fbpx
Wikipedia

Glycolysis

Glycolysis is the metabolic pathway that converts glucose (C6H12O6) into pyruvate and, in most organisms, occurs in the liquid part of cells (the cytosol). The free energy released in this process is used to form the high-energy molecules adenosine triphosphate (ATP) and reduced nicotinamide adenine dinucleotide (NADH).[1] Glycolysis is a sequence of ten reactions catalyzed by enzymes.

The metabolic pathway of glycolysis converts glucose to pyruvate via a series of intermediate metabolites.    Each chemical modification is performed by a different enzyme.    Steps 1 and 3 consume ATP and    steps 7 and 10 produce ATP. Since steps 6–10 occur twice per glucose molecule, this leads to a net production of ATP.
Summary of aerobic respiration
Summary of the 10 reactions of the glycolysis pathway

The wide occurrence of glycolysis in other species indicates that it is an ancient metabolic pathway.[2] Indeed, the reactions that make up glycolysis and its parallel pathway, the pentose phosphate pathway, can occur in the oxygen-free conditions of the Archean oceans, also in the absence of enzymes, catalyzed by metal ions, meaning this is a plausible prebiotic pathway for abiogenesis.[3]

The most common type of glycolysis is the Embden–Meyerhof–Parnas (EMP) pathway, which was discovered by Gustav Embden, Otto Meyerhof, and Jakub Karol Parnas. Glycolysis also refers to other pathways, such as the Entner–Doudoroff pathway and various heterofermentative and homofermentative pathways. However, the discussion here will be limited to the Embden–Meyerhof–Parnas pathway.[4]

The glycolysis pathway can be separated into two phases:[5]

  1. Investment phase – wherein ATP is consumed
  2. Yield phase – wherein more ATP is produced than originally consumed

Overview edit

The overall reaction of glycolysis is:

 

+ 2 [NAD]+
+ 2 [ADP]
+ 2 [P]i

 

 

2 × Pyruvate

2 ×   

 

+ 2 [NADH]
+ 2 H+
+ 2 [ATP]
+ 2 H2O
 
Glycolysis pathway overview.

The use of symbols in this equation makes it appear unbalanced with respect to oxygen atoms, hydrogen atoms, and charges. Atom balance is maintained by the two phosphate (Pi) groups:[6]

  • Each exists in the form of a hydrogen phosphate anion ([HPO4]2−), dissociating to contribute 2H+ overall
  • Each liberates an oxygen atom when it binds to an adenosine diphosphate (ADP) molecule, contributing 2 O overall

Charges are balanced by the difference between ADP and ATP. In the cellular environment, all three hydroxyl groups of ADP dissociate into −O and H+, giving ADP3−, and this ion tends to exist in an ionic bond with Mg2+, giving ADPMg. ATP behaves identically except that it has four hydroxyl groups, giving ATPMg2−. When these differences along with the true charges on the two phosphate groups are considered together, the net charges of −4 on each side are balanced.

For simple fermentations, the metabolism of one molecule of glucose to two molecules of pyruvate has a net yield of two molecules of ATP. Most cells will then carry out further reactions to "repay" the used NAD+ and produce a final product of ethanol or lactic acid. Many bacteria use inorganic compounds as hydrogen acceptors to regenerate the NAD+.

Cells performing aerobic respiration synthesize much more ATP, but not as part of glycolysis. These further aerobic reactions use pyruvate, and NADH + H+ from glycolysis. Eukaryotic aerobic respiration produces approximately 34 additional molecules of ATP for each glucose molecule, however most of these are produced by a mechanism vastly different from the substrate-level phosphorylation in glycolysis.

The lower-energy production, per glucose, of anaerobic respiration relative to aerobic respiration, results in greater flux through the pathway under hypoxic (low-oxygen) conditions, unless alternative sources of anaerobically oxidizable substrates, such as fatty acids, are found.

History edit

The pathway of glycolysis as it is known today took almost 100 years to fully elucidate.[7] The combined results of many smaller experiments were required in order to understand the pathway as a whole.

The first steps in understanding glycolysis began in the nineteenth century with the wine industry. For economic reasons, the French wine industry sought to investigate why wine sometimes turned distasteful, instead of fermenting into alcohol. French scientist Louis Pasteur researched this issue during the 1850s, and the results of his experiments began the long road to elucidating the pathway of glycolysis.[8] His experiments showed that fermentation occurs by the action of living microorganisms, yeasts, and that yeast's glucose consumption decreased under aerobic conditions of fermentation, in comparison to anaerobic conditions (the Pasteur effect).[9]

 
Eduard Buchner. Discovered cell-free fermentation.

Insight into the component steps of glycolysis were provided by the non-cellular fermentation experiments of Eduard Buchner during the 1890s.[10][11] Buchner demonstrated that the conversion of glucose to ethanol was possible using a non-living extract of yeast, due to the action of enzymes in the extract.[12]: 135–148  This experiment not only revolutionized biochemistry, but also allowed later scientists to analyze this pathway in a more controlled laboratory setting. In a series of experiments (1905-1911), scientists Arthur Harden and William Young discovered more pieces of glycolysis.[13] They discovered the regulatory effects of ATP on glucose consumption during alcohol fermentation. They also shed light on the role of one compound as a glycolysis intermediate: fructose 1,6-bisphosphate.[12]: 151–158 

The elucidation of fructose 1,6-bisphosphate was accomplished by measuring CO2 levels when yeast juice was incubated with glucose. CO2 production increased rapidly then slowed down. Harden and Young noted that this process would restart if an inorganic phosphate (Pi) was added to the mixture. Harden and Young deduced that this process produced organic phosphate esters, and further experiments allowed them to extract fructose diphosphate (F-1,6-DP).

Arthur Harden and William Young along with Nick Sheppard determined, in a second experiment, that a heat-sensitive high-molecular-weight subcellular fraction (the enzymes) and a heat-insensitive low-molecular-weight cytoplasm fraction (ADP, ATP and NAD+ and other cofactors) are required together for fermentation to proceed. This experiment begun by observing that dialyzed (purified) yeast juice could not ferment or even create a sugar phosphate. This mixture was rescued with the addition of undialyzed yeast extract that had been boiled. Boiling the yeast extract renders all proteins inactive (as it denatures them). The ability of boiled extract plus dialyzed juice to complete fermentation suggests that the cofactors were non-protein in character.[13]

 
Otto Meyerhof. One of the main scientists involved in completing the puzzle of glycolysis

In the 1920s Otto Meyerhof was able to link together some of the many individual pieces of glycolysis discovered by Buchner, Harden, and Young. Meyerhof and his team were able to extract different glycolytic enzymes from muscle tissue, and combine them to artificially create the pathway from glycogen to lactic acid.[14][15]

In one paper, Meyerhof and scientist Renate Junowicz-Kockolaty investigated the reaction that splits fructose 1,6-diphosphate into the two triose phosphates. Previous work proposed that the split occurred via 1,3-diphosphoglyceraldehyde plus an oxidizing enzyme and cozymase. Meyerhoff and Junowicz found that the equilibrium constant for the isomerase and aldoses reaction were not affected by inorganic phosphates or any other cozymase or oxidizing enzymes. They further removed diphosphoglyceraldehyde as a possible intermediate in glycolysis.[15]

With all of these pieces available by the 1930s, Gustav Embden proposed a detailed, step-by-step outline of that pathway we now know as glycolysis.[16] The biggest difficulties in determining the intricacies of the pathway were due to the very short lifetime and low steady-state concentrations of the intermediates of the fast glycolytic reactions. By the 1940s, Meyerhof, Embden and many other biochemists had finally completed the puzzle of glycolysis.[15] The understanding of the isolated pathway has been expanded in the subsequent decades, to include further details of its regulation and integration with other metabolic pathways.

Sequence of reactions edit

Summary of reactions edit


Preparatory phase edit

The first five steps of Glycolysis are regarded as the preparatory (or investment) phase, since they consume energy to convert the glucose into two three-carbon sugar phosphates[5] (G3P).

d-Glucose (Glc) Hexokinase glucokinase (HK)
a transferase
α-d-Glucose-6-phosphate (G6P)
     
ATP H+ + ADP
 
 
 

Once glucose enters the cell, the first step is phosphorylation of glucose by a family of enzymes called hexokinases to form glucose 6-phosphate (G6P). This reaction consumes ATP, but it acts to keep the glucose concentration inside the cell low, promoting continuous transport of blood glucose into the cell through the plasma membrane transporters. In addition, phosphorylation blocks the glucose from leaking out – the cell lacks transporters for G6P, and free diffusion out of the cell is prevented due to the charged nature of G6P. Glucose may alternatively be formed from the phosphorolysis or hydrolysis of intracellular starch or glycogen.

In animals, an isozyme of hexokinase called glucokinase is also used in the liver, which has a much lower affinity for glucose (Km in the vicinity of normal glycemia), and differs in regulatory properties. The different substrate affinity and alternate regulation of this enzyme are a reflection of the role of the liver in maintaining blood sugar levels.

Cofactors: Mg2+


α-d-Glucose 6-phosphate (G6P) Phosphoglucoisomerase (PGI)
an isomerase
β-d-Fructose 6-phosphate (F6P)
     
 
 
 

G6P is then rearranged into fructose 6-phosphate (F6P) by glucose phosphate isomerase. Fructose can also enter the glycolytic pathway by phosphorylation at this point.

The change in structure is an isomerization, in which the G6P has been converted to F6P. The reaction requires an enzyme, phosphoglucose isomerase, to proceed. This reaction is freely reversible under normal cell conditions. However, it is often driven forward because of a low concentration of F6P, which is constantly consumed during the next step of glycolysis. Under conditions of high F6P concentration, this reaction readily runs in reverse. This phenomenon can be explained through Le Chatelier's Principle. Isomerization to a keto sugar is necessary for carbanion stabilization in the fourth reaction step (below).


β-d-Fructose 6-phosphate (F6P) Phosphofructokinase (PFK-1)
a transferase
β-d-Fructose 1,6-bisphosphate (F1,6BP)
     
ATP H+ + ADP
 
 
 

The energy expenditure of another ATP in this step is justified in 2 ways: The glycolytic process (up to this step) becomes irreversible, and the energy supplied destabilizes the molecule. Because the reaction catalyzed by phosphofructokinase 1 (PFK-1) is coupled to the hydrolysis of ATP (an energetically favorable step) it is, in essence, irreversible, and a different pathway must be used to do the reverse conversion during gluconeogenesis. This makes the reaction a key regulatory point (see below).

Furthermore, the second phosphorylation event is necessary to allow the formation of two charged groups (rather than only one) in the subsequent step of glycolysis, ensuring the prevention of free diffusion of substrates out of the cell.

The same reaction can also be catalyzed by pyrophosphate-dependent phosphofructokinase (PFP or PPi-PFK), which is found in most plants, some bacteria, archea, and protists, but not in animals. This enzyme uses pyrophosphate (PPi) as a phosphate donor instead of ATP. It is a reversible reaction, increasing the flexibility of glycolytic metabolism.[17] A rarer ADP-dependent PFK enzyme variant has been identified in archaean species.[18]

Cofactors: Mg2+


Destabilizing the molecule in the previous reaction allows the hexose ring to be split by aldolase into two triose sugars: dihydroxyacetone phosphate (a ketose), and glyceraldehyde 3-phosphate (an aldose). There are two classes of aldolases: class I aldolases, present in animals and plants, and class II aldolases, present in fungi and bacteria; the two classes use different mechanisms in cleaving the ketose ring.

Electrons delocalized in the carbon-carbon bond cleavage associate with the alcohol group. The resulting carbanion is stabilized by the structure of the carbanion itself via resonance charge distribution and by the presence of a charged ion prosthetic group.


Dihydroxyacetone phosphate (DHAP) Triosephosphate isomerase (TPI)
an isomerase
d-Glyceraldehyde 3-phosphate (GADP)
     
 
 
 

Triosephosphate isomerase rapidly interconverts dihydroxyacetone phosphate with glyceraldehyde 3-phosphate (GADP) that proceeds further into glycolysis. This is advantageous, as it directs dihydroxyacetone phosphate down the same pathway as glyceraldehyde 3-phosphate, simplifying regulation.

Pay-off phase edit

The second half of glycolysis is known as the pay-off phase, characterised by a net gain of the energy-rich molecules ATP and NADH.[5] Since glucose leads to two triose sugars in the preparatory phase, each reaction in the pay-off phase occurs twice per glucose molecule. This yields 2 NADH molecules and 4 ATP molecules, leading to a net gain of 2 NADH molecules and 2 ATP molecules from the glycolytic pathway per glucose.

Glyceraldehyde 3-phosphate (GADP) Glyceraldehyde phosphate dehydrogenase (GAPDH)
an oxidoreductase
d-1,3-Bisphosphoglycerate (1,3BPG)
     
NAD+ + Pi NADH + H+
 
   
 
 

The aldehyde groups of the triose sugars are oxidised, and inorganic phosphate is added to them, forming 1,3-bisphosphoglycerate.

The hydrogen is used to reduce two molecules of NAD+, a hydrogen carrier, to give NADH + H+ for each triose.

Hydrogen atom balance and charge balance are both maintained because the phosphate (Pi) group actually exists in the form of a hydrogen phosphate anion (HPO2−4),[6] which dissociates to contribute the extra H+ ion and gives a net charge of -3 on both sides.

Here, arsenate ([AsO4]3−), an anion akin to inorganic phosphate may replace phosphate as a substrate to form 1-arseno-3-phosphoglycerate. This, however, is unstable and readily hydrolyzes to form 3-phosphoglycerate, the intermediate in the next step of the pathway. As a consequence of bypassing this step, the molecule of ATP generated from 1-3 bisphosphoglycerate in the next reaction will not be made, even though the reaction proceeds. As a result, arsenate is an uncoupler of glycolysis.[19]


This step is the enzymatic transfer of a phosphate group from 1,3-bisphosphoglycerate to ADP by phosphoglycerate kinase, forming ATP and 3-phosphoglycerate. At this step, glycolysis has reached the break-even point: 2 molecules of ATP were consumed, and 2 new molecules have now been synthesized. This step, one of the two substrate-level phosphorylation steps, requires ADP; thus, when the cell has plenty of ATP (and little ADP), this reaction does not occur. Because ATP decays relatively quickly when it is not metabolized, this is an important regulatory point in the glycolytic pathway.

ADP actually exists as ADPMg, and ATP as ATPMg2−, balancing the charges at −5 both sides.

Cofactors: Mg2+


Phosphoglycerate mutase isomerises 3-phosphoglycerate into 2-phosphoglycerate.


2-Phosphoglycerate (2PG) Enolase (ENO)
a lyase
Phosphoenolpyruvate (PEP)
     
H2O
 
 
 
  Enolase (ENO)

Enolase next converts 2-phosphoglycerate to phosphoenolpyruvate. This reaction is an elimination reaction involving an E1cB mechanism.

Cofactors: 2 Mg2+, one "conformational" ion to coordinate with the carboxylate group of the substrate, and one "catalytic" ion that participates in the dehydration.


Phosphoenolpyruvate (PEP) Pyruvate kinase (PK)
a transferase
Pyruvate (Pyr)
     
ADP + H+ ATP
 
 
 

A final substrate-level phosphorylation now forms a molecule of pyruvate and a molecule of ATP by means of the enzyme pyruvate kinase. This serves as an additional regulatory step, similar to the phosphoglycerate kinase step.

Cofactors: Mg2+

Biochemical logic edit

The existence of more than one point of regulation indicates that intermediates between those points enter and leave the glycolysis pathway by other processes. For example, in the first regulated step, hexokinase converts glucose into glucose-6-phosphate. Instead of continuing through the glycolysis pathway, this intermediate can be converted into glucose storage molecules, such as glycogen or starch. The reverse reaction, breaking down, e.g., glycogen, produces mainly glucose-6-phosphate; very little free glucose is formed in the reaction. The glucose-6-phosphate so produced can enter glycolysis after the first control point.

In the second regulated step (the third step of glycolysis), phosphofructokinase converts fructose-6-phosphate into fructose-1,6-bisphosphate, which then is converted into glyceraldehyde-3-phosphate and dihydroxyacetone phosphate. The dihydroxyacetone phosphate can be removed from glycolysis by conversion into glycerol-3-phosphate, which can be used to form triglycerides.[20] Conversely, triglycerides can be broken down into fatty acids and glycerol; the latter, in turn, can be converted into dihydroxyacetone phosphate, which can enter glycolysis after the second control point.

Free energy changes edit

Concentrations of metabolites in erythrocytes[21]: 584 
Compound Concentration / mM
Glucose 5.0
Glucose-6-phosphate 0.083
Fructose-6-phosphate 0.014
Fructose-1,6-bisphosphate 0.031
Dihydroxyacetone phosphate 0.14
Glyceraldehyde-3-phosphate 0.019
1,3-Bisphosphoglycerate 0.001
2,3-Bisphosphoglycerate 4.0
3-Phosphoglycerate 0.12
2-Phosphoglycerate 0.03
Phosphoenolpyruvate 0.023
Pyruvate 0.051
ATP 1.85
ADP 0.14
Pi 1.0

The change in free energy, ΔG, for each step in the glycolysis pathway can be calculated using ΔG = ΔG°′ + RTln Q, where Q is the reaction quotient. This requires knowing the concentrations of the metabolites. All of these values are available for erythrocytes, with the exception of the concentrations of NAD+ and NADH. The ratio of NAD+ to NADH in the cytoplasm is approximately 1000, which makes the oxidation of glyceraldehyde-3-phosphate (step 6) more favourable.

Using the measured concentrations of each step, and the standard free energy changes, the actual free energy change can be calculated. (Neglecting this is very common - the delta G of ATP hydrolysis in cells is not the standard free energy change of ATP hydrolysis quoted in textbooks).

Change in free energy for each step of glycolysis[21]: 582–583 
Step Reaction ΔG°′
(kJ/mol)
ΔG
(kJ/mol)
1 Glucose + ATP4− → Glucose-6-phosphate2− + ADP3− + H+ −16.7 −34
2 Glucose-6-phosphate2− → Fructose-6-phosphate2− 1.67 −2.9
3 Fructose-6-phosphate2− + ATP4− → Fructose-1,6-bisphosphate4− + ADP3− + H+ −14.2 −19
4 Fructose-1,6-bisphosphate4− → Dihydroxyacetone phosphate2− + Glyceraldehyde-3-phosphate2− 23.9 −0.23
5 Dihydroxyacetone phosphate2− → Glyceraldehyde-3-phosphate2− 7.56 2.4
6 Glyceraldehyde-3-phosphate2− + Pi2− + NAD+ → 1,3-Bisphosphoglycerate4− + NADH + H+ 6.30 −1.29
7 1,3-Bisphosphoglycerate4− + ADP3− → 3-Phosphoglycerate3− + ATP4− −18.9 0.09
8 3-Phosphoglycerate3− → 2-Phosphoglycerate3− 4.4 0.83
9 2-Phosphoglycerate3− → Phosphoenolpyruvate3− + H2O 1.8 1.1
10 Phosphoenolpyruvate3− + ADP3− + H+ → Pyruvate + ATP4− −31.7 −23.0

From measuring the physiological concentrations of metabolites in an erythrocyte it seems that about seven of the steps in glycolysis are in equilibrium for that cell type. Three of the steps — the ones with large negative free energy changes — are not in equilibrium and are referred to as irreversible; such steps are often subject to regulation.

Step 5 in the figure is shown behind the other steps, because that step is a side-reaction that can decrease or increase the concentration of the intermediate glyceraldehyde-3-phosphate. That compound is converted to dihydroxyacetone phosphate by the enzyme triose phosphate isomerase, which is a catalytically perfect enzyme; its rate is so fast that the reaction can be assumed to be in equilibrium. The fact that ΔG is not zero indicates that the actual concentrations in the erythrocyte are not accurately known.

Regulation edit

The enzymes that catalyse glycolysis are regulated via a range of biological mechanisms in order to control overall flux though the pathway. This is vital for both homeostatsis in a static environment, and metabolic adaptation to a changing environment or need.[22] The details of regulation for some enzymes are highly conserved between species, whereas others vary widely.[23][24]

  1. Gene Expression: Firstly, the cellular concentrations of glycolytic enzymes are modulated via regulation of gene expression via transcription factors,[25] with several glycolysis enzymes themselves acting as regulatory protein kinases in the nucleus.[26]
  2. Allosteric inhibition and activation by metabolites: In particular end-product inhibition of regulated enzymes by metabolites such as ATP serves as negative feedback regulation of the pathway.[23][27]
  3. Allosteric inhibition and activation by Protein-protein interactions (PPI).[28] Indeed, some proteins interact with and regulate multiple glycolytic enzymes.[29]
  4. Post-translational modification (PTM).[30] In particular, phosphorylation and dephosphorylation is a key mechanism of regulation of pyruvate kinase in the liver.
  5. Localization[27]

Regulation by insulin in animals edit

In animals, regulation of blood glucose levels by the pancreas in conjunction with the liver is a vital part of homeostasis. The beta cells in the pancreatic islets are sensitive to the blood glucose concentration.[31] A rise in the blood glucose concentration causes them to release insulin into the blood, which has an effect particularly on the liver, but also on fat and muscle cells, causing these tissues to remove glucose from the blood. When the blood sugar falls the pancreatic beta cells cease insulin production, but, instead, stimulate the neighboring pancreatic alpha cells to release glucagon into the blood.[31] This, in turn, causes the liver to release glucose into the blood by breaking down stored glycogen, and by means of gluconeogenesis. If the fall in the blood glucose level is particularly rapid or severe, other glucose sensors cause the release of epinephrine from the adrenal glands into the blood. This has the same action as glucagon on glucose metabolism, but its effect is more pronounced.[31] In the liver glucagon and epinephrine cause the phosphorylation of the key, regulated enzymes of glycolysis, fatty acid synthesis, cholesterol synthesis, gluconeogenesis, and glycogenolysis. Insulin has the opposite effect on these enzymes.[32] The phosphorylation and dephosphorylation of these enzymes (ultimately in response to the glucose level in the blood) is the dominant manner by which these pathways are controlled in the liver, fat, and muscle cells. Thus the phosphorylation of phosphofructokinase inhibits glycolysis, whereas its dephosphorylation through the action of insulin stimulates glycolysis.[32]

Regulated Enzymes in Glycolysis edit

The three regulatory enzymes are hexokinase (or glucokinase in the liver), phosphofructokinase, and pyruvate kinase. The flux through the glycolytic pathway is adjusted in response to conditions both inside and outside the cell. The internal factors that regulate glycolysis do so primarily to provide ATP in adequate quantities for the cell's needs. The external factors act primarily on the liver, fat tissue, and muscles, which can remove large quantities of glucose from the blood after meals (thus preventing hyperglycemia by storing the excess glucose as fat or glycogen, depending on the tissue type). The liver is also capable of releasing glucose into the blood between meals, during fasting, and exercise thus preventing hypoglycemia by means of glycogenolysis and gluconeogenesis. These latter reactions coincide with the halting of glycolysis in the liver.

In addition hexokinase and glucokinase act independently of the hormonal effects as controls at the entry points of glucose into the cells of different tissues. Hexokinase responds to the glucose-6-phosphate (G6P) level in the cell, or, in the case of glucokinase, to the blood sugar level in the blood to impart entirely intracellular controls of the glycolytic pathway in different tissues (see below).[32]

When glucose has been converted into G6P by hexokinase or glucokinase, it can either be converted to glucose-1-phosphate (G1P) for conversion to glycogen, or it is alternatively converted by glycolysis to pyruvate, which enters the mitochondrion where it is converted into acetyl-CoA and then into citrate. Excess citrate is exported from the mitochondrion back into the cytosol, where ATP citrate lyase regenerates acetyl-CoA and oxaloacetate (OAA). The acetyl-CoA is then used for fatty acid synthesis and cholesterol synthesis, two important ways of utilizing excess glucose when its concentration is high in blood. The regulated enzymes catalyzing these reactions perform these functions when they have been dephosphorylated through the action of insulin on the liver cells. Between meals, during fasting, exercise or hypoglycemia, glucagon and epinephrine are released into the blood. This causes liver glycogen to be converted back to G6P, and then converted to glucose by the liver-specific enzyme glucose 6-phosphatase and released into the blood. Glucagon and epinephrine also stimulate gluconeogenesis, which coverts non-carbohydrate substrates into G6P, which joins the G6P derived from glycogen, or substitutes for it when the liver glycogen store have been depleted. This is critical for brain function, since the brain utilizes glucose as an energy source under most conditions.[33] The simultaneously phosphorylation of, particularly, phosphofructokinase, but also, to a certain extent pyruvate kinase, prevents glycolysis occurring at the same time as gluconeogenesis and glycogenolysis.

Hexokinase and glucokinase edit

 
Yeast hexokinase B (PDB: 1IG8​)

All cells contain the enzyme hexokinase, which catalyzes the conversion of glucose that has entered the cell into glucose-6-phosphate (G6P). Since the cell membrane is impervious to G6P, hexokinase essentially acts to transport glucose into the cells from which it can then no longer escape. Hexokinase is inhibited by high levels of G6P in the cell. Thus the rate of entry of glucose into cells partially depends on how fast G6P can be disposed of by glycolysis, and by glycogen synthesis (in the cells which store glycogen, namely liver and muscles).[32][34]

Glucokinase, unlike hexokinase, is not inhibited by G6P. It occurs in liver cells, and will only phosphorylate the glucose entering the cell to form glucose-6-phosphate (G6P), when the glucose in the blood is abundant. This being the first step in the glycolytic pathway in the liver, it therefore imparts an additional layer of control of the glycolytic pathway in this organ.[32]

Phosphofructokinase edit

 
Bacillus stearothermophilus phosphofructokinase (PDB: 6PFK​)

Phosphofructokinase is an important control point in the glycolytic pathway, since it is one of the irreversible steps and has key allosteric effectors, AMP and fructose 2,6-bisphosphate (F2,6BP).

Fructose 2,6-bisphosphate (F2,6BP) is a very potent activator of phosphofructokinase (PFK-1) that is synthesized when F6P is phosphorylated by a second phosphofructokinase (PFK2). In the liver, when blood sugar is low and glucagon elevates cAMP, PFK2 is phosphorylated by protein kinase A. The phosphorylation inactivates PFK2, and another domain on this protein becomes active as fructose bisphosphatase-2, which converts F2,6BP back to F6P. Both glucagon and epinephrine cause high levels of cAMP in the liver. The result of lower levels of liver fructose-2,6-bisphosphate is a decrease in activity of phosphofructokinase and an increase in activity of fructose 1,6-bisphosphatase, so that gluconeogenesis (in essence, "glycolysis in reverse") is favored. This is consistent with the role of the liver in such situations, since the response of the liver to these hormones is to release glucose to the blood.

ATP competes with AMP for the allosteric effector site on the PFK enzyme. ATP concentrations in cells are much higher than those of AMP, typically 100-fold higher,[35] but the concentration of ATP does not change more than about 10% under physiological conditions, whereas a 10% drop in ATP results in a 6-fold increase in AMP.[36] Thus, the relevance of ATP as an allosteric effector is questionable. An increase in AMP is a consequence of a decrease in energy charge in the cell.

Citrate inhibits phosphofructokinase when tested in vitro by enhancing the inhibitory effect of ATP. However, it is doubtful that this is a meaningful effect in vivo, because citrate in the cytosol is utilized mainly for conversion to acetyl-CoA for fatty acid and cholesterol synthesis.

TIGAR, a p53 induced enzyme, is responsible for the regulation of phosphofructokinase and acts to protect against oxidative stress.[37] TIGAR is a single enzyme with dual function that regulates F2,6BP. It can behave as a phosphatase (fructuose-2,6-bisphosphatase) which cleaves the phosphate at carbon-2 producing F6P. It can also behave as a kinase (PFK2) adding a phosphate onto carbon-2 of F6P which produces F2,6BP. In humans, the TIGAR protein is encoded by C12orf5 gene. The TIGAR enzyme will hinder the forward progression of glycolysis, by creating a build up of fructose-6-phosphate (F6P) which is isomerized into glucose-6-phosphate (G6P). The accumulation of G6P will shunt carbons into the pentose phosphate pathway.[38][39]

Pyruvate kinase edit

 
Yeast pyruvate kinase (PDB: 1A3W​)

The final step of glycolysis is catalysed by pyruvate kinase to form pyruvate and another ATP. It is regulated by a range of different transcriptional, covalent and non-covalent regulation mechanisms, which can vary widely in different tissues.[40][41][42] For example, in the liver, pyruvate kinase is regulated based on glucose availability. During fasting (no glucose available), glucagon activates protein kinase A which phosphorylates pyruvate kinase to inhibit it.[43] An increase in blood sugar leads to secretion of insulin, which activates protein phosphatase 1, leading to dephosphorylation and re-activation of pyruvate kinase.[43] These controls prevent pyruvate kinase from being active at the same time as the enzymes that catalyze the reverse reaction (pyruvate carboxylase and phosphoenolpyruvate carboxykinase), preventing a futile cycle.[43] Conversely, the isoform of pyruvate kinasein found in muscle is not affected by protein kinase A (which is activated by adrenaline in that tissue), so that glycolysis remains active in muscles even during fasting.[43]

Post-glycolysis processes edit

The overall process of glycolysis is:

Glucose + 2 NAD+ + 2 ADP + 2 Pi → 2 pyruvate + 2 NADH + 2 H+ + 2 ATP

If glycolysis were to continue indefinitely, all of the NAD+ would be used up, and glycolysis would stop. To allow glycolysis to continue, organisms must be able to oxidize NADH back to NAD+. How this is performed depends on which external electron acceptor is available.

Anoxic regeneration of NAD+ edit

One method of doing this is to simply have the pyruvate do the oxidation; in this process, pyruvate is converted to lactate (the conjugate base of lactic acid) in a process called lactic acid fermentation:

Pyruvate + NADH + H+ → lactate + NAD+

This process occurs in the bacteria involved in making yogurt (the lactic acid causes the milk to curdle). This process also occurs in animals under hypoxic (or partially anaerobic) conditions, found, for example, in overworked muscles that are starved of oxygen. In many tissues, this is a cellular last resort for energy; most animal tissue cannot tolerate anaerobic conditions for an extended period of time.

Some organisms, such as yeast, convert NADH back to NAD+ in a process called ethanol fermentation. In this process, the pyruvate is converted first to acetaldehyde and carbon dioxide, and then to ethanol.

Lactic acid fermentation and ethanol fermentation can occur in the absence of oxygen. This anaerobic fermentation allows many single-cell organisms to use glycolysis as their only energy source.

Anoxic regeneration of NAD+ is only an effective means of energy production during short, intense exercise in vertebrates, for a period ranging from 10 seconds to 2 minutes during a maximal effort in humans. (At lower exercise intensities it can sustain muscle activity in diving animals, such as seals, whales and other aquatic vertebrates, for very much longer periods of time.) Under these conditions NAD+ is replenished by NADH donating its electrons to pyruvate to form lactate. This produces 2 ATP molecules per glucose molecule, or about 5% of glucose's energy potential (38 ATP molecules in bacteria). But the speed at which ATP is produced in this manner is about 100 times that of oxidative phosphorylation. The pH in the cytoplasm quickly drops when hydrogen ions accumulate in the muscle, eventually inhibiting the enzymes involved in glycolysis.

The burning sensation in muscles during hard exercise can be attributed to the release of hydrogen ions during the shift to glucose fermentation from glucose oxidation to carbon dioxide and water, when aerobic metabolism can no longer keep pace with the energy demands of the muscles. These hydrogen ions form a part of lactic acid. The body falls back on this less efficient but faster method of producing ATP under low oxygen conditions. This is thought to have been the primary means of energy production in earlier organisms before oxygen reached high concentrations in the atmosphere between 2000 and 2500 million years ago, and thus would represent a more ancient form of energy production than the aerobic replenishment of NAD+ in cells.

The liver in mammals gets rid of this excess lactate by transforming it back into pyruvate under aerobic conditions; see Cori cycle.

Fermentation of pyruvate to lactate is sometimes also called "anaerobic glycolysis", however, glycolysis ends with the production of pyruvate regardless of the presence or absence of oxygen.

In the above two examples of fermentation, NADH is oxidized by transferring two electrons to pyruvate. However, anaerobic bacteria use a wide variety of compounds as the terminal electron acceptors in cellular respiration: nitrogenous compounds, such as nitrates and nitrites; sulfur compounds, such as sulfates, sulfites, sulfur dioxide, and elemental sulfur; carbon dioxide; iron compounds; manganese compounds; cobalt compounds; and uranium compounds.

Aerobic regeneration of NAD+ and further catabolism of pyruvate edit

In aerobic eukaryotes, a complex mechanism has developed to use the oxygen in air as the final electron acceptor, in a process called oxidative phosphorylation. Aerobic prokaryotes, which lack mitochondria, use a variety of simpler mechanisms.

  • Firstly, the NADH + H+ generated by glycolysis has to be transferred to the mitochondrion to be oxidized, and thus to regenerate the NAD+ necessary for glycolysis to continue. However the inner mitochondrial membrane is impermeable to NADH and NAD+.[44] Use is therefore made of two “shuttles” to transport the electrons from NADH across the mitochondrial membrane. They are the malate-aspartate shuttle and the glycerol phosphate shuttle. In the former the electrons from NADH are transferred to cytosolic oxaloacetate to form malate. The malate then traverses the inner mitochondrial membrane into the mitochondrial matrix, where it is reoxidized by NAD+ forming intra-mitochondrial oxaloacetate and NADH. The oxaloacetate is then re-cycled to the cytosol via its conversion to aspartate which is readily transported out of the mitochondrion. In the glycerol phosphate shuttle electrons from cytosolic NADH are transferred to dihydroxyacetone to form glycerol-3-phosphate which readily traverses the outer mitochondrial membrane. Glycerol-3-phosphate is then reoxidized to dihydroxyacetone, donating its electrons to FAD instead of NAD+.[44] This reaction takes place on the inner mitochondrial membrane, allowing FADH2 to donate its electrons directly to coenzyme Q (ubiquinone) which is part of the electron transport chain which ultimately transfers electrons to molecular oxygen O2, with the formation of water, and the release of energy eventually captured in the form of ATP.
  • The glycolytic end-product, pyruvate (plus NAD+) is converted to acetyl-CoA, CO2 and NADH + H+ within the mitochondria in a process called pyruvate decarboxylation.
  • The resulting acetyl-CoA enters the citric acid cycle (or Krebs Cycle), where the acetyl group of the acetyl-CoA is converted into carbon dioxide by two decarboxylation reactions with the formation of yet more intra-mitochondrial NADH + H+.
  • The intra-mitochondrial NADH + H+ is oxidized to NAD+ by the electron transport chain, using oxygen as the final electron acceptor to form water. The energy released during this process is used to create a hydrogen ion (or proton) gradient across the inner membrane of the mitochondrion.
  • Finally, the proton gradient is used to produce about 2.5 ATP for every NADH + H+ oxidized in a process called oxidative phosphorylation.[44]

Conversion of carbohydrates into fatty acids and cholesterol edit

The pyruvate produced by glycolysis is an important intermediary in the conversion of carbohydrates into fatty acids and cholesterol.[45] This occurs via the conversion of pyruvate into acetyl-CoA in the mitochondrion. However, this acetyl CoA needs to be transported into cytosol where the synthesis of fatty acids and cholesterol occurs. This cannot occur directly. To obtain cytosolic acetyl-CoA, citrate (produced by the condensation of acetyl CoA with oxaloacetate) is removed from the citric acid cycle and carried across the inner mitochondrial membrane into the cytosol.[45] There it is cleaved by ATP citrate lyase into acetyl-CoA and oxaloacetate. The oxaloacetate is returned to mitochondrion as malate (and then back into oxaloacetate to transfer more acetyl-CoA out of the mitochondrion). The cytosolic acetyl-CoA can be carboxylated by acetyl-CoA carboxylase into malonyl CoA, the first committed step in the synthesis of fatty acids, or it can be combined with acetoacetyl-CoA to form 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) which is the rate limiting step controlling the synthesis of cholesterol.[46] Cholesterol can be used as is, as a structural component of cellular membranes, or it can be used to synthesize the steroid hormones, bile salts, and vitamin D.[34][45][46]

Conversion of pyruvate into oxaloacetate for the citric acid cycle edit

Pyruvate molecules produced by glycolysis are actively transported across the inner mitochondrial membrane, and into the matrix where they can either be oxidized and combined with coenzyme A to form CO2, acetyl-CoA, and NADH,[34] or they can be carboxylated (by pyruvate carboxylase) to form oxaloacetate. This latter reaction "fills up" the amount of oxaloacetate in the citric acid cycle, and is therefore an anaplerotic reaction (from the Greek meaning to "fill up"), increasing the cycle's capacity to metabolize acetyl-CoA when the tissue's energy needs (e.g. in heart and skeletal muscle) are suddenly increased by activity.[47] In the citric acid cycle all the intermediates (e.g. citrate, iso-citrate, alpha-ketoglutarate, succinate, fumarate, malate and oxaloacetate) are regenerated during each turn of the cycle. Adding more of any of these intermediates to the mitochondrion therefore means that that additional amount is retained within the cycle, increasing all the other intermediates as one is converted into the other. Hence the addition of oxaloacetate greatly increases the amounts of all the citric acid intermediates, thereby increasing the cycle's capacity to metabolize acetyl CoA, converting its acetate component into CO2 and water, with the release of enough energy to form 11 ATP and 1 GTP molecule for each additional molecule of acetyl CoA that combines with oxaloacetate in the cycle.[47]

To cataplerotically remove oxaloacetate from the citric cycle, malate can be transported from the mitochondrion into the cytoplasm, decreasing the amount of oxaloacetate that can be regenerated.[47] Furthermore, citric acid intermediates are constantly used to form a variety of substances such as the purines, pyrimidines and porphyrins.[47]

Intermediates for other pathways edit

This article concentrates on the catabolic role of glycolysis with regard to converting potential chemical energy to usable chemical energy during the oxidation of glucose to pyruvate. Many of the metabolites in the glycolytic pathway are also used by anabolic pathways, and, as a consequence, flux through the pathway is critical to maintain a supply of carbon skeletons for biosynthesis.[citation needed]

The following metabolic pathways are all strongly reliant on glycolysis as a source of metabolites: and many more.

Although gluconeogenesis and glycolysis share many intermediates the one is not functionally a branch or tributary of the other. There are two regulatory steps in both pathways which, when active in the one pathway, are automatically inactive in the other. The two processes can therefore not be simultaneously active.[48] Indeed, if both sets of reactions were highly active at the same time the net result would be the hydrolysis of four high energy phosphate bonds (two ATP and two GTP) per reaction cycle.[48]

NAD+ is the oxidizing agent in glycolysis, as it is in most other energy yielding metabolic reactions (e.g. beta-oxidation of fatty acids, and during the citric acid cycle). The NADH thus produced is primarily used to ultimately transfer electrons to O2 to produce water, or, when O2 is not available, to produced compounds such as lactate or ethanol (see Anoxic regeneration of NAD+ above). NADH is rarely used for synthetic processes, the notable exception being gluconeogenesis. During fatty acid and cholesterol synthesis the reducing agent is NADPH. This difference exemplifies a general principle that NADPH is consumed during biosynthetic reactions, whereas NADH is generated in energy-yielding reactions.[48] The source of the NADPH is two-fold. When malate is oxidatively decarboxylated by “NADP+-linked malic enzyme" pyruvate, CO2 and NADPH are formed. NADPH is also formed by the pentose phosphate pathway which converts glucose into ribose, which can be used in synthesis of nucleotides and nucleic acids, or it can be catabolized to pyruvate.[48]

Glycolysis in disease edit

Diabetes edit

Cellular uptake of glucose occurs in response to insulin signals, and glucose is subsequently broken down through glycolysis, lowering blood sugar levels. However, the low insulin levels seen in diabetes result in hyperglycemia, where glucose levels in the blood rise and glucose is not properly taken up by cells. Hepatocytes further contribute to this hyperglycemia through gluconeogenesis. Glycolysis in hepatocytes controls hepatic glucose production, and when glucose is overproduced by the liver without having a means of being broken down by the body, hyperglycemia results.[49]

Genetic diseases edit

Glycolytic mutations are generally rare due to importance of the metabolic pathway; the majority of occurring mutations result in an inability of the cell to respire, and therefore cause the death of the cell at an early stage. However, some mutations (glycogen storage diseases and other inborn errors of carbohydrate metabolism) are seen with one notable example being pyruvate kinase deficiency, leading to chronic hemolytic anemia.[citation needed]

Cancer edit

Malignant tumor cells perform glycolysis at a rate that is ten times faster than their noncancerous tissue counterparts.[50] During their genesis, limited capillary support often results in hypoxia (decreased O2 supply) within the tumor cells. Thus, these cells rely on anaerobic metabolic processes such as glycolysis for ATP (adenosine triphosphate). Some tumor cells overexpress specific glycolytic enzymes which result in higher rates of glycolysis.[51] Often these enzymes are Isoenzymes, of traditional glycolysis enzymes, that vary in their susceptibility to traditional feedback inhibition. The increase in glycolytic activity ultimately counteracts the effects of hypoxia by generating sufficient ATP from this anaerobic pathway.[52] This phenomenon was first described in 1930 by Otto Warburg and is referred to as the Warburg effect. The Warburg hypothesis claims that cancer is primarily caused by dysfunctionality in mitochondrial metabolism, rather than because of the uncontrolled growth of cells. A number of theories have been advanced to explain the Warburg effect. One such theory suggests that the increased glycolysis is a normal protective process of the body and that malignant change could be primarily caused by energy metabolism.[53]

This high glycolysis rate has important medical applications, as high aerobic glycolysis by malignant tumors is utilized clinically to diagnose and monitor treatment responses of cancers by imaging uptake of 2-18F-2-deoxyglucose (FDG) (a radioactive modified hexokinase substrate) with positron emission tomography (PET).[54][55]

There is ongoing research to affect mitochondrial metabolism and treat cancer by reducing glycolysis and thus starving cancerous cells in various new ways, including a ketogenic diet.[56][57][58]

Interactive pathway map edit

The diagram below shows human protein names. Names in other organisms may be different and the number of isozymes (such as HK1, HK2, ...) is likely to be different too.

Click on genes, proteins and metabolites below to link to respective articles.[§ 1]

[[File:
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
[[
]]
|alt=Glycolysis and Gluconeogenesis edit]]
Glycolysis and Gluconeogenesis edit
  1. ^ The interactive pathway map can be edited at WikiPathways: "GlycolysisGluconeogenesis_WP534".

Alternative nomenclature edit

Some of the metabolites in glycolysis have alternative names and nomenclature. In part, this is because some of them are common to other pathways, such as the Calvin cycle.

This article Alternative
1 Glucose Glc Dextrose
2 Glucose-6-phosphate G6P
3 Fructose-6-phosphate F6P
4 Fructose-1,6-bisphosphate F1,6BP Fructose 1,6-diphosphate FBP; FDP; F1,6DP
5 Dihydroxyacetone phosphate DHAP Glycerone phosphate
6 Glyceraldehyde-3-phosphate GADP 3-Phosphoglyceraldehyde PGAL; G3P; GALP; GAP; TP
7 1,3-Bisphosphoglycerate 1,3BPG Glycerate-1,3-bisphosphate,
glycerate-1,3-diphosphate,
1,3-diphosphoglycerate
PGAP; BPG; DPG
8 3-Phosphoglycerate 3PG Glycerate-3-phosphate PGA; GP
9 2-Phosphoglycerate 2PG Glycerate-2-phosphate
10 Phosphoenolpyruvate PEP
11 Pyruvate Pyr Pyruvic acid conjugate base

Structure of glycolysis components in Fischer projections and polygonal model edit

The intermediates of glycolysis depicted in Fischer projections show the chemical changing step by step. Such image can be compared to polygonal model representation.[59] Another comparation of Fischer projections and Poligonal Model in glycolysis is shown in a video.[60] Video animations in the same channel in YouTube can be seen for another metabolic pathway (Krebs Cycle) and the representation and applying of Polygonal Model in Organic Chemistry [61]

 
Glycolysis - Structure of anaerobic glycolysis components showed using Fischer projections, left, and polygonal model, right. The compounds correspond to glucose (GLU), glucose 6-phosphate (G6P), fructose 6-phosphate (F6P), fructose 1,6-bisphosphate ( F16BP), dihydroxyacetone phosphate (DHAP), glyceraldehyde 3-phosphate(GA3P), 1,3-bisphosphoglycerate (13BPG), 3-phosphoglycerate (3PG), 2-phosphoglycerate (2PG), phosphoenolpyruvate (PEP), pyruvate (PIR), and lactate (LAC). The enzymes which participate of this pathway are indicated by underlined numbers, and correspond to hexokinase (1), glucose-6-phosphate isomerase (2), phosphofructokinase-1 (3), fructose-bisphosphate aldolase (4), triosephosphate isomerase (5), glyceraldehyde-3-phosphate dehydrogenase (5), phosphoglycerate kinase (7), phosphoglycerate mutase (8), phosphopyruvate hydratase (enolase) (9), pyruvate kinase (10), and lactate dehydrogenase (11). The participant coenzymes (NAD+, NADH + H+, ATP and ADP), inorganic phosphate, H2O and CO2 were omitted in these representations. The phosphorylation reactions from ATP, as well the ADP phosphorylation reactions in later steps of glycolysis are shown as ~P respectively entering or going out the pathway. The oxireduction reactions using NAD+ or NADH are observed as hydrogens “2H” going out or entering the pathway.

See also edit

References edit

  1. ^ Alfarouk KO, Verduzco D, Rauch C, Muddathir AK, Adil HH, Elhassan GO, et al. (18 December 2014). "Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question". Oncoscience. 1 (12): 777–802. doi:10.18632/oncoscience.109. PMC 4303887. PMID 25621294.
  2. ^ Romano AH, Conway T (1996). "Evolution of carbohydrate metabolic pathways". Research in Microbiology. 147 (6–7): 448–455. doi:10.1016/0923-2508(96)83998-2. PMID 9084754.
  3. ^ Keller MA, Turchyn AV, Ralser M (April 2014). "Non-enzymatic glycolysis and pentose phosphate pathway-like reactions in a plausible Archean ocean". Molecular Systems Biology. 10 (4): 725. doi:10.1002/msb.20145228. PMC 4023395. PMID 24771084.
  4. ^ Kim BH, Gadd GM. (2011) Bacterial Physiology and Metabolism, 3rd edition.
  5. ^ a b c Mehta S (20 September 2011). "Glycolysis – Animation and Notes". PharmaXchange.
  6. ^ a b Lane AN, Fan TW, Higashi RM (2009). "Metabolic acidosis and the importance of balanced equations". Metabolomics. 5 (2): 163–165. doi:10.1007/s11306-008-0142-2. S2CID 35500999.
  7. ^ Barnett JA (April 2003). "A history of research on yeasts 5: the fermentation pathway". Yeast. 20 (6): 509–543. doi:10.1002/yea.986. PMID 12722184. S2CID 26805351.
  8. ^ . www.pasteurbrewing.com. Archived from the original on 2011-01-13. Retrieved 2016-02-23.
  9. ^ Alba-Lois L, Segal-Kischinevzky C (January 2010). "Yeast fermentation and the making of beer and wine". Nature Education. 3 (9): 17.
  10. ^ Kohler R (1971-03-01). "The background to Eduard Buchner's discovery of cell-free fermentation". Journal of the History of Biology. 4 (1): 35–61. doi:10.1007/BF00356976. PMID 11609437. S2CID 46573308.
  11. ^ "Eduard Buchner - Biographical". www.nobelprize.org. Retrieved 2016-02-23.
  12. ^ a b Cornish-Bowden A (1997). "Harden and Young's Discovery of Fructose 1,6-Bisphosphate". New Beer in an Old Bottle: Eduard Buchner and the Growth of Biochemical Knowledge. Valencia, Spain.{{cite book}}: CS1 maint: location missing publisher (link)
  13. ^ a b Palmer G. "Chapter 3: The History of Glycolysis: An Example of a Linear Metabolic Pathway.". (PDF). Archived from the original (PDF) on 18 November 2017.
  14. ^ "Otto Meyerhof - Biographical". www.nobelprize.org. Retrieved 2016-02-23.
  15. ^ a b c Kresge N, Simoni RD, Hill RL (January 2005). "Otto Fritz Meyerhof and the elucidation of the glycolytic pathway". The Journal of Biological Chemistry. 280 (4): e3. doi:10.1016/S0021-9258(20)76366-0. PMID 15665335.
  16. ^ "Embden, Gustav – Dictionary definition of Embden, Gustav | Encyclopedia.com: FREE online dictionary". www.encyclopedia.com. Retrieved 2016-02-23.
  17. ^ Reeves RE, South DJ, Blytt HJ, Warren LG (December 1974). "Pyrophosphate:D-fructose 6-phosphate 1-phosphotransferase. A new enzyme with the glycolytic function of 6-phosphofructokinase". The Journal of Biological Chemistry. 249 (24): 7737–7741. doi:10.1016/S0021-9258(19)42029-2. PMID 4372217.
  18. ^ Selig M, Xavier KB, Santos H, Schönheit P (April 1997). "Comparative analysis of Embden-Meyerhof and Entner-Doudoroff glycolytic pathways in hyperthermophilic archaea and the bacterium Thermotoga". Archives of Microbiology. 167 (4): 217–232. doi:10.1007/BF03356097. PMID 9075622. S2CID 19489719.
  19. ^ Garrett RH, Grisham CM (2012). Biochemistry (5th ed.). Cengage Learning. ISBN 978-1-133-10629-6.
  20. ^ Berg JM, Tymoczko JL, Stryer L (2007). Biochemistry (6th ed.). New York: Freeman. p. 622. ISBN 978-0716787242.
  21. ^ a b Garrett R, Grisham CM (2005). Biochemistry (3rd ed.). Belmont, CA: Thomson Brooks/Cole. p. 584. ISBN 978-0-534-49033-1.
  22. ^ Shimizu K, Matsuoka Y (March 2019). "Regulation of glycolytic flux and overflow metabolism depending on the source of energy generation for energy demand". Biotechnology Advances. 37 (2): 284–305. doi:10.1016/j.biotechadv.2018.12.007. PMID 30576718. S2CID 58591361.
  23. ^ a b Chubukov V, Gerosa L, Kochanowski K, Sauer U (May 2014). "Coordination of microbial metabolism". Nature Reviews. Microbiology. 12 (5): 327–340. doi:10.1038/nrmicro3238. PMID 24658329. S2CID 28413431.
  24. ^ Hochachka PW (1999). "Cross-Species Studies of Glycolytic Function". In Roach RC, Wagner PD, Hackett PH (eds.). Hypoxia. Advances in Experimental Medicine and Biology. Vol. 474. Boston, MA: Springer US. pp. 219–229. doi:10.1007/978-1-4615-4711-2_18. ISBN 978-1-4613-7134-2. PMID 10635004.
  25. ^ Lemaigre FP, Rousseau GG (October 1994). "Transcriptional control of genes that regulate glycolysis and gluconeogenesis in adult liver". The Biochemical Journal. 303 (1): 1–14. doi:10.1042/bj3030001. PMC 1137548. PMID 7945228.
  26. ^ Bian X, Jiang H, Meng Y, Li YP, Fang J, Lu Z (March 2022). "Regulation of gene expression by glycolytic and gluconeogenic enzymes". Trends in Cell Biology. 32 (9): 786–799. doi:10.1016/j.tcb.2022.02.003. PMID 35300892. S2CID 247459973.
  27. ^ a b Gerosa L, Sauer U (August 2011). "Regulation and control of metabolic fluxes in microbes". Current Opinion in Biotechnology. 22 (4): 566–575. doi:10.1016/j.copbio.2011.04.016. PMID 21600757.
  28. ^ Chowdhury S, Hepper S, Lodi MK, Saier MH, Uetz P (April 2021). "The Protein Interactome of Glycolysis in Escherichia coli". Proteomes. 9 (2): 16. doi:10.3390/proteomes9020016. PMC 8167557. PMID 33917325.
  29. ^ Rodionova IA, Zhang Z, Mehla J, Goodacre N, Babu M, Emili A, et al. (August 2017). "The phosphocarrier protein HPr of the bacterial phosphotransferase system globally regulates energy metabolism by directly interacting with multiple enzymes in Escherichia coli". The Journal of Biological Chemistry. 292 (34): 14250–14257. doi:10.1074/jbc.M117.795294. PMC 5572926. PMID 28634232.
  30. ^ Pisithkul T, Patel NM, Amador-Noguez D (April 2015). "Post-translational modifications as key regulators of bacterial metabolic fluxes". Current Opinion in Microbiology. 24: 29–37. doi:10.1016/j.mib.2014.12.006. PMID 25597444.
  31. ^ a b c Koeslag JH, Saunders PT, Terblanche E (June 2003). "A reappraisal of the blood glucose homeostat which comprehensively explains the type 2 diabetes mellitus-syndrome X complex". The Journal of Physiology (published 2003). 549 (Pt 2): 333–346. doi:10.1113/jphysiol.2002.037895. PMC 2342944. PMID 12717005.
  32. ^ a b c d e Stryer L (1995). "Glycolysis.". Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 483–508. ISBN 0-7167-2009-4.
  33. ^ Stryer L (1995). Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. p. 773. ISBN 0-7167-2009-4.
  34. ^ a b c Voet D, Voet JG, Pratt CW (2006). Fundamentals of Biochemistry (2nd ed.). John Wiley and Sons, Inc. pp. 547, 556. ISBN 978-0-471-21495-3.
  35. ^ Beis I, Newsholme EA (October 1975). "The contents of adenine nucleotides, phosphagens and some glycolytic intermediates in resting muscles from vertebrates and invertebrates". The Biochemical Journal. 152 (1): 23–32. doi:10.1042/bj1520023. PMC 1172435. PMID 1212224.
  36. ^ Voet D, Voet JG (2004). Biochemistry (3rd ed.). New York: John Wiley & Sons, Inc.
  37. ^ Lackie J (2010). TIGAR. Oxford Reference Online: Oxford University Press. ISBN 9780199549351.
  38. ^ Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, et al. (July 2006). "TIGAR, a p53-inducible regulator of glycolysis and apoptosis". Cell. 126 (1): 107–120. doi:10.1016/j.cell.2006.05.036. PMID 16839880. S2CID 15006256.
  39. ^ "TIGAR TP53 induced glycolysis regulatory phosphatase [Homo sapiens (human)] - Gene - NCBI". www.ncbi.nlm.nih.gov. Retrieved 2018-05-17.
  40. ^ Carbonell J, Felíu JE, Marco R, Sols A (August 1973). "Pyruvate kinase. Classes of regulatory isoenzymes in mammalian tissues". European Journal of Biochemistry. 37 (1): 148–156. doi:10.1111/j.1432-1033.1973.tb02969.x. hdl:10261/78345. PMID 4729424.
  41. ^ Valentini G, Chiarelli L, Fortin R, Speranza ML, Galizzi A, Mattevi A (June 2000). "The allosteric regulation of pyruvate kinase". The Journal of Biological Chemistry. 275 (24): 18145–18152. doi:10.1074/jbc.m001870200. PMID 10751408.
  42. ^ Israelsen WJ, Vander Heiden MG (July 2015). "Pyruvate kinase: Function, regulation and role in cancer". Seminars in Cell & Developmental Biology. 43: 43–51. doi:10.1016/j.semcdb.2015.08.004. PMC 4662905. PMID 26277545.
  43. ^ a b c d Engström L (1978). "The regulation of liver pyruvate kinase by phosphorylation--dephosphorylation". Current Topics in Cellular Regulation. Elsevier. 13: 28–51. doi:10.1016/b978-0-12-152813-3.50006-9. ISBN 978-0-12-152813-3. PMID 208818.
  44. ^ a b c Stryer L (1995). "Oxidative phosphorylation.". Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 537–549. ISBN 0-7167-2009-4.
  45. ^ a b c Stryer L (1995). "Fatty acid metabolism.". Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 603–628. ISBN 0-7167-2009-4.
  46. ^ a b Stryer L (1995). "Biosynthesis of membrane lipids and steroids.". Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 691–707. ISBN 0-7167-2009-4.
  47. ^ a b c d Stryer L (1995). "Citric acid cycle.". Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 509–527, 569–579, 614–616, 638–641, 732–735, 739–748, 770–773. ISBN 0-7167-2009-4.
  48. ^ a b c d Stryer L (1995). Biochemistry (Fourth ed.). New York: W.H. Freeman and Company. pp. 559–565, 574–576, 614–623. ISBN 0-7167-2009-4.
  49. ^ Guo X, Li H, Xu H, Woo S, Dong H, Lu F, et al. (2012-08-01). "Glycolysis in the control of blood glucose homeostasis". Acta Pharmaceutica Sinica B. 2 (4): 358–367. doi:10.1016/j.apsb.2012.06.002. ISSN 2211-3835.
  50. ^ Alfarouk KO, Verduzco D, Rauch C, Muddathir AK, Adil HH, Elhassan GO, et al. (2014). "Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question". Oncoscience. 1 (12): 777–802. doi:10.18632/oncoscience.109. PMC 4303887. PMID 25621294.
  51. ^ Alfarouk KO, Shayoub ME, Muddathir AK, Elhassan GO, Bashir AH (July 2011). "Evolution of Tumor Metabolism might Reflect Carcinogenesis as a Reverse Evolution process (Dismantling of Multicellularity)". Cancers. 3 (3): 3002–3017. doi:10.3390/cancers3033002. PMC 3759183. PMID 24310356.
  52. ^ Nelson DL, Cox MM (2005). Lehninger principles of biochemistry (4th ed.). New York: W.H. Freeman. ISBN 978-0-7167-4339-2.
  53. ^ Gold J (October 2011). . Archived from the original on May 19, 2018. Retrieved September 8, 2012.
  54. ^ Pauwels EK, Sturm EJ, Bombardieri E, Cleton FJ, Stokkel MP (October 2000). "Positron-emission tomography with [18F]fluorodeoxyglucose. Part I. Biochemical uptake mechanism and its implication for clinical studies". Journal of Cancer Research and Clinical Oncology. 126 (10): 549–59. doi:10.1007/pl00008465. PMID 11043392. S2CID 2725555.
  55. ^ "PET Scan: PET Scan Info Reveals ..." Retrieved December 5, 2005.
  56. ^ Schwartz L, Seyfried T, Alfarouk KO, Da Veiga Moreira J, Fais S (April 2017). "Out of Warburg effect: An effective cancer treatment targeting the tumor specific metabolism and dysregulated pH". Seminars in Cancer Biology. 43: 134–138. doi:10.1016/j.semcancer.2017.01.005. PMID 28122260.
  57. ^ Schwartz L, Supuran CT, Alfarouk KO (2017). "The Warburg Effect and the Hallmarks of Cancer". Anti-Cancer Agents in Medicinal Chemistry. 17 (2): 164–170. doi:10.2174/1871520616666161031143301. PMID 27804847.
  58. ^ Maroon J, Bost J, Amos A, Zuccoli G (August 2013). "Restricted calorie ketogenic diet for the treatment of glioblastoma multiforme". Journal of Child Neurology. 28 (8): 1002–1008. doi:10.1177/0883073813488670. PMID 23670248. S2CID 1994087.
  59. ^ Bonafe CF, Bispo JA, de Jesus MB (January 2018). "The polygonal model: A simple representation of biomolecules as a tool for teaching metabolism". Biochemistry and Molecular Biology Education. 46 (1): 66–75. doi:10.1002/bmb.21093. PMID 29131491. S2CID 31317102.
  60. ^ Bonafe C (23 September 2019). "Introduction to Polygonal Model - PART 1. Glycolysis and Structure of the Participant Molecules". YouTube. Archived from the original on 2021-11-04.
  61. ^ "Metabolism Animation and Polygonal Model". YouTube. Retrieved 2019-12-11.

External links edit

  • A Detailed Glycolysis Animation provided by IUBMB (Adobe Flash Required)
  • at RCSB PDB
  • Glycolytic cycle with animations at wdv.com
  • Metabolism, Cellular Respiration and Photosynthesis - The Virtual Library of Biochemistry, Molecular Biology and Cell Biology
  • at ufp.pt
  • Expasy biochemical pathways poster at ExPASy
  • MedicalMnemonics.com: 317 5468
  • metpath: Interactive representation of glycolysis

glycolysis, metabolic, pathway, that, converts, glucose, c6h12o6, into, pyruvate, most, organisms, occurs, liquid, part, cells, cytosol, free, energy, released, this, process, used, form, high, energy, molecules, adenosine, triphosphate, reduced, nicotinamide,. Glycolysis is the metabolic pathway that converts glucose C6H12O6 into pyruvate and in most organisms occurs in the liquid part of cells the cytosol The free energy released in this process is used to form the high energy molecules adenosine triphosphate ATP and reduced nicotinamide adenine dinucleotide NADH 1 Glycolysis is a sequence of ten reactions catalyzed by enzymes Glucose G6P F6P F1 6BP GADP DHAP 1 3BPG 3PG 2PG PEP Pyruvate HK PGI PFK ALDO TPI GAPDH PGK PGM ENO PK Glycolysis The metabolic pathway of glycolysis converts glucose to pyruvate via a series of intermediate metabolites Each chemical modification is performed by a different enzyme Steps 1 and 3 consume ATP and steps 7 and 10 produce ATP Since steps 6 10 occur twice per glucose molecule this leads to a net production of ATP Summary of aerobic respiration Summary of the 10 reactions of the glycolysis pathwayThe wide occurrence of glycolysis in other species indicates that it is an ancient metabolic pathway 2 Indeed the reactions that make up glycolysis and its parallel pathway the pentose phosphate pathway can occur in the oxygen free conditions of the Archean oceans also in the absence of enzymes catalyzed by metal ions meaning this is a plausible prebiotic pathway for abiogenesis 3 The most common type of glycolysis is the Embden Meyerhof Parnas EMP pathway which was discovered by Gustav Embden Otto Meyerhof and Jakub Karol Parnas Glycolysis also refers to other pathways such as the Entner Doudoroff pathway and various heterofermentative and homofermentative pathways However the discussion here will be limited to the Embden Meyerhof Parnas pathway 4 The glycolysis pathway can be separated into two phases 5 Investment phase wherein ATP is consumed Yield phase wherein more ATP is produced than originally consumedContents 1 Overview 2 History 3 Sequence of reactions 3 1 Summary of reactions 3 2 Preparatory phase 3 3 Pay off phase 3 4 Biochemical logic 3 5 Free energy changes 4 Regulation 4 1 Regulation by insulin in animals 4 2 Regulated Enzymes in Glycolysis 4 2 1 Hexokinase and glucokinase 4 2 2 Phosphofructokinase 4 2 3 Pyruvate kinase 5 Post glycolysis processes 5 1 Anoxic regeneration of NAD 5 2 Aerobic regeneration of NAD and further catabolism of pyruvate 5 3 Conversion of carbohydrates into fatty acids and cholesterol 5 4 Conversion of pyruvate into oxaloacetate for the citric acid cycle 6 Intermediates for other pathways 7 Glycolysis in disease 7 1 Diabetes 7 2 Genetic diseases 7 3 Cancer 8 Interactive pathway map 9 Alternative nomenclature 10 Structure of glycolysis components in Fischer projections and polygonal model 11 See also 12 References 13 External linksOverview editThe overall reaction of glycolysis is d Glucose nbsp 2 NAD 2 ADP 2 P i nbsp 2 Pyruvate2 nbsp 2 NADH 2 H 2 ATP 2 H2O nbsp Glycolysis pathway overview The use of symbols in this equation makes it appear unbalanced with respect to oxygen atoms hydrogen atoms and charges Atom balance is maintained by the two phosphate Pi groups 6 Each exists in the form of a hydrogen phosphate anion HPO4 2 dissociating to contribute 2H overall Each liberates an oxygen atom when it binds to an adenosine diphosphate ADP molecule contributing 2 O overallCharges are balanced by the difference between ADP and ATP In the cellular environment all three hydroxyl groups of ADP dissociate into O and H giving ADP3 and this ion tends to exist in an ionic bond with Mg2 giving ADPMg ATP behaves identically except that it has four hydroxyl groups giving ATPMg2 When these differences along with the true charges on the two phosphate groups are considered together the net charges of 4 on each side are balanced For simple fermentations the metabolism of one molecule of glucose to two molecules of pyruvate has a net yield of two molecules of ATP Most cells will then carry out further reactions to repay the used NAD and produce a final product of ethanol or lactic acid Many bacteria use inorganic compounds as hydrogen acceptors to regenerate the NAD Cells performing aerobic respiration synthesize much more ATP but not as part of glycolysis These further aerobic reactions use pyruvate and NADH H from glycolysis Eukaryotic aerobic respiration produces approximately 34 additional molecules of ATP for each glucose molecule however most of these are produced by a mechanism vastly different from the substrate level phosphorylation in glycolysis The lower energy production per glucose of anaerobic respiration relative to aerobic respiration results in greater flux through the pathway under hypoxic low oxygen conditions unless alternative sources of anaerobically oxidizable substrates such as fatty acids are found Metabolism of common monosaccharides including glycolysis gluconeogenesis glycogenesis and glycogenolysis nbsp History editThe pathway of glycolysis as it is known today took almost 100 years to fully elucidate 7 The combined results of many smaller experiments were required in order to understand the pathway as a whole The first steps in understanding glycolysis began in the nineteenth century with the wine industry For economic reasons the French wine industry sought to investigate why wine sometimes turned distasteful instead of fermenting into alcohol French scientist Louis Pasteur researched this issue during the 1850s and the results of his experiments began the long road to elucidating the pathway of glycolysis 8 His experiments showed that fermentation occurs by the action of living microorganisms yeasts and that yeast s glucose consumption decreased under aerobic conditions of fermentation in comparison to anaerobic conditions the Pasteur effect 9 nbsp Eduard Buchner Discovered cell free fermentation Insight into the component steps of glycolysis were provided by the non cellular fermentation experiments of Eduard Buchner during the 1890s 10 11 Buchner demonstrated that the conversion of glucose to ethanol was possible using a non living extract of yeast due to the action of enzymes in the extract 12 135 148 This experiment not only revolutionized biochemistry but also allowed later scientists to analyze this pathway in a more controlled laboratory setting In a series of experiments 1905 1911 scientists Arthur Harden and William Young discovered more pieces of glycolysis 13 They discovered the regulatory effects of ATP on glucose consumption during alcohol fermentation They also shed light on the role of one compound as a glycolysis intermediate fructose 1 6 bisphosphate 12 151 158 The elucidation of fructose 1 6 bisphosphate was accomplished by measuring CO2 levels when yeast juice was incubated with glucose CO2 production increased rapidly then slowed down Harden and Young noted that this process would restart if an inorganic phosphate Pi was added to the mixture Harden and Young deduced that this process produced organic phosphate esters and further experiments allowed them to extract fructose diphosphate F 1 6 DP Arthur Harden and William Young along with Nick Sheppard determined in a second experiment that a heat sensitive high molecular weight subcellular fraction the enzymes and a heat insensitive low molecular weight cytoplasm fraction ADP ATP and NAD and other cofactors are required together for fermentation to proceed This experiment begun by observing that dialyzed purified yeast juice could not ferment or even create a sugar phosphate This mixture was rescued with the addition of undialyzed yeast extract that had been boiled Boiling the yeast extract renders all proteins inactive as it denatures them The ability of boiled extract plus dialyzed juice to complete fermentation suggests that the cofactors were non protein in character 13 nbsp Otto Meyerhof One of the main scientists involved in completing the puzzle of glycolysisIn the 1920s Otto Meyerhof was able to link together some of the many individual pieces of glycolysis discovered by Buchner Harden and Young Meyerhof and his team were able to extract different glycolytic enzymes from muscle tissue and combine them to artificially create the pathway from glycogen to lactic acid 14 15 In one paper Meyerhof and scientist Renate Junowicz Kockolaty investigated the reaction that splits fructose 1 6 diphosphate into the two triose phosphates Previous work proposed that the split occurred via 1 3 diphosphoglyceraldehyde plus an oxidizing enzyme and cozymase Meyerhoff and Junowicz found that the equilibrium constant for the isomerase and aldoses reaction were not affected by inorganic phosphates or any other cozymase or oxidizing enzymes They further removed diphosphoglyceraldehyde as a possible intermediate in glycolysis 15 With all of these pieces available by the 1930s Gustav Embden proposed a detailed step by step outline of that pathway we now know as glycolysis 16 The biggest difficulties in determining the intricacies of the pathway were due to the very short lifetime and low steady state concentrations of the intermediates of the fast glycolytic reactions By the 1940s Meyerhof Embden and many other biochemists had finally completed the puzzle of glycolysis 15 The understanding of the isolated pathway has been expanded in the subsequent decades to include further details of its regulation and integration with other metabolic pathways Sequence of reactions editSummary of reactions edit Glucose nbsp HexokinaseATP ADP nbsp Glucose 6 phosphate nbsp Glucose 6 phosphateisomerase nbsp Fructose 6 phosphate nbsp Phosphofructokinase 1ATP ADP nbsp Fructose 1 6 bisphosphate nbsp Fructose bisphosphatealdolase nbsp Dihydroxyacetone phosphate nbsp Glyceraldehyde 3 phosphate nbsp Triosephosphateisomerase nbsp 2 Glyceraldehyde 3 phosphate2 nbsp Glyceraldehyde 3 phosphatedehydrogenaseNAD Pi NADH H nbsp NAD Pi NADH H 2 1 3 Bisphosphoglycerate2 nbsp Phosphoglycerate kinaseADP ATP nbsp ADP ATP 2 3 Phosphoglycerate2 nbsp Phosphoglycerate mutase nbsp 2 2 Phosphoglycerate2 nbsp Phosphopyruvatehydratase enolase H2O nbsp H2O 2 Phosphoenolpyruvate2 nbsp Pyruvate kinaseADP ATP nbsp 2 Pyruvate2 nbsp Preparatory phase edit The first five steps of Glycolysis are regarded as the preparatory or investment phase since they consume energy to convert the glucose into two three carbon sugar phosphates 5 G3P d Glucose Glc Hexokinase glucokinase HK a transferase a d Glucose 6 phosphate G6P nbsp nbsp ATP H ADP nbsp Once glucose enters the cell the first step is phosphorylation of glucose by a family of enzymes called hexokinases to form glucose 6 phosphate G6P This reaction consumes ATP but it acts to keep the glucose concentration inside the cell low promoting continuous transport of blood glucose into the cell through the plasma membrane transporters In addition phosphorylation blocks the glucose from leaking out the cell lacks transporters for G6P and free diffusion out of the cell is prevented due to the charged nature of G6P Glucose may alternatively be formed from the phosphorolysis or hydrolysis of intracellular starch or glycogen In animals an isozyme of hexokinase called glucokinase is also used in the liver which has a much lower affinity for glucose Km in the vicinity of normal glycemia and differs in regulatory properties The different substrate affinity and alternate regulation of this enzyme are a reflection of the role of the liver in maintaining blood sugar levels Cofactors Mg2 a d Glucose 6 phosphate G6P Phosphoglucoisomerase PGI an isomerase b d Fructose 6 phosphate F6P nbsp nbsp nbsp G6P is then rearranged into fructose 6 phosphate F6P by glucose phosphate isomerase Fructose can also enter the glycolytic pathway by phosphorylation at this point The change in structure is an isomerization in which the G6P has been converted to F6P The reaction requires an enzyme phosphoglucose isomerase to proceed This reaction is freely reversible under normal cell conditions However it is often driven forward because of a low concentration of F6P which is constantly consumed during the next step of glycolysis Under conditions of high F6P concentration this reaction readily runs in reverse This phenomenon can be explained through Le Chatelier s Principle Isomerization to a keto sugar is necessary for carbanion stabilization in the fourth reaction step below b d Fructose 6 phosphate F6P Phosphofructokinase PFK 1 a transferase b d Fructose 1 6 bisphosphate F1 6BP nbsp nbsp ATP H ADP nbsp The energy expenditure of another ATP in this step is justified in 2 ways The glycolytic process up to this step becomes irreversible and the energy supplied destabilizes the molecule Because the reaction catalyzed by phosphofructokinase 1 PFK 1 is coupled to the hydrolysis of ATP an energetically favorable step it is in essence irreversible and a different pathway must be used to do the reverse conversion during gluconeogenesis This makes the reaction a key regulatory point see below Furthermore the second phosphorylation event is necessary to allow the formation of two charged groups rather than only one in the subsequent step of glycolysis ensuring the prevention of free diffusion of substrates out of the cell The same reaction can also be catalyzed by pyrophosphate dependent phosphofructokinase PFP or PPi PFK which is found in most plants some bacteria archea and protists but not in animals This enzyme uses pyrophosphate PPi as a phosphate donor instead of ATP It is a reversible reaction increasing the flexibility of glycolytic metabolism 17 A rarer ADP dependent PFK enzyme variant has been identified in archaean species 18 Cofactors Mg2 b d Fructose 1 6 bisphosphate F1 6BP Fructose bisphosphate aldolase ALDO a lyase d Glyceraldehyde 3 phosphate GADP Dihydroxyacetone phosphate DHAP nbsp nbsp nbsp nbsp Destabilizing the molecule in the previous reaction allows the hexose ring to be split by aldolase into two triose sugars dihydroxyacetone phosphate a ketose and glyceraldehyde 3 phosphate an aldose There are two classes of aldolases class I aldolases present in animals and plants and class II aldolases present in fungi and bacteria the two classes use different mechanisms in cleaving the ketose ring Electrons delocalized in the carbon carbon bond cleavage associate with the alcohol group The resulting carbanion is stabilized by the structure of the carbanion itself via resonance charge distribution and by the presence of a charged ion prosthetic group Dihydroxyacetone phosphate DHAP Triosephosphate isomerase TPI an isomerase d Glyceraldehyde 3 phosphate GADP nbsp nbsp nbsp Triosephosphate isomerase rapidly interconverts dihydroxyacetone phosphate with glyceraldehyde 3 phosphate GADP that proceeds further into glycolysis This is advantageous as it directs dihydroxyacetone phosphate down the same pathway as glyceraldehyde 3 phosphate simplifying regulation Pay off phase edit The second half of glycolysis is known as the pay off phase characterised by a net gain of the energy rich molecules ATP and NADH 5 Since glucose leads to two triose sugars in the preparatory phase each reaction in the pay off phase occurs twice per glucose molecule This yields 2 NADH molecules and 4 ATP molecules leading to a net gain of 2 NADH molecules and 2 ATP molecules from the glycolytic pathway per glucose Glyceraldehyde 3 phosphate GADP Glyceraldehyde phosphate dehydrogenase GAPDH an oxidoreductase d 1 3 Bisphosphoglycerate 1 3BPG nbsp nbsp NAD Pi NADH H nbsp The aldehyde groups of the triose sugars are oxidised and inorganic phosphate is added to them forming 1 3 bisphosphoglycerate The hydrogen is used to reduce two molecules of NAD a hydrogen carrier to give NADH H for each triose Hydrogen atom balance and charge balance are both maintained because the phosphate Pi group actually exists in the form of a hydrogen phosphate anion HPO2 4 6 which dissociates to contribute the extra H ion and gives a net charge of 3 on both sides Here arsenate AsO4 3 an anion akin to inorganic phosphate may replace phosphate as a substrate to form 1 arseno 3 phosphoglycerate This however is unstable and readily hydrolyzes to form 3 phosphoglycerate the intermediate in the next step of the pathway As a consequence of bypassing this step the molecule of ATP generated from 1 3 bisphosphoglycerate in the next reaction will not be made even though the reaction proceeds As a result arsenate is an uncoupler of glycolysis 19 1 3 Bisphosphoglycerate 1 3BPG Phosphoglycerate kinase PGK a transferase 3 Phosphoglycerate 3PG nbsp nbsp ADP ATP nbsp Phosphoglycerate kinase PGK This step is the enzymatic transfer of a phosphate group from 1 3 bisphosphoglycerate to ADP by phosphoglycerate kinase forming ATP and 3 phosphoglycerate At this step glycolysis has reached the break even point 2 molecules of ATP were consumed and 2 new molecules have now been synthesized This step one of the two substrate level phosphorylation steps requires ADP thus when the cell has plenty of ATP and little ADP this reaction does not occur Because ATP decays relatively quickly when it is not metabolized this is an important regulatory point in the glycolytic pathway ADP actually exists as ADPMg and ATP as ATPMg2 balancing the charges at 5 both sides Cofactors Mg2 3 Phosphoglycerate 3PG Phosphoglycerate mutase PGM a mutase 2 Phosphoglycerate 2PG nbsp nbsp nbsp Phosphoglycerate mutase isomerises 3 phosphoglycerate into 2 phosphoglycerate 2 Phosphoglycerate 2PG Enolase ENO a lyase Phosphoenolpyruvate PEP nbsp nbsp H2O nbsp Enolase ENO Enolase next converts 2 phosphoglycerate to phosphoenolpyruvate This reaction is an elimination reaction involving an E1cB mechanism Cofactors 2 Mg2 one conformational ion to coordinate with the carboxylate group of the substrate and one catalytic ion that participates in the dehydration Phosphoenolpyruvate PEP Pyruvate kinase PK a transferase Pyruvate Pyr nbsp nbsp ADP H ATP nbsp A final substrate level phosphorylation now forms a molecule of pyruvate and a molecule of ATP by means of the enzyme pyruvate kinase This serves as an additional regulatory step similar to the phosphoglycerate kinase step Cofactors Mg2 Biochemical logic edit The existence of more than one point of regulation indicates that intermediates between those points enter and leave the glycolysis pathway by other processes For example in the first regulated step hexokinase converts glucose into glucose 6 phosphate Instead of continuing through the glycolysis pathway this intermediate can be converted into glucose storage molecules such as glycogen or starch The reverse reaction breaking down e g glycogen produces mainly glucose 6 phosphate very little free glucose is formed in the reaction The glucose 6 phosphate so produced can enter glycolysis after the first control point In the second regulated step the third step of glycolysis phosphofructokinase converts fructose 6 phosphate into fructose 1 6 bisphosphate which then is converted into glyceraldehyde 3 phosphate and dihydroxyacetone phosphate The dihydroxyacetone phosphate can be removed from glycolysis by conversion into glycerol 3 phosphate which can be used to form triglycerides 20 Conversely triglycerides can be broken down into fatty acids and glycerol the latter in turn can be converted into dihydroxyacetone phosphate which can enter glycolysis after the second control point Free energy changes edit Concentrations of metabolites in erythrocytes 21 584 Compound Concentration mMGlucose 5 0Glucose 6 phosphate 0 083Fructose 6 phosphate 0 014Fructose 1 6 bisphosphate 0 031Dihydroxyacetone phosphate 0 14Glyceraldehyde 3 phosphate 0 0191 3 Bisphosphoglycerate 0 0012 3 Bisphosphoglycerate 4 03 Phosphoglycerate 0 122 Phosphoglycerate 0 03Phosphoenolpyruvate 0 023Pyruvate 0 051ATP 1 85ADP 0 14Pi 1 0The change in free energy DG for each step in the glycolysis pathway can be calculated using DG DG RTln Q where Q is the reaction quotient This requires knowing the concentrations of the metabolites All of these values are available for erythrocytes with the exception of the concentrations of NAD and NADH The ratio of NAD to NADH in the cytoplasm is approximately 1000 which makes the oxidation of glyceraldehyde 3 phosphate step 6 more favourable Using the measured concentrations of each step and the standard free energy changes the actual free energy change can be calculated Neglecting this is very common the delta G of ATP hydrolysis in cells is not the standard free energy change of ATP hydrolysis quoted in textbooks Change in free energy for each step of glycolysis 21 582 583 Step Reaction DG kJ mol DG kJ mol 1 Glucose ATP4 Glucose 6 phosphate2 ADP3 H 16 7 342 Glucose 6 phosphate2 Fructose 6 phosphate2 1 67 2 93 Fructose 6 phosphate2 ATP4 Fructose 1 6 bisphosphate4 ADP3 H 14 2 194 Fructose 1 6 bisphosphate4 Dihydroxyacetone phosphate2 Glyceraldehyde 3 phosphate2 23 9 0 235 Dihydroxyacetone phosphate2 Glyceraldehyde 3 phosphate2 7 56 2 46 Glyceraldehyde 3 phosphate2 Pi2 NAD 1 3 Bisphosphoglycerate4 NADH H 6 30 1 297 1 3 Bisphosphoglycerate4 ADP3 3 Phosphoglycerate3 ATP4 18 9 0 098 3 Phosphoglycerate3 2 Phosphoglycerate3 4 4 0 839 2 Phosphoglycerate3 Phosphoenolpyruvate3 H2O 1 8 1 110 Phosphoenolpyruvate3 ADP3 H Pyruvate ATP4 31 7 23 0From measuring the physiological concentrations of metabolites in an erythrocyte it seems that about seven of the steps in glycolysis are in equilibrium for that cell type Three of the steps the ones with large negative free energy changes are not in equilibrium and are referred to as irreversible such steps are often subject to regulation Step 5 in the figure is shown behind the other steps because that step is a side reaction that can decrease or increase the concentration of the intermediate glyceraldehyde 3 phosphate That compound is converted to dihydroxyacetone phosphate by the enzyme triose phosphate isomerase which is a catalytically perfect enzyme its rate is so fast that the reaction can be assumed to be in equilibrium The fact that DG is not zero indicates that the actual concentrations in the erythrocyte are not accurately known Regulation editThe enzymes that catalyse glycolysis are regulated via a range of biological mechanisms in order to control overall flux though the pathway This is vital for both homeostatsis in a static environment and metabolic adaptation to a changing environment or need 22 The details of regulation for some enzymes are highly conserved between species whereas others vary widely 23 24 Gene Expression Firstly the cellular concentrations of glycolytic enzymes are modulated via regulation of gene expression via transcription factors 25 with several glycolysis enzymes themselves acting as regulatory protein kinases in the nucleus 26 Allosteric inhibition and activation by metabolites In particular end product inhibition of regulated enzymes by metabolites such as ATP serves as negative feedback regulation of the pathway 23 27 Allosteric inhibition and activation by Protein protein interactions PPI 28 Indeed some proteins interact with and regulate multiple glycolytic enzymes 29 Post translational modification PTM 30 In particular phosphorylation and dephosphorylation is a key mechanism of regulation of pyruvate kinase in the liver Localization 27 Regulation by insulin in animals edit In animals regulation of blood glucose levels by the pancreas in conjunction with the liver is a vital part of homeostasis The beta cells in the pancreatic islets are sensitive to the blood glucose concentration 31 A rise in the blood glucose concentration causes them to release insulin into the blood which has an effect particularly on the liver but also on fat and muscle cells causing these tissues to remove glucose from the blood When the blood sugar falls the pancreatic beta cells cease insulin production but instead stimulate the neighboring pancreatic alpha cells to release glucagon into the blood 31 This in turn causes the liver to release glucose into the blood by breaking down stored glycogen and by means of gluconeogenesis If the fall in the blood glucose level is particularly rapid or severe other glucose sensors cause the release of epinephrine from the adrenal glands into the blood This has the same action as glucagon on glucose metabolism but its effect is more pronounced 31 In the liver glucagon and epinephrine cause the phosphorylation of the key regulated enzymes of glycolysis fatty acid synthesis cholesterol synthesis gluconeogenesis and glycogenolysis Insulin has the opposite effect on these enzymes 32 The phosphorylation and dephosphorylation of these enzymes ultimately in response to the glucose level in the blood is the dominant manner by which these pathways are controlled in the liver fat and muscle cells Thus the phosphorylation of phosphofructokinase inhibits glycolysis whereas its dephosphorylation through the action of insulin stimulates glycolysis 32 Regulated Enzymes in Glycolysis edit The three regulatory enzymes are hexokinase or glucokinase in the liver phosphofructokinase and pyruvate kinase The flux through the glycolytic pathway is adjusted in response to conditions both inside and outside the cell The internal factors that regulate glycolysis do so primarily to provide ATP in adequate quantities for the cell s needs The external factors act primarily on the liver fat tissue and muscles which can remove large quantities of glucose from the blood after meals thus preventing hyperglycemia by storing the excess glucose as fat or glycogen depending on the tissue type The liver is also capable of releasing glucose into the blood between meals during fasting and exercise thus preventing hypoglycemia by means of glycogenolysis and gluconeogenesis These latter reactions coincide with the halting of glycolysis in the liver In addition hexokinase and glucokinase act independently of the hormonal effects as controls at the entry points of glucose into the cells of different tissues Hexokinase responds to the glucose 6 phosphate G6P level in the cell or in the case of glucokinase to the blood sugar level in the blood to impart entirely intracellular controls of the glycolytic pathway in different tissues see below 32 When glucose has been converted into G6P by hexokinase or glucokinase it can either be converted to glucose 1 phosphate G1P for conversion to glycogen or it is alternatively converted by glycolysis to pyruvate which enters the mitochondrion where it is converted into acetyl CoA and then into citrate Excess citrate is exported from the mitochondrion back into the cytosol where ATP citrate lyase regenerates acetyl CoA and oxaloacetate OAA The acetyl CoA is then used for fatty acid synthesis and cholesterol synthesis two important ways of utilizing excess glucose when its concentration is high in blood The regulated enzymes catalyzing these reactions perform these functions when they have been dephosphorylated through the action of insulin on the liver cells Between meals during fasting exercise or hypoglycemia glucagon and epinephrine are released into the blood This causes liver glycogen to be converted back to G6P and then converted to glucose by the liver specific enzyme glucose 6 phosphatase and released into the blood Glucagon and epinephrine also stimulate gluconeogenesis which coverts non carbohydrate substrates into G6P which joins the G6P derived from glycogen or substitutes for it when the liver glycogen store have been depleted This is critical for brain function since the brain utilizes glucose as an energy source under most conditions 33 The simultaneously phosphorylation of particularly phosphofructokinase but also to a certain extent pyruvate kinase prevents glycolysis occurring at the same time as gluconeogenesis and glycogenolysis Hexokinase and glucokinase edit nbsp Yeast hexokinase B PDB 1IG8 All cells contain the enzyme hexokinase which catalyzes the conversion of glucose that has entered the cell into glucose 6 phosphate G6P Since the cell membrane is impervious to G6P hexokinase essentially acts to transport glucose into the cells from which it can then no longer escape Hexokinase is inhibited by high levels of G6P in the cell Thus the rate of entry of glucose into cells partially depends on how fast G6P can be disposed of by glycolysis and by glycogen synthesis in the cells which store glycogen namely liver and muscles 32 34 Glucokinase unlike hexokinase is not inhibited by G6P It occurs in liver cells and will only phosphorylate the glucose entering the cell to form glucose 6 phosphate G6P when the glucose in the blood is abundant This being the first step in the glycolytic pathway in the liver it therefore imparts an additional layer of control of the glycolytic pathway in this organ 32 Phosphofructokinase edit nbsp Bacillus stearothermophilus phosphofructokinase PDB 6PFK Phosphofructokinase is an important control point in the glycolytic pathway since it is one of the irreversible steps and has key allosteric effectors AMP and fructose 2 6 bisphosphate F2 6BP Fructose 2 6 bisphosphate F2 6BP is a very potent activator of phosphofructokinase PFK 1 that is synthesized when F6P is phosphorylated by a second phosphofructokinase PFK2 In the liver when blood sugar is low and glucagon elevates cAMP PFK2 is phosphorylated by protein kinase A The phosphorylation inactivates PFK2 and another domain on this protein becomes active as fructose bisphosphatase 2 which converts F2 6BP back to F6P Both glucagon and epinephrine cause high levels of cAMP in the liver The result of lower levels of liver fructose 2 6 bisphosphate is a decrease in activity of phosphofructokinase and an increase in activity of fructose 1 6 bisphosphatase so that gluconeogenesis in essence glycolysis in reverse is favored This is consistent with the role of the liver in such situations since the response of the liver to these hormones is to release glucose to the blood ATP competes with AMP for the allosteric effector site on the PFK enzyme ATP concentrations in cells are much higher than those of AMP typically 100 fold higher 35 but the concentration of ATP does not change more than about 10 under physiological conditions whereas a 10 drop in ATP results in a 6 fold increase in AMP 36 Thus the relevance of ATP as an allosteric effector is questionable An increase in AMP is a consequence of a decrease in energy charge in the cell Citrate inhibits phosphofructokinase when tested in vitro by enhancing the inhibitory effect of ATP However it is doubtful that this is a meaningful effect in vivo because citrate in the cytosol is utilized mainly for conversion to acetyl CoA for fatty acid and cholesterol synthesis TIGAR a p53 induced enzyme is responsible for the regulation of phosphofructokinase and acts to protect against oxidative stress 37 TIGAR is a single enzyme with dual function that regulates F2 6BP It can behave as a phosphatase fructuose 2 6 bisphosphatase which cleaves the phosphate at carbon 2 producing F6P It can also behave as a kinase PFK2 adding a phosphate onto carbon 2 of F6P which produces F2 6BP In humans the TIGAR protein is encoded by C12orf5 gene The TIGAR enzyme will hinder the forward progression of glycolysis by creating a build up of fructose 6 phosphate F6P which is isomerized into glucose 6 phosphate G6P The accumulation of G6P will shunt carbons into the pentose phosphate pathway 38 39 Pyruvate kinase edit nbsp Yeast pyruvate kinase PDB 1A3W Main article Pyruvate kinase The final step of glycolysis is catalysed by pyruvate kinase to form pyruvate and another ATP It is regulated by a range of different transcriptional covalent and non covalent regulation mechanisms which can vary widely in different tissues 40 41 42 For example in the liver pyruvate kinase is regulated based on glucose availability During fasting no glucose available glucagon activates protein kinase A which phosphorylates pyruvate kinase to inhibit it 43 An increase in blood sugar leads to secretion of insulin which activates protein phosphatase 1 leading to dephosphorylation and re activation of pyruvate kinase 43 These controls prevent pyruvate kinase from being active at the same time as the enzymes that catalyze the reverse reaction pyruvate carboxylase and phosphoenolpyruvate carboxykinase preventing a futile cycle 43 Conversely the isoform of pyruvate kinasein found in muscle is not affected by protein kinase A which is activated by adrenaline in that tissue so that glycolysis remains active in muscles even during fasting 43 Post glycolysis processes editThe overall process of glycolysis is Glucose 2 NAD 2 ADP 2 Pi 2 pyruvate 2 NADH 2 H 2 ATPIf glycolysis were to continue indefinitely all of the NAD would be used up and glycolysis would stop To allow glycolysis to continue organisms must be able to oxidize NADH back to NAD How this is performed depends on which external electron acceptor is available Anoxic regeneration of NAD edit This section does not cite any sources Please help improve this section by adding citations to reliable sources Unsourced material may be challenged and removed June 2022 Learn how and when to remove this template message One method of doing this is to simply have the pyruvate do the oxidation in this process pyruvate is converted to lactate the conjugate base of lactic acid in a process called lactic acid fermentation Pyruvate NADH H lactate NAD This process occurs in the bacteria involved in making yogurt the lactic acid causes the milk to curdle This process also occurs in animals under hypoxic or partially anaerobic conditions found for example in overworked muscles that are starved of oxygen In many tissues this is a cellular last resort for energy most animal tissue cannot tolerate anaerobic conditions for an extended period of time Some organisms such as yeast convert NADH back to NAD in a process called ethanol fermentation In this process the pyruvate is converted first to acetaldehyde and carbon dioxide and then to ethanol Lactic acid fermentation and ethanol fermentation can occur in the absence of oxygen This anaerobic fermentation allows many single cell organisms to use glycolysis as their only energy source Anoxic regeneration of NAD is only an effective means of energy production during short intense exercise in vertebrates for a period ranging from 10 seconds to 2 minutes during a maximal effort in humans At lower exercise intensities it can sustain muscle activity in diving animals such as seals whales and other aquatic vertebrates for very much longer periods of time Under these conditions NAD is replenished by NADH donating its electrons to pyruvate to form lactate This produces 2 ATP molecules per glucose molecule or about 5 of glucose s energy potential 38 ATP molecules in bacteria But the speed at which ATP is produced in this manner is about 100 times that of oxidative phosphorylation The pH in the cytoplasm quickly drops when hydrogen ions accumulate in the muscle eventually inhibiting the enzymes involved in glycolysis The burning sensation in muscles during hard exercise can be attributed to the release of hydrogen ions during the shift to glucose fermentation from glucose oxidation to carbon dioxide and water when aerobic metabolism can no longer keep pace with the energy demands of the muscles These hydrogen ions form a part of lactic acid The body falls back on this less efficient but faster method of producing ATP under low oxygen conditions This is thought to have been the primary means of energy production in earlier organisms before oxygen reached high concentrations in the atmosphere between 2000 and 2500 million years ago and thus would represent a more ancient form of energy production than the aerobic replenishment of NAD in cells The liver in mammals gets rid of this excess lactate by transforming it back into pyruvate under aerobic conditions see Cori cycle Fermentation of pyruvate to lactate is sometimes also called anaerobic glycolysis however glycolysis ends with the production of pyruvate regardless of the presence or absence of oxygen In the above two examples of fermentation NADH is oxidized by transferring two electrons to pyruvate However anaerobic bacteria use a wide variety of compounds as the terminal electron acceptors in cellular respiration nitrogenous compounds such as nitrates and nitrites sulfur compounds such as sulfates sulfites sulfur dioxide and elemental sulfur carbon dioxide iron compounds manganese compounds cobalt compounds and uranium compounds Aerobic regeneration of NAD and further catabolism of pyruvate edit In aerobic eukaryotes a complex mechanism has developed to use the oxygen in air as the final electron acceptor in a process called oxidative phosphorylation Aerobic prokaryotes which lack mitochondria use a variety of simpler mechanisms Firstly the NADH H generated by glycolysis has to be transferred to the mitochondrion to be oxidized and thus to regenerate the NAD necessary for glycolysis to continue However the inner mitochondrial membrane is impermeable to NADH and NAD 44 Use is therefore made of two shuttles to transport the electrons from NADH across the mitochondrial membrane They are the malate aspartate shuttle and the glycerol phosphate shuttle In the former the electrons from NADH are transferred to cytosolic oxaloacetate to form malate The malate then traverses the inner mitochondrial membrane into the mitochondrial matrix where it is reoxidized by NAD forming intra mitochondrial oxaloacetate and NADH The oxaloacetate is then re cycled to the cytosol via its conversion to aspartate which is readily transported out of the mitochondrion In the glycerol phosphate shuttle electrons from cytosolic NADH are transferred to dihydroxyacetone to form glycerol 3 phosphate which readily traverses the outer mitochondrial membrane Glycerol 3 phosphate is then reoxidized to dihydroxyacetone donating its electrons to FAD instead of NAD 44 This reaction takes place on the inner mitochondrial membrane allowing FADH2 to donate its electrons directly to coenzyme Q ubiquinone which is part of the electron transport chain which ultimately transfers electrons to molecular oxygen O2 with the formation of water and the release of energy eventually captured in the form of ATP The glycolytic end product pyruvate plus NAD is converted to acetyl CoA CO2 and NADH H within the mitochondria in a process called pyruvate decarboxylation The resulting acetyl CoA enters the citric acid cycle or Krebs Cycle where the acetyl group of the acetyl CoA is converted into carbon dioxide by two decarboxylation reactions with the formation of yet more intra mitochondrial NADH H The intra mitochondrial NADH H is oxidized to NAD by the electron transport chain using oxygen as the final electron acceptor to form water The energy released during this process is used to create a hydrogen ion or proton gradient across the inner membrane of the mitochondrion Finally the proton gradient is used to produce about 2 5 ATP for every NADH H oxidized in a process called oxidative phosphorylation 44 Conversion of carbohydrates into fatty acids and cholesterol edit The pyruvate produced by glycolysis is an important intermediary in the conversion of carbohydrates into fatty acids and cholesterol 45 This occurs via the conversion of pyruvate into acetyl CoA in the mitochondrion However this acetyl CoA needs to be transported into cytosol where the synthesis of fatty acids and cholesterol occurs This cannot occur directly To obtain cytosolic acetyl CoA citrate produced by the condensation of acetyl CoA with oxaloacetate is removed from the citric acid cycle and carried across the inner mitochondrial membrane into the cytosol 45 There it is cleaved by ATP citrate lyase into acetyl CoA and oxaloacetate The oxaloacetate is returned to mitochondrion as malate and then back into oxaloacetate to transfer more acetyl CoA out of the mitochondrion The cytosolic acetyl CoA can be carboxylated by acetyl CoA carboxylase into malonyl CoA the first committed step in the synthesis of fatty acids or it can be combined with acetoacetyl CoA to form 3 hydroxy 3 methylglutaryl CoA HMG CoA which is the rate limiting step controlling the synthesis of cholesterol 46 Cholesterol can be used as is as a structural component of cellular membranes or it can be used to synthesize the steroid hormones bile salts and vitamin D 34 45 46 Conversion of pyruvate into oxaloacetate for the citric acid cycle edit Pyruvate molecules produced by glycolysis are actively transported across the inner mitochondrial membrane and into the matrix where they can either be oxidized and combined with coenzyme A to form CO2 acetyl CoA and NADH 34 or they can be carboxylated by pyruvate carboxylase to form oxaloacetate This latter reaction fills up the amount of oxaloacetate in the citric acid cycle and is therefore an anaplerotic reaction from the Greek meaning to fill up increasing the cycle s capacity to metabolize acetyl CoA when the tissue s energy needs e g in heart and skeletal muscle are suddenly increased by activity 47 In the citric acid cycle all the intermediates e g citrate iso citrate alpha ketoglutarate succinate fumarate malate and oxaloacetate are regenerated during each turn of the cycle Adding more of any of these intermediates to the mitochondrion therefore means that that additional amount is retained within the cycle increasing all the other intermediates as one is converted into the other Hence the addition of oxaloacetate greatly increases the amounts of all the citric acid intermediates thereby increasing the cycle s capacity to metabolize acetyl CoA converting its acetate component into CO2 and water with the release of enough energy to form 11 ATP and 1 GTP molecule for each additional molecule of acetyl CoA that combines with oxaloacetate in the cycle 47 To cataplerotically remove oxaloacetate from the citric cycle malate can be transported from the mitochondrion into the cytoplasm decreasing the amount of oxaloacetate that can be regenerated 47 Furthermore citric acid intermediates are constantly used to form a variety of substances such as the purines pyrimidines and porphyrins 47 Intermediates for other pathways editThis article concentrates on the catabolic role of glycolysis with regard to converting potential chemical energy to usable chemical energy during the oxidation of glucose to pyruvate Many of the metabolites in the glycolytic pathway are also used by anabolic pathways and as a consequence flux through the pathway is critical to maintain a supply of carbon skeletons for biosynthesis citation needed The following metabolic pathways are all strongly reliant on glycolysis as a source of metabolites and many more Pentose phosphate pathway which begins with the dehydrogenation of glucose 6 phosphate the first intermediate to be produced by glycolysis produces various pentose sugars and NADPH for the synthesis of fatty acids and cholesterol Glycogen synthesis also starts with glucose 6 phosphate at the beginning of the glycolytic pathway Glycerol for the formation of triglycerides and phospholipids is produced from the glycolytic intermediate glyceraldehyde 3 phosphate Various post glycolytic pathways Fatty acid synthesis Cholesterol synthesis The citric acid cycle which in turn leads to Amino acid synthesis Nucleotide synthesis Tetrapyrrole synthesis dd Although gluconeogenesis and glycolysis share many intermediates the one is not functionally a branch or tributary of the other There are two regulatory steps in both pathways which when active in the one pathway are automatically inactive in the other The two processes can therefore not be simultaneously active 48 Indeed if both sets of reactions were highly active at the same time the net result would be the hydrolysis of four high energy phosphate bonds two ATP and two GTP per reaction cycle 48 NAD is the oxidizing agent in glycolysis as it is in most other energy yielding metabolic reactions e g beta oxidation of fatty acids and during the citric acid cycle The NADH thus produced is primarily used to ultimately transfer electrons to O2 to produce water or when O2 is not available to produced compounds such as lactate or ethanol see Anoxic regeneration of NAD above NADH is rarely used for synthetic processes the notable exception being gluconeogenesis During fatty acid and cholesterol synthesis the reducing agent is NADPH This difference exemplifies a general principle that NADPH is consumed during biosynthetic reactions whereas NADH is generated in energy yielding reactions 48 The source of the NADPH is two fold When malate is oxidatively decarboxylated by NADP linked malic enzyme pyruvate CO2 and NADPH are formed NADPH is also formed by the pentose phosphate pathway which converts glucose into ribose which can be used in synthesis of nucleotides and nucleic acids or it can be catabolized to pyruvate 48 Glycolysis in disease editDiabetes edit Cellular uptake of glucose occurs in response to insulin signals and glucose is subsequently broken down through glycolysis lowering blood sugar levels However the low insulin levels seen in diabetes result in hyperglycemia where glucose levels in the blood rise and glucose is not properly taken up by cells Hepatocytes further contribute to this hyperglycemia through gluconeogenesis Glycolysis in hepatocytes controls hepatic glucose production and when glucose is overproduced by the liver without having a means of being broken down by the body hyperglycemia results 49 Genetic diseases edit Glycolytic mutations are generally rare due to importance of the metabolic pathway the majority of occurring mutations result in an inability of the cell to respire and therefore cause the death of the cell at an early stage However some mutations glycogen storage diseases and other inborn errors of carbohydrate metabolism are seen with one notable example being pyruvate kinase deficiency leading to chronic hemolytic anemia citation needed Cancer edit Malignant tumor cells perform glycolysis at a rate that is ten times faster than their noncancerous tissue counterparts 50 During their genesis limited capillary support often results in hypoxia decreased O2 supply within the tumor cells Thus these cells rely on anaerobic metabolic processes such as glycolysis for ATP adenosine triphosphate Some tumor cells overexpress specific glycolytic enzymes which result in higher rates of glycolysis 51 Often these enzymes are Isoenzymes of traditional glycolysis enzymes that vary in their susceptibility to traditional feedback inhibition The increase in glycolytic activity ultimately counteracts the effects of hypoxia by generating sufficient ATP from this anaerobic pathway 52 This phenomenon was first described in 1930 by Otto Warburg and is referred to as the Warburg effect The Warburg hypothesis claims that cancer is primarily caused by dysfunctionality in mitochondrial metabolism rather than because of the uncontrolled growth of cells A number of theories have been advanced to explain the Warburg effect One such theory suggests that the increased glycolysis is a normal protective process of the body and that malignant change could be primarily caused by energy metabolism 53 This high glycolysis rate has important medical applications as high aerobic glycolysis by malignant tumors is utilized clinically to diagnose and monitor treatment responses of cancers by imaging uptake of 2 18F 2 deoxyglucose FDG a radioactive modified hexokinase substrate with positron emission tomography PET 54 55 There is ongoing research to affect mitochondrial metabolism and treat cancer by reducing glycolysis and thus starving cancerous cells in various new ways including a ketogenic diet 56 57 58 Interactive pathway map editThe diagram below shows human protein names Names in other organisms may be different and the number of isozymes such as HK1 HK2 is likely to be different too Click on genes proteins and metabolites below to link to respective articles 1 File nbsp nbsp alt Glycolysis and Gluconeogenesis edit Glycolysis and Gluconeogenesis edit The interactive pathway map can be edited at WikiPathways GlycolysisGluconeogenesis WP534 Alternative nomenclature editSome of the metabolites in glycolysis have alternative names and nomenclature In part this is because some of them are common to other pathways such as the Calvin cycle This article Alternative1 Glucose Glc Dextrose2 Glucose 6 phosphate G6P3 Fructose 6 phosphate F6P4 Fructose 1 6 bisphosphate F1 6BP Fructose 1 6 diphosphate FBP FDP F1 6DP5 Dihydroxyacetone phosphate DHAP Glycerone phosphate6 Glyceraldehyde 3 phosphate GADP 3 Phosphoglyceraldehyde PGAL G3P GALP GAP TP7 1 3 Bisphosphoglycerate 1 3BPG Glycerate 1 3 bisphosphate glycerate 1 3 diphosphate 1 3 diphosphoglycerate PGAP BPG DPG8 3 Phosphoglycerate 3PG Glycerate 3 phosphate PGA GP9 2 Phosphoglycerate 2PG Glycerate 2 phosphate10 Phosphoenolpyruvate PEP11 Pyruvate Pyr Pyruvic acid conjugate baseStructure of glycolysis components in Fischer projections and polygonal model editThe intermediates of glycolysis depicted in Fischer projections show the chemical changing step by step Such image can be compared to polygonal model representation 59 Another comparation of Fischer projections and Poligonal Model in glycolysis is shown in a video 60 Video animations in the same channel in YouTube can be seen for another metabolic pathway Krebs Cycle and the representation and applying of Polygonal Model in Organic Chemistry 61 nbsp Glycolysis Structure of anaerobic glycolysis components showed using Fischer projections left and polygonal model right The compounds correspond to glucose GLU glucose 6 phosphate G6P fructose 6 phosphate F6P fructose 1 6 bisphosphate F16BP dihydroxyacetone phosphate DHAP glyceraldehyde 3 phosphate GA3P 1 3 bisphosphoglycerate 13BPG 3 phosphoglycerate 3PG 2 phosphoglycerate 2PG phosphoenolpyruvate PEP pyruvate PIR and lactate LAC The enzymes which participate of this pathway are indicated by underlined numbers and correspond to hexokinase 1 glucose 6 phosphate isomerase 2 phosphofructokinase 1 3 fructose bisphosphate aldolase 4 triosephosphate isomerase 5 glyceraldehyde 3 phosphate dehydrogenase 5 phosphoglycerate kinase 7 phosphoglycerate mutase 8 phosphopyruvate hydratase enolase 9 pyruvate kinase 10 and lactate dehydrogenase 11 The participant coenzymes NAD NADH H ATP and ADP inorganic phosphate H2O and CO2 were omitted in these representations The phosphorylation reactions from ATP as well the ADP phosphorylation reactions in later steps of glycolysis are shown as P respectively entering or going out the pathway The oxireduction reactions using NAD or NADH are observed as hydrogens 2H going out or entering the pathway See also edit nbsp Biology portal nbsp Wikimedia Commons has media related to Glycolysis Carbohydrate catabolism Citric acid cycle Cori cycle Fermentation biochemistry Gluconeogenesis Glycolytic oscillation Glycogenoses glycogen storage diseases Inborn errors of carbohydrate metabolism Pentose phosphate pathway Pyruvate decarboxylation Triose kinaseReferences edit Alfarouk KO Verduzco D Rauch C Muddathir AK Adil HH Elhassan GO et al 18 December 2014 Glycolysis tumor metabolism cancer growth and dissemination A new pH based etiopathogenic perspective and therapeutic approach to an old cancer question Oncoscience 1 12 777 802 doi 10 18632 oncoscience 109 PMC 4303887 PMID 25621294 Romano AH Conway T 1996 Evolution of carbohydrate metabolic pathways Research in Microbiology 147 6 7 448 455 doi 10 1016 0923 2508 96 83998 2 PMID 9084754 Keller MA Turchyn AV Ralser M April 2014 Non enzymatic glycolysis and pentose phosphate pathway like reactions in a plausible Archean ocean Molecular Systems Biology 10 4 725 doi 10 1002 msb 20145228 PMC 4023395 PMID 24771084 Kim BH Gadd GM 2011 Bacterial Physiology and Metabolism 3rd edition a b c Mehta S 20 September 2011 Glycolysis Animation and Notes PharmaXchange a b Lane AN Fan TW Higashi RM 2009 Metabolic acidosis and the importance of balanced equations Metabolomics 5 2 163 165 doi 10 1007 s11306 008 0142 2 S2CID 35500999 Barnett JA April 2003 A history of research on yeasts 5 the fermentation pathway Yeast 20 6 509 543 doi 10 1002 yea 986 PMID 12722184 S2CID 26805351 Louis Pasteur and Alcoholic Fermentation www pasteurbrewing com Archived from the original on 2011 01 13 Retrieved 2016 02 23 Alba Lois L Segal Kischinevzky C January 2010 Yeast fermentation and the making of beer and wine Nature Education 3 9 17 Kohler R 1971 03 01 The background to Eduard Buchner s discovery of cell free fermentation Journal of the History of Biology 4 1 35 61 doi 10 1007 BF00356976 PMID 11609437 S2CID 46573308 Eduard Buchner Biographical www nobelprize org Retrieved 2016 02 23 a b Cornish Bowden A 1997 Harden and Young s Discovery of Fructose 1 6 Bisphosphate New Beer in an Old Bottle Eduard Buchner and the Growth of Biochemical Knowledge Valencia Spain a href Template Cite book html title Template Cite book cite book a CS1 maint location missing publisher link a b Palmer G Chapter 3 The History of Glycolysis An Example of a Linear Metabolic Pathway Bios 302 PDF Archived from the original PDF on 18 November 2017 Otto Meyerhof Biographical www nobelprize org Retrieved 2016 02 23 a b c Kresge N Simoni RD Hill RL January 2005 Otto Fritz Meyerhof and the elucidation of the glycolytic pathway The Journal of Biological Chemistry 280 4 e3 doi 10 1016 S0021 9258 20 76366 0 PMID 15665335 Embden Gustav Dictionary definition of Embden Gustav Encyclopedia com FREE online dictionary www encyclopedia com Retrieved 2016 02 23 Reeves RE South DJ Blytt HJ Warren LG December 1974 Pyrophosphate D fructose 6 phosphate 1 phosphotransferase A new enzyme with the glycolytic function of 6 phosphofructokinase The Journal of Biological Chemistry 249 24 7737 7741 doi 10 1016 S0021 9258 19 42029 2 PMID 4372217 Selig M Xavier KB Santos H Schonheit P April 1997 Comparative analysis of Embden Meyerhof and Entner Doudoroff glycolytic pathways in hyperthermophilic archaea and the bacterium Thermotoga Archives of Microbiology 167 4 217 232 doi 10 1007 BF03356097 PMID 9075622 S2CID 19489719 Garrett RH Grisham CM 2012 Biochemistry 5th ed Cengage Learning ISBN 978 1 133 10629 6 Berg JM Tymoczko JL Stryer L 2007 Biochemistry 6th ed New York Freeman p 622 ISBN 978 0716787242 a b Garrett R Grisham CM 2005 Biochemistry 3rd ed Belmont CA Thomson Brooks Cole p 584 ISBN 978 0 534 49033 1 Shimizu K Matsuoka Y March 2019 Regulation of glycolytic flux and overflow metabolism depending on the source of energy generation for energy demand Biotechnology Advances 37 2 284 305 doi 10 1016 j biotechadv 2018 12 007 PMID 30576718 S2CID 58591361 a b Chubukov V Gerosa L Kochanowski K Sauer U May 2014 Coordination of microbial metabolism Nature Reviews Microbiology 12 5 327 340 doi 10 1038 nrmicro3238 PMID 24658329 S2CID 28413431 Hochachka PW 1999 Cross Species Studies of Glycolytic Function In Roach RC Wagner PD Hackett PH eds Hypoxia Advances in Experimental Medicine and Biology Vol 474 Boston MA Springer US pp 219 229 doi 10 1007 978 1 4615 4711 2 18 ISBN 978 1 4613 7134 2 PMID 10635004 Lemaigre FP Rousseau GG October 1994 Transcriptional control of genes that regulate glycolysis and gluconeogenesis in adult liver The Biochemical Journal 303 1 1 14 doi 10 1042 bj3030001 PMC 1137548 PMID 7945228 Bian X Jiang H Meng Y Li YP Fang J Lu Z March 2022 Regulation of gene expression by glycolytic and gluconeogenic enzymes Trends in Cell Biology 32 9 786 799 doi 10 1016 j tcb 2022 02 003 PMID 35300892 S2CID 247459973 a b Gerosa L Sauer U August 2011 Regulation and control of metabolic fluxes in microbes Current Opinion in Biotechnology 22 4 566 575 doi 10 1016 j copbio 2011 04 016 PMID 21600757 Chowdhury S Hepper S Lodi MK Saier MH Uetz P April 2021 The Protein Interactome of Glycolysis in Escherichia coli Proteomes 9 2 16 doi 10 3390 proteomes9020016 PMC 8167557 PMID 33917325 Rodionova IA Zhang Z Mehla J Goodacre N Babu M Emili A et al August 2017 The phosphocarrier protein HPr of the bacterial phosphotransferase system globally regulates energy metabolism by directly interacting with multiple enzymes in Escherichia coli The Journal of Biological Chemistry 292 34 14250 14257 doi 10 1074 jbc M117 795294 PMC 5572926 PMID 28634232 Pisithkul T Patel NM Amador Noguez D April 2015 Post translational modifications as key regulators of bacterial metabolic fluxes Current Opinion in Microbiology 24 29 37 doi 10 1016 j mib 2014 12 006 PMID 25597444 a b c Koeslag JH Saunders PT Terblanche E June 2003 A reappraisal of the blood glucose homeostat which comprehensively explains the type 2 diabetes mellitus syndrome X complex The Journal of Physiology published 2003 549 Pt 2 333 346 doi 10 1113 jphysiol 2002 037895 PMC 2342944 PMID 12717005 a b c d e Stryer L 1995 Glycolysis Biochemistry Fourth ed New York W H Freeman and Company pp 483 508 ISBN 0 7167 2009 4 Stryer L 1995 Biochemistry Fourth ed New York W H Freeman and Company p 773 ISBN 0 7167 2009 4 a b c Voet D Voet JG Pratt CW 2006 Fundamentals of Biochemistry 2nd ed John Wiley and Sons Inc pp 547 556 ISBN 978 0 471 21495 3 Beis I Newsholme EA October 1975 The contents of adenine nucleotides phosphagens and some glycolytic intermediates in resting muscles from vertebrates and invertebrates The Biochemical Journal 152 1 23 32 doi 10 1042 bj1520023 PMC 1172435 PMID 1212224 Voet D Voet JG 2004 Biochemistry 3rd ed New York John Wiley amp Sons Inc Lackie J 2010 TIGAR Oxford Reference Online Oxford University Press ISBN 9780199549351 Bensaad K Tsuruta A Selak MA Vidal MN Nakano K Bartrons R et al July 2006 TIGAR a p53 inducible regulator of glycolysis and apoptosis Cell 126 1 107 120 doi 10 1016 j cell 2006 05 036 PMID 16839880 S2CID 15006256 TIGAR TP53 induced glycolysis regulatory phosphatase Homo sapiens human Gene NCBI www ncbi nlm nih gov Retrieved 2018 05 17 Carbonell J Feliu JE Marco R Sols A August 1973 Pyruvate kinase Classes of regulatory isoenzymes in mammalian tissues European Journal of Biochemistry 37 1 148 156 doi 10 1111 j 1432 1033 1973 tb02969 x hdl 10261 78345 PMID 4729424 Valentini G Chiarelli L Fortin R Speranza ML Galizzi A Mattevi A June 2000 The allosteric regulation of pyruvate kinase The Journal of Biological Chemistry 275 24 18145 18152 doi 10 1074 jbc m001870200 PMID 10751408 Israelsen WJ Vander Heiden MG July 2015 Pyruvate kinase Function regulation and role in cancer Seminars in Cell amp Developmental Biology 43 43 51 doi 10 1016 j semcdb 2015 08 004 PMC 4662905 PMID 26277545 a b c d Engstrom L 1978 The regulation of liver pyruvate kinase by phosphorylation dephosphorylation Current Topics in Cellular Regulation Elsevier 13 28 51 doi 10 1016 b978 0 12 152813 3 50006 9 ISBN 978 0 12 152813 3 PMID 208818 a b c Stryer L 1995 Oxidative phosphorylation Biochemistry Fourth ed New York W H Freeman and Company pp 537 549 ISBN 0 7167 2009 4 a b c Stryer L 1995 Fatty acid metabolism Biochemistry Fourth ed New York W H Freeman and Company pp 603 628 ISBN 0 7167 2009 4 a b Stryer L 1995 Biosynthesis of membrane lipids and steroids Biochemistry Fourth ed New York W H Freeman and Company pp 691 707 ISBN 0 7167 2009 4 a b c d Stryer L 1995 Citric acid cycle Biochemistry Fourth ed New York W H Freeman and Company pp 509 527 569 579 614 616 638 641 732 735 739 748 770 773 ISBN 0 7167 2009 4 a b c d Stryer L 1995 Biochemistry Fourth ed New York W H Freeman and Company pp 559 565 574 576 614 623 ISBN 0 7167 2009 4 Guo X Li H Xu H Woo S Dong H Lu F et al 2012 08 01 Glycolysis in the control of blood glucose homeostasis Acta Pharmaceutica Sinica B 2 4 358 367 doi 10 1016 j apsb 2012 06 002 ISSN 2211 3835 Alfarouk KO Verduzco D Rauch C Muddathir AK Adil HH Elhassan GO et al 2014 Glycolysis tumor metabolism cancer growth and dissemination A new pH based etiopathogenic perspective and therapeutic approach to an old cancer question Oncoscience 1 12 777 802 doi 10 18632 oncoscience 109 PMC 4303887 PMID 25621294 Alfarouk KO Shayoub ME Muddathir AK Elhassan GO Bashir AH July 2011 Evolution of Tumor Metabolism might Reflect Carcinogenesis as a Reverse Evolution process Dismantling of Multicellularity Cancers 3 3 3002 3017 doi 10 3390 cancers3033002 PMC 3759183 PMID 24310356 Nelson DL Cox MM 2005 Lehninger principles of biochemistry 4th ed New York W H Freeman ISBN 978 0 7167 4339 2 Gold J October 2011 What is Cancer Archived from the original on May 19 2018 Retrieved September 8 2012 Pauwels EK Sturm EJ Bombardieri E Cleton FJ Stokkel MP October 2000 Positron emission tomography with 18F fluorodeoxyglucose Part I Biochemical uptake mechanism and its implication for clinical studies Journal of Cancer Research and Clinical Oncology 126 10 549 59 doi 10 1007 pl00008465 PMID 11043392 S2CID 2725555 PET Scan PET Scan Info Reveals Retrieved December 5 2005 Schwartz L Seyfried T Alfarouk KO Da Veiga Moreira J Fais S April 2017 Out of Warburg effect An effective cancer treatment targeting the tumor specific metabolism and dysregulated pH Seminars in Cancer Biology 43 134 138 doi 10 1016 j semcancer 2017 01 005 PMID 28122260 Schwartz L Supuran CT Alfarouk KO 2017 The Warburg Effect and the Hallmarks of Cancer Anti Cancer Agents in Medicinal Chemistry 17 2 164 170 doi 10 2174 1871520616666161031143301 PMID 27804847 Maroon J Bost J Amos A Zuccoli G August 2013 Restricted calorie ketogenic diet for the treatment of glioblastoma multiforme Journal of Child Neurology 28 8 1002 1008 doi 10 1177 0883073813488670 PMID 23670248 S2CID 1994087 Bonafe CF Bispo JA de Jesus MB January 2018 The polygonal model A simple representation of biomolecules as a tool for teaching metabolism Biochemistry and Molecular Biology Education 46 1 66 75 doi 10 1002 bmb 21093 PMID 29131491 S2CID 31317102 Bonafe C 23 September 2019 Introduction to Polygonal Model PART 1 Glycolysis and Structure of the Participant Molecules YouTube Archived from the original on 2021 11 04 Metabolism Animation and Polygonal Model YouTube Retrieved 2019 12 11 External links editA Detailed Glycolysis Animation provided by IUBMB Adobe Flash Required The Glycolytic enzymes in Glycolysis at RCSB PDB Glycolytic cycle with animations at wdv com Metabolism Cellular Respiration and Photosynthesis The Virtual Library of Biochemistry Molecular Biology and Cell Biology The chemical logic behind glycolysis at ufp pt Expasy biochemical pathways poster at ExPASy MedicalMnemonics com 317 5468 metpath Interactive representation of glycolysis Retrieved from https en wikipedia org w index php title Glycolysis amp oldid 1204587647, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.