

The pathway cycle for production of ascorbic acid from D-glucose is given below:

Figure 2.4 Linster and E V Schaftingen (2006) gives Pathway cycle for production of Ascorbic acid in Animals.

Ascorbic acid is synthesized by many vertebrates. The occurrence of ascorbic acid


biosynthesis in sea lamprey suggests that this feature appeared early in the evolutionary
history of fishes (590±500 million years ago), i.e. prior to terrestrial vertebrate emergence [2 .
The biosynthetic capacity has, however, subsequently been lost in a number of species, such
as teleost fishes, passeriform birds, bats, guinea pigs, and primates including humans, for
whom ascorbate has thus become a vitamin. Fish, amphibians and reptiles synthesize
ascorbate in the kidney, whereas mammals produce it in the liver.
In animals, d-glucuronate, derived from UDP-glucuronate, is reduced to l-gulonate, which
leads to inversion of the numbering of the carbon chain (µinversion of configuration¶) since
c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
the aldehyde function of d-glucuronate (C1) becomes a hydroxymethyl group in the resulting
l-gulonate. In plants, the pathway starts with GDP-d-mannose, which is converted (without
change in carbon numbering) to l-galactonolactone, the substrate for the plant homologue of
GLO, l-galactonolactone dehydrogenase.

Formation of glucuronate from UDP-glucuronate (Linster and E. V. Schaftingen


2006)

The formation of glucuronate from UDP-glucuronate could hypothetically involve


following three mechanisms to convert glucuronate from UDP-glucuronate.
(a) The cleavage of UDPglucuronate to glucuronate 1-phosphate, followed by
dephosphorylation of the latter by a glucuronate-1- phosphatase;
(b) The formation of a glucuronidated intermediate followed by its hydrolysis by b-
glucuronidase or esterases (which could hydrolyze acyl-glucuronides); or
(c) Direct hydrolysis of UDP-glucuronate to UDP and glucuronate.
Mechanism (a), It is show that a series of nonglucuronidable xenobiotics rapidly
stimulate the formation of glucuronate in isolated hepatocytes, the same xenobiotics also
stimulated the formation of glucuronate from UDP-glucuronate in liver cell-free extracts
enriched with ATP or in liver microsomes supplemented with ATP and a heat-stable cofactor
identified as coenzyme A. Quantitatively, the synthesis of glucuronate observed under these
conditions accounted for the formation of glucuronate observed in intact cells, indicating that
glucuronate is formed from UDP-glucuronate by a microsomal enzyme. These enzymes are
present in the endoplasmic reticulum as, similarly to UGTs, it is stimulated by UDP-N-
acetylglucosamine, which enhances the transport of UDP-glucuronate into vesicles derived
from the endoplasmic reticulum
Mechanism (b), it is ruled out by the fact that glucuronate formation from UDP-
glucuronate occurs in rat liver microsomes in the absence of UGT substrates and is actually
inhibited by such substrates. Furthermore, inhibitors of ȕ-glucuronidase and esterases do not
affect the formation of glucuronate from UDP-glucuronate by microsomal preparations.
Mechanism (c). Taken together, these observations lead to the conclusion that
glucuronate is formed by direct hydrolysis of UDP-glucuronate by a UDP-glucuronidase. It is
likely that the ability of UGTs to hydrolyze UDP-glucuronate varies among isoforms and
depends on the phospholipidic environment. This last point may explain the observation that
UDPglucuronidase is inhibited in rat liver microsomes by the addition of ATP and coenzyme
A, the latter combination of cofactors could allow the reesterification of lipids in a subcellular
fraction known to contain free fatty acids, acyl-CoA synthetase and acyltransferases.
c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid

Glucuronate reductase (aldehyde reductase) (Linster and E. V. Schaftingen 2006)

The reduction of d-glucuronate to L-gulonate is catalyzed by an NADPH-dependent


reductase, with broad specificity, known as aldehyde reductase or TPN L-hexonate
dehydrogenase and now referred to as aldo-keto reductase for the human enzyme.
The compound D-glucuronate and D-glucuronolactoneare converted by aldehyde reductase to
L-gulonate and L-gulonolactone, respectively. Aldehyde reductase belongs to the large group
of monomeric NADPH-dependent oxidoreductases, known as aldo-keto reductase, which
comprise many members in the human genome, including aldose reductase and
hydroxysteroid dehydrogenases. These enzymes show broad substrate specificities and it
would therefore not be surprising that, besides aldehyde reductase, other members of the
aldo-keto reductase super family participate in the reduction of d-glucuronate. Aldose
reductase appears to be much less efficient than aldehyde reductase in this respect.

Urono- and gulonolactonase:


The conversion of D-glucuronate to L-gulonolactone requires the action of two
enzymes, a reductase and a lactonase, proceeding either via D-glucuronolactone if the
lactonization is the first step, or via l-gulonate if the first reaction is the reduction. Three
different types of lactonases acting on sugar derivatives have been characterized in
mammalian tissues: 6-phosphogluconolactonase, uronolactonase and aldonolactonase. The
first one is an enzyme of the pentose phosphate pathway, which belongs, in mammals, to the
same family of proteins as glucosamine 6-phosphate isomerase and has no direct role to play
in the formation of vitamin C. Uronolactonase, a microsomal enzyme, hydrolyzes D-
glucurono-3,6-lactone, but is inactive against aldonolactones. Aldonolactonase
(gulonolactonase) is also a metal dependent enzyme, acting best with Mn2+, which is present
in the cytosol and hydrolyzes a number of C- and D-lactones of a variety of 5-, 6-, and 7-
carbon aldonates, including L-gulono-1, 4-lactone and D-glucono-1, 5- lactone. It also
catalyzes the lactonization of aldonates, e.g. of L-gulonate, and can therefore participate in
the formation of vitamin C.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
L-Gulonolactone oxidase (Linster and E. V. Schaftingen 2006)

GLO, a microsomal enzyme, catalyzes aerobically the conversion of L-gulonolactone


to L-ascorbate with production of H2 O2. The immediate oxidation product of GLO is 2-keto-
l-gulonolactone, an intermediate that spontaneously isomerizes to L-ascorbate. The preferred
substrate of the enzyme is L-gulono- 1, 4-lactone, but it also acts on L-galactono-, D-
mannono- and d-altrono-1, 4-lactone. Other C-lactones, including L-idono- and D-
gluconolactone, were not oxidized by the enzyme, indicating its configurationally specificity
for the hydroxyl group at C2. The production of H2 O2 is unusual for an enzyme of the
endoplasmic reticulum, and one may wonder if this membrane-bound oxidoreductase does
not transfer its electrons to another acceptor in intact cells, most particularly because its plant
homologues do so. The latter share 30% sequence identity with mammalian GLO and differ
from this enzyme in three main aspects:
(1) They act specifically on l-galactono-1, 4-lactone, which is their physiological substrate;
(2) They are bound to the inner mitochondrial membrane; and
(3) They do not transfer electrons directly to O2, but to cytochrome c.

3.4 Commercial method for Ascorbic acid synthesis

Reichstein Process

The use of Reichstein¶s procedure into an industrial process is marked by great efforts
to improve each reaction step. As a result of many technical and chemical modifications each
step gives over 90% yields. The overall yield of ascorbic acid from d-glucose is now 60%
(ULLMANN¶S Encyclopedia vol. 38).

D-Sorbitol.

The catalytic hydrogenation of D-glucose to D-sorbitol is accomplished at high


pressures and elevated temperatures in the presence of a Raney nickel catalyst. The
hydrogenation is carried out in batch or continuous operations and affords almost quantitative
yields with minimal formation of D-mannitol and L-iditol. After removal of the catalyst, the
sorbitol solution is employed in fermentation without any further purification. Therefore,
manufacturers of ascorbic acid make high demands on the quality of D-glucose; crystalline
dextrose or purified starch hydrolysates are used 8, (ULLMANN¶S Encyclopedia vol. 38).

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
L-Sorbose

L-sorbose is obtained in the pyranose form by microbiological oxidation of sterile


aqueous solutions of D-sorbitol in batch or continuous operations in the presence of air. The
most frequently used strains belong to the Ñ    
   family, which converts
dsorbitol to L-sorbose with more than 90% efficiency. Large-scale fermentations have to be
carried out at pH 4±6 and at 30 ± 35oC under sterile conditions to avoid loss of product during
oxidation and work-up by filtration and crystallization (ULLMANN¶S Encyclopedia vol. 38).

Figure 2.5 Hancock and R Viola (2002) gives Pathway cycle for production of Ascorbic acid by Reichstein
Process.

,3:4,6-Di-O-isopropylidene-M-l-sorbofuranose.

The protection of the 2,3-and 4,6-hydroxyl groups by forming cyclic ketals is


achieved in acetone with excess sulfuric acid as catalyst and dehydrating agent, generally at
low temperatures (e.g., 4C). In the final reaction mixture 2,3:4,6-di- -isopropylidene-M-
lsorbofuranose is obtained as main product. Byproducts are 2,3- -isopropylidene-M-l-
sorbofuranose and 1,2- -isopropylidene-M- l-sorbopyranose. After neutralization, excess
acetone is recovered by distillation. 2, 3:4, 6-di- -isopropylidene-M-l-sorbofuranose is
extracted from the aqueous solution with aromatic solvents (e.g., toluene). The remaining
monoisopropylidenesorboses are also recovered from the aqueous solution and returned to
the Process.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
2, 3:4, 6-Di-O-isopropylidene-2-ketol-gulonic acid.

Originally, the oxidation of 2,3 : 4,6-di- -isopropylidene-M-lsorbofuranose to 2,3 :


4,6-di- -isopropylidene- 2-keto-l-gulonic acid was performed at elevated temperatures in
dilute sodium hydroxide withKMnO4, yielding ca. 90% of product. Much less expensive
oxidation methods are applied in modern continuous processes: sodium hypochlorite,
electrochemical oxidation, or catalytic air oxidation. Oxidation with hypochlorite in the
presence of catalytic amounts of nickel chloride or sulfate at 60C yields !93% of 2,3:4,6-di-
-isopropylidene-2-keto-l-gulonic acid. The active oxidant is presumably nickel peroxide.
Electrochemical oxidation is performed with nickel or nickel oxide electrodes in alkaline
solution. Hydrogen, evolved at the cathodes, can be used for hydrogenation of D-glucose. It
can be advantageous to stop electrochemical conversion to 2,3:4,6-di- -isopropylidene-2-
keto-l-gulonic acid at 90% to avoid a drop in selectivity at the end of electrolysis. Oxidation
can be completed with sodium hypochlorite solution. At the end of the electro oxidation,
selectivity can be improved by electrolytic cells with high electrode surfaces, such as Swiss
roll cells or cells with three-dimensional electrodes. Alternatively, the oxidation has been
performed by using air or oxygen and a metal catalyst in alkaline solution. Good yields are
achieved with palladium or platinum on carbon. The reaction product 2,3:4,6-di- -
isopropylidene-2-keto-l-gulonic acid is isolated by acidification and precipitation as the
monohydrate.

L-Ascorbic acid.

The conversion of 2,3:4,6-di- -isopropylidene-2-keto-l-gulonic acid to l-ascorbic


acid is achieved by two different procedures:

1) Deprotection to give 2-keto-l-gulonic acid, followed by esterification with methanol and


base-catalyzed cyclization.

2) Acid-catalyzed cyclization to ascorbic acid directly from the protected or released 2-keto-l-
gulonic acid.

The starting material for base-catalyzed reactions is methyl 2-keto-l-gulonate,


prepared by treatment of the 2,3:4,6-di- -isopropylidene-2-keto-l-gulonic acid with acidic
methanol. Finally, reaction of the methyl ester with sodium hydrogencarbonate or sodium
acetate affords sodium ascorbate in high yield. The first acid-catalyzed route to ascorbic acid
was published only few years after discovery of the Reichstein process. Today, the industrial
process is performed with an inert solvent (e.g., trichloromethane or toluene) in the presence
c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
of hydrochloric acid. The advantage of this method is that ascorbic acid precipitates from the
mixture as it is formed, minimizing decomposition during reaction. The crude product is
obtained by filtration in high yield and high purity. After dissolution of crude ascorbic acid in
water, impurities are removed by refining with activated carbon, decolorizing resins, or ion
exchange resins, followed by crystallization.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
r

 

Ascorbic acid is synthesized by different pathways using Bacteria, Algae and by


means of chemical reactions called Reichstein process. They all have their merits as well
demerits. In Bacterial biosynthesis the overall yield is 60 g/l. of 2-KLG from L-Sorbose or D-
sorbitol with 60% conversion in D-Sorbitol pathway (Shrikant et al., 2006).while that of 2-
Keto-D-Gluconic Acid Pathway have 85% yield using genetically modified strain
(Glucanobacter Oxydans)(Shrikant et al., 2006) and 10.5 g/l concentration of 2-KLG from D-
Glucose. Using genetically modified strain erwina citerus gives 19.8 g/l of 2-KLG with 49%
conversion (Hancock and R. Viola, 2001). So the D-Sorbitol pathway is more economical
pathway in bacterial biosynthesis of Ascorbic acid.

The other pathway is Reichstein pathway in these process seven steps is included of
which one is fermentation and others are chemical oriented. The yield of each step is 90%
(ULLMANN¶S Encyclopedia vol. 38). Overall yield of the process is 50% (Hancock and R.
Viola, 2001). So that Reichstein process is more favorable for production of Ascorbic acid
industrially.

The intermediate steps in Reichstein process are energy consuming and required high
temperature , pressure condition make it non feasible for synthesis of ascorbic acid. For this
reason the intermediate are synthesize biotechnologically for higher yield and good
conversion.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid

    

1.c Anderson D. Barata-Soares, Maria Luiza P. A. Gomez, Carlos Henrique de Mesquita


and Franco M. Lajolo (2004) Ascorbic acid biosynthesis: a precursor study on plants,
Brazilian Journal of Plant Physiology, 16(3):147-154.

2.c Carole L. Linster and Emile Van Schaftingen (2006) Biosynthesis, recycling and
degradation in mammals, FEBS Journal, 274:1±22.

3.c Christoph Bremus, Ute Herrmann, Stephanie Bringer-Meyer, Hermann Sahm (2006)
The use of microorganisms in l-ascorbic acid production, Journal of Biotechnology,
124:196±205.

4.c Glen L. Wheeler, Mark A. Jones and Nicholas Smirnoff (1998) The Biosynthetic
Pathway of Vitamin C in Higher Plants, Nature, vol. 393:365-369.

5.c J. F. Grindley,T M. A. Payton,T H. Van De Pol, And K. G. Hardyt (1988) Conversion


of Glucose to 2-Keto-L-Gulonate, an Intermediate in L-Ascorbate Synthesis, by a
Recombinant Strain of Erwinia citreus, Applied And Environmental Microbiology,
Vol. 54, No. 7:1770-1775.

6.c Robert D. Hancock and Roberto Viola (2002) Biotechnological approaches for L-
ascorbic acid production, Trends In Biotechnology Vol.20 No.7:299-305.

7.c Robert D. Hancock and Roberto Viola (2001) The use of micro-organisms for L-
ascorbic acid production: current status and future perspectives, Applied
Microbiology Biotechnology, 56:567±576.

c
Institute Of Chemical Engineering, Mumbai c
ccccccccccccccccccccccccccccccccccccccccccc cccccccccccccccccccccccccccccccccc Metabolic pathway engineering for the production of L-ascorbic acid
8.c Shrikant A. Survase, Ishwar B. Bajaj and Rekha S. Singhal (2006) Biotechnological
Production of Vitamins, Food Technology Biotechnology""(3): 381±396.

9.c Takahiro Ishikawa, John Dowdle and Nicholas Smirnoff (2006) Progress in
manipulating ascorbic acid biosynthesis and accumulation in plants, Physiologia
Plantarum, 126: 343±355.

10.cTeruhide Sugisawa, Taro Miyazaki, and Tatsuo Hoshino (2005) Microbial Production
Of L-Ascorbic Acid From D-sorbitol, L-sorbose, L-Glucose, and L-Sorbosone by
Ketogulonicigenium vulgare DSM 4025, Bioscience Biotechnology
Biochemicals,69(3):659-662.

11.cBernd Oster and Ulrich Fechtel, E. Merck, Darmstadt, Federal Republic of Germany,
(2003) ULLMANN¶S Encyclopedia of Industrial Chemistry (Wiley- VCH), sixth edn.
Vol. 38:218-231.

c
Institute Of Chemical Engineering, Mumbai c

Você também pode gostar