Mannose phosphate isomerase

Last updated
Mannose-6 phosphate isomerase
3h1m.jpg
Identifiers
EC no. 5.3.1.8
CAS no. 9023-88-5
Databases
IntEnz IntEnz view
BRENDA BRENDA entry
ExPASy NiceZyme view
KEGG KEGG entry
MetaCyc metabolic pathway
PRIAM profile
PDB structures RCSB PDB PDBe PDBsum
Search
PMC articles
PubMed articles
NCBI proteins

Mannose-6 phosphate isomerase (MPI), alternately phosphomannose isomerase (PMI) (EC 5.3.1.8) is an enzyme which facilitates the interconversion of fructose 6-phosphate (F6P) and mannose-6-phosphate (M6P). Mannose-6-phosphate isomerase may also enable the synthesis of GDP-mannose in eukaryotic organisms. M6P can be converted to F6P by mannose-6-phosphate isomerase and subsequently utilized in several metabolic pathways including glycolysis and capsular polysaccharide biosynthesis. [1] PMI is monomeric and metallodependent on zinc as a cofactor ligand. [2] PMI is inhibited by erythrose 4-phosphate, mannitol 1-phosphate, and to a lesser extent, the alpha anomer of M6P. [3]

Contents

Mechanism

Rough mechanism showing cis-enediol intermediate PMIroughmechanism.png
Rough mechanism showing cis-enediol intermediate

MPI must convert an aldose (mannose) to a ketose (fructose), in addition to opening and closing the rings for these sugars. [4] In humans a mechanism has been suggested which involves a hydrogen transfer between C1 and C2, mediated by Tyr278, and the movement of a proton from O1 and O2 mediated by the associated Zn2+ ion. [5] The ring opening step may be catalyzed by His99 and Asp270, and isomerization is likely a cis-enediol mechanism. [6] [7]

PMI shows a high degree of selectivity for the beta anomer of M6P, and the alpha anomer has no activity, and may in fact act as an inhibitor. [8] Phosphoglucose isomerase (PGI) has a very similar function to PMI, (as it catalyzes the interconversion of glucose 6-phosphate and F6P) however PGI can anomerize alpha and beta G6P, and may also catalyze the conversion of alpha M6P to beta M6P, while PMI may not anomerize M6P. [8] It is likely the cis-enediol intermediate formed by PMI is the same intermediate formed by PGI. [9]

Structure

MPI is composed of 440 amino acid residues, with one active site and one zinc ion (Zn2+) ligand. Amino acids GLN 111A, HIS 113A, GLU 138A, HIS 285A, and HOH 798A are involved with the zinc ligand bonding. [2] Structure differs from phosphoglucose isomerase by a threonine residue (Thr291) which creates extra space in the active site of PMI to accommodate the different stereochemistry of M6P. This increased space created by the threonine allows the rotation of the C2-C3 bond, which enables the necessary cis-enediol intermediate to be formed. Because mannose and glucose are stereoisomers at C2, which is crucial to the mechanism for both enzymes, PMI must allow extra space in the active site to allow for rotation of mannose to form the cis-enediol intermediate, which is the same intermediate formed by phosphoglucose isomerase. [10]

Biological relevance

PMI has several contributions to necessary metabolic pathways. It enables cells to transform M6P into F6P, which can then be entered into Glycolysis. PMI also allows cells to convert F6P into M6P, which is a common glycolytic cellular identifier for cellular transport and cell membrane identification in prokaryotic and eukaryotic organisms. [4]

Medical relevance

PMI may be helpful in the development of new antifungal treatments, as lack of PMI activity in yeast cells can lead to cell lysis and the enzyme may be a target for inhibition. [11] This may be due to the role of PMI in the formation cell walls and capsular polysaccharide biosynthesis. Additionally M6P is an important signaling molecule, especially for transport to lysosomes: disorders affecting MPI activity may affect cellular ability to quickly produce M6P from plentiful F6P, and therefore vesicle traffic to lysosomes and endosomes may be altered, potentially negatively impacting the cell. [12]

See also

Notes

  1. EBI Database, IPRO16305 Mannose-6-phosphate Isomerase.
  2. 1 2 "1pmi". PDBe.
  3. Gao H, Yu Y, Leary JA (September 2005). "Mechanism and kinetics of metalloenzyme phosphomannose isomerase: measurement of dissociation constants and effect of zinc binding using ESI-FTICR mass spectrometry". Analytical Chemistry. 77 (17): 5596–603. doi:10.1021/ac050549m. PMID   16131071.
  4. 1 2 Berg JM, Tymoczko JL, Stryer L (2007). Biochemistry (6th ed.). New York, New York: W.H. Freeman & Co.
  5. Xiao J, Guo Z, Guo Y, Chu F, Sun P (November 2006). "Computational study of human phosphomannose isomerase: Insights from homology modeling and molecular dynamics simulation of enzyme bound substrate". Journal of Molecular Graphics & Modelling. 25 (3): 289–95. doi:10.1016/j.jmgm.2006.01.001. PMID   16488169.
  6. Sagurthi SR, Gowda G, Savithri HS, Murthy MR (July 2009). "Structures of mannose-6-phosphate isomerase from Salmonella typhimurium bound to metal atoms and substrate: implications for catalytic mechanism" (PDF). Acta Crystallographica Section D. 65 (Pt 7): 724–32. doi:10.1107/S0907444909013328. PMID   19564693.
  7. Gracy RW, Noltmann EA (October 1968). "Studies on phosphomannose isomerase. 3. A mechanism for catalysis and for the role of zinc in the enzymatic and the nonenzymatic isomerization". The Journal of Biological Chemistry. 243 (20): 5410–9. PMID   4973622.
  8. 1 2 Rose IA, O'Connell EL, Schray KJ (March 1973). "Mannose 6-phosphate: anomeric form used by phosphomannose isomerase and its 1-epimerization by phosphoglucose isomerase". The Journal of Biological Chemistry. 248 (6): 2232–4. PMID   4570473.
  9. Wu R, Xie H, Cao Z, Mo Y (June 2008). "Combined quantum mechanics/molecular mechanics study on the reversible isomerization of glucose and fructose catalyzed by Pyrococcus furiosus phosphoglucose isomerase" (PDF). Journal of the American Chemical Society. 130 (22): 7022–31. doi:10.1021/ja710633c. PMID   18470986.
  10. Swan MK, Hansen T, Schönheit P, Davies C (November 2004). "Structural basis for phosphomannose isomerase activity in phosphoglucose isomerase from Pyrobaculum aerophilum: a subtle difference between distantly related enzymes". Biochemistry. 43 (44): 14088–95. doi:10.1021/bi048608y. PMID   15518558.
  11. Cleasby A, Wonacott A, Skarzynski T, Hubbard RE, Davies GJ, Proudfoot AE, Bernard AR, Payton MA, Wells TN (May 1996). "The x-ray crystal structure of phosphomannose isomerase from Candida albicans at 1.7 angstrom resolution". Nature Structural Biology. 3 (5): 470–9. doi:10.1038/nsb0596-470. PMID   8612079.
  12. Jaeken J, Matthijs G (2001). "Congenital disorders of glycosylation". Annual Review of Genomics and Human Genetics. 2: 129–51. doi:10.1146/annurev.genom.2.1.129. PMID   11701646.

Related Research Articles

<span class="mw-page-title-main">Glycolysis</span> Catabolic pathway

Glycolysis is the metabolic pathway that converts glucose 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). Glycolysis is a sequence of ten reactions catalyzed by enzymes.

<span class="mw-page-title-main">Phosphorylation</span> Chemical process of introducing a phosphate

In biochemistry, phosphorylation is the attachment of a phosphate group to a molecule or an ion. This process and its inverse, dephosphorylation, are common in biology. Protein phosphorylation often activates many enzymes.

A congenital disorder of glycosylation is one of several rare inborn errors of metabolism in which glycosylation of a variety of tissue proteins and/or lipids is deficient or defective. Congenital disorders of glycosylation are sometimes known as CDG syndromes. They often cause serious, sometimes fatal, malfunction of several different organ systems in affected infants. The most common sub-type is PMM2-CDG where the genetic defect leads to the loss of phosphomannomutase 2 (PMM2), the enzyme responsible for the conversion of mannose-6-phosphate into mannose-1-phosphate

<span class="mw-page-title-main">Mannose</span> Chemical compound

Mannose is a sugar monomer of the aldohexose series of carbohydrates. It is a C-2 epimer of glucose. Mannose is important in human metabolism, especially in the glycosylation of certain proteins. Several congenital disorders of glycosylation are associated with mutations in enzymes involved in mannose metabolism.

<span class="mw-page-title-main">Tumor hypoxia</span> Situation where tumor cells have been deprived of oxygen

Tumor hypoxia is the situation where tumor cells have been deprived of oxygen. As a tumor grows, it rapidly outgrows its blood supply, leaving portions of the tumor with regions where the oxygen concentration is significantly lower than in healthy tissues. Hypoxic microenvironements in solid tumors are a result of available oxygen being consumed within 70 to 150 μm of tumour vasculature by rapidly proliferating tumor cells thus limiting the amount of oxygen available to diffuse further into the tumor tissue. In order to support continuous growth and proliferation in challenging hypoxic environments, cancer cells are found to alter their metabolism. Furthermore, hypoxia is known to change cell behavior and is associated with extracellular matrix remodeling and increased migratory and metastatic behavior.

Glycosylation is the reaction in which a carbohydrate, i.e. a glycosyl donor, is attached to a hydroxyl or other functional group of another molecule in order to form a glycoconjugate. In biology, glycosylation usually refers to an enzyme-catalysed reaction, whereas glycation may refer to a non-enzymatic reaction.

<span class="mw-page-title-main">Phosphofructokinase 1</span> Class of enzymes

Phosphofructokinase-1 (PFK-1) is one of the most important regulatory enzymes of glycolysis. It is an allosteric enzyme made of 4 subunits and controlled by many activators and inhibitors. PFK-1 catalyzes the important "committed" step of glycolysis, the conversion of fructose 6-phosphate and ATP to fructose 1,6-bisphosphate and ADP. Glycolysis is the foundation for respiration, both anaerobic and aerobic. Because phosphofructokinase (PFK) catalyzes the ATP-dependent phosphorylation to convert fructose-6-phosphate into fructose 1,6-bisphosphate and ADP, it is one of the key regulatory steps of glycolysis. PFK is able to regulate glycolysis through allosteric inhibition, and in this way, the cell can increase or decrease the rate of glycolysis in response to the cell's energy requirements. For example, a high ratio of ATP to ADP will inhibit PFK and glycolysis. The key difference between the regulation of PFK in eukaryotes and prokaryotes is that in eukaryotes PFK is activated by fructose 2,6-bisphosphate. The purpose of fructose 2,6-bisphosphate is to supersede ATP inhibition, thus allowing eukaryotes to have greater sensitivity to regulation by hormones like glucagon and insulin.

<span class="mw-page-title-main">Phosphoglucomutase</span>

Phosphoglucomutase is an enzyme that transfers a phosphate group on an α-D-glucose monomer from the 1 to the 6 position in the forward direction or the 6 to the 1 position in the reverse direction.

Isomerases are a general class of enzymes that convert a molecule from one isomer to another. Isomerases facilitate intramolecular rearrangements in which bonds are broken and formed. The general form of such a reaction is as follows:

A tetrose is a monosaccharide with 4 carbon atoms. They have either an aldehyde functional group in position 1 (aldotetroses) or a ketone functional group in position 2 (ketotetroses).

<span class="mw-page-title-main">Glucose-6-phosphate isomerase</span> Mammalian protein found in Homo sapiens

Glucose-6-phosphate isomerase (GPI), alternatively known as phosphoglucose isomerase/phosphoglucoisomerase (PGI) or phosphohexose isomerase (PHI), is an enzyme that in humans is encoded by the GPI gene on chromosome 19. This gene encodes a member of the glucose phosphate isomerase protein family. The encoded protein has been identified as a moonlighting protein based on its ability to perform mechanistically distinct functions. In the cytoplasm, the gene product functions as a glycolytic enzyme that interconverts glucose-6-phosphate (G6P) and fructose-6-phosphate (F6P). Extracellularly, the encoded protein functions as a neurotrophic factor that promotes survival of skeletal motor neurons and sensory neurons, and as a lymphokine that induces immunoglobulin secretion. The encoded protein is also referred to as autocrine motility factor (AMF) based on an additional function as a tumor-secreted cytokine and angiogenic factor. Defects in this gene are the cause of nonspherocytic hemolytic anemia, and a severe enzyme deficiency can be associated with hydrops fetalis, immediate neonatal death and neurological impairment. Alternative splicing results in multiple transcript variants. [provided by RefSeq, Jan 2014]

<span class="mw-page-title-main">Triosephosphate isomerase</span> Enzyme involved in glycolysis

Triose-phosphate isomerase is an enzyme that catalyzes the reversible interconversion of the triose phosphate isomers dihydroxyacetone phosphate and D-glyceraldehyde 3-phosphate.

<span class="mw-page-title-main">Ribose 5-phosphate</span> Chemical compound

Ribose 5-phosphate (R5P) is both a product and an intermediate of the pentose phosphate pathway. The last step of the oxidative reactions in the pentose phosphate pathway is the production of ribulose 5-phosphate. Depending on the body's state, ribulose 5-phosphate can reversibly isomerize to ribose 5-phosphate. Ribulose 5-phosphate can alternatively undergo a series of isomerizations as well as transaldolations and transketolations that result in the production of other pentose phosphates as well as fructose 6-phosphate and glyceraldehyde 3-phosphate.

<span class="mw-page-title-main">Mannose 6-phosphate</span> Chemical compound

Mannose-6-phosphate (M6P) is a molecule bound by lectin in the immune system. M6P is converted to fructose 6-phosphate by mannose phosphate isomerase.

<span class="mw-page-title-main">Sucrose phosphorylase</span>

Sucrose phosphorylase is an important enzyme in the metabolism of sucrose and regulation of other metabolic intermediates. Sucrose phosphorylase is in the class of hexosyltransferases. More specifically it has been placed in the retaining glycoside hydrolases family although it catalyzes a transglycosidation rather than hydrolysis. Sucrose phosphorylase catalyzes the conversion of sucrose to D-fructose and α-D-glucose-1-phosphate. It has been shown in multiple experiments that the enzyme catalyzes this conversion by a double displacement mechanism.

<span class="mw-page-title-main">Phosphomannomutase</span>

In enzymology, a phosphomannomutase is an enzyme that catalyzes the chemical reaction

In enzymology, a mannose-1-phosphate guanylyltransferase is an enzyme that catalyzes the chemical reaction

<span class="mw-page-title-main">PMM1</span> Protein-coding gene in the species Homo sapiens

Phosphomannomutase 1 is an enzyme that in humans is encoded by the PMM1 gene.

<span class="mw-page-title-main">TPI1</span> Protein-coding gene in the species Homo sapiens

Triosephosphate isomerase is an enzyme that in humans is encoded by the TPI1 gene.

<span class="mw-page-title-main">MPI-CDG</span> Medical condition

MPI-CDG is an autosomal recessive congenital disorder of glycosylation caused by biallelic pathogenic variants in MPI. The clinical symptoms in MPI-CDG are caused by deficient activity of the enzyme mannose phosphate isomerase. Clinically, the most common symptoms of MPI-CDG are chronic diarrhea, failure to thrive, protein-losing enteropathy, and coagulopathy. MPI-CDG differs from most other described glycosylation disorders due to its lack of central nervous system involvement, and because it has treatment options besides supportive care. Treatment with oral mannose has been shown to improve most symptoms of the disease. If left untreated, MPI-CDG can be fatal. MPI-CDG was previously known as CDG-IB. The disorder was first described clinically in 1986, and the underlying genetic defect was identified in 1998.