Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(21)2022 Oct 30.
Article in English | MEDLINE | ID: mdl-36361985

ABSTRACT

Phosphoglycerate mutase (PGAM) is a glycolytic enzyme converting 3-phosphoglycerate to 2-phosphoglycerate, which in mammalian cells is expressed in two isoforms: brain (PGAM1) and muscle (PGAM2). Recently, it was shown that besides its enzymatic function, PGAM2 can be imported to the cell nucleus where it co-localizes with the nucleoli. It was suggested that it functions there to stabilize the nucleolar structure, maintain mRNA expression, and assist in the assembly of new pre-ribosomal subunits. However, the precise mechanism by which the protein translocates to the nucleus is unknown. In this study, we present the first crystal structure of PGAM2, identify the residues involved in the nuclear localization of the protein and propose that PGAM contains a "quaternary nuclear localization sequence (NLS)", i.e., one that consists of residues from different protein chains. Additionally, we identify potential interaction partners for PGAM2 in the nucleoli and demonstrate that 14-3-3ζ/δ is indeed an interaction partner of PGAM2 in the nucleus. We also present evidence that the insulin/IGF1-PI3K-Akt-mTOR signaling pathway is responsible for the nuclear localization of PGAM2.


Subject(s)
Phosphatidylinositol 3-Kinases , Phosphoglycerate Mutase , Animals , Phosphoglycerate Mutase/genetics , Active Transport, Cell Nucleus , Phosphatidylinositol 3-Kinases/metabolism , 14-3-3 Proteins/metabolism , Muscles/metabolism , Mammals/metabolism
2.
Brain Commun ; 3(2): fcab036, 2021.
Article in English | MEDLINE | ID: mdl-33977262

ABSTRACT

Leukodystrophies are genetic disorders of cerebral white matter that almost exclusively have a progressive disease course. We became aware of three members of a family with a disorder characterized by a sudden loss of all previously acquired abilities around 1 year of age followed by almost complete recovery within 2 years. Cerebral MRI and myelin sensitive imaging showed a pronounced demyelination that progressed for several months despite signs of clinical improvement and was followed by remyelination. Exome sequencing did not-identify any mutations in known leukodystrophy genes but revealed a heterozygous variant in the FBP2 gene, c.343G>A, p. Val115Met, shared by the affected family members. Cerebral MRI of other family members demonstrated similar white matter abnormalities in all carriers of the variant in FBP2. The FBP2 gene codes for muscle fructose 1,6-bisphosphatase, an enzyme involved in gluconeogenesis that is highly expressed in brain tissue. Biochemical analysis showed that the variant has a dominant negative effect on enzymatic activity, substrate affinity, cooperativity and thermal stability. Moreover, it also affects the non-canonical functions of muscle fructose 1,6-bisphosphatase involved in mitochondrial protection and regulation of several nuclear processes. In patients' fibroblasts, muscle fructose 1,6-bisphosphatase shows no colocalization with mitochondria and nuclei leading to increased reactive oxygen species production and a disturbed mitochondrial network. In conclusion, the results of this study indicate that the variant in FBP2 disturbs cerebral energy metabolism and is associated with a novel remitting leukodystrophy.

3.
Acta Biochim Pol ; 68(1): 5-14, 2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33502838

ABSTRACT

Muscle fructose-1,6-bisphosphatase (FBPase), which catalyzes the hydrolysis of fructose-1,6-bisphosphate (F1,6BP) to fructose-6-phosphate (F6P) and inorganic phosphate, regulates glucose homeostasis by controlling the glyconeogenic pathway. FBPase requires divalent cations, such as Mg2+, Mn2+, or Zn2+, for its catalytic activity; however, calcium ions inhibit the muscle isoform of FBPase by interrupting the movement of the catalytic loop. It has been shown that residue E69 in this loop plays a key role in the sensitivity of muscle FBPase towards calcium ions. The study presented here is based on five crystal structures of wild-type human muscle FBPase and its E69Q mutant in complexes with the substrate and product of the enzymatic reaction, namely F1,6BP and F6P. The ligands are bound in the active site of the studied proteins in the same manner and have excellent definition in the electron density maps. In all studied crystals, the homotetrameric enzyme assumes the same cruciform quaternary structure, with the κ angle, which describes the orientation of the upper dimer with respect to the lower dimer, of -85o. This unusual quaternary arrangement of the subunits, characteristic of the R-state of muscle FBPase, is also observed in solution by small-angle X-ray scattering (SAXS).


Subject(s)
Fructose-Bisphosphatase/chemistry , Fructose-Bisphosphatase/metabolism , Muscles/enzymology , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallization , Fructosephosphates/chemistry , Fructosephosphates/metabolism , Humans , Hydrogen Bonding , Hydrolysis , Ligands , Models, Molecular , Molecular Weight , Muscles/metabolism , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Scattering, Small Angle , X-Ray Diffraction/methods
4.
Cell Death Dis ; 10(10): 712, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558701

ABSTRACT

Muscle fructose-1,6-bisphosphate aldolase (ALDOA) is among the most abundant glycolytic enzymes in all cancer cells. Here, we show that the enzyme plays a previously unknown and critical role in a cancer cell survival. Simultaneous inhibition of ALDOA activity and interaction with F-actin cytoskeleton using ALDOA slow-binding inhibitor UM0112176 leads to a rapid cofilin-dependent loss of F-actin stress fibers which is associated with elevated ROS production, inhibition of ATP synthesis, increase in calcium levels, caspase activation and arrested cellular proliferation. These effects can be reproduced by silencing of ALDOA. The mechanism of pharmacological action is, however, independent of the catalytic function of the enzyme, specific to cancer cells, and is most deleterious to cells undergoing the epithelial-mesenchymal transition, a process facilitating cancer cell invasion. Our results demonstrate that the overabundance of ALDOA in cancer cells is associated with its moonlighting rather than catalytic functions. This may have significant implications for development of novel broad-based anti-cancer therapies.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Fructose-Bisphosphate Aldolase/metabolism , Neoplasms/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epithelial-Mesenchymal Transition , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Fructose-Bisphosphate Aldolase/genetics , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , Hexokinase/metabolism , Humans , Ion Channels/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neoplasms/enzymology , Neoplasms/pathology , RNA, Small Interfering , Reactive Oxygen Species/metabolism
5.
Adv Biol Regul ; 72: 41-50, 2019 05.
Article in English | MEDLINE | ID: mdl-30871972

ABSTRACT

Fructose-1,6-bisphosphatase (FBPase) is one of the ancient, evolutionarily conserved enzymes of carbohydrate metabolism. It has been described for a first time in 1943, however, for the next half a century mostly kinetic and structural parameters of animal FBPases have been studied. Discovery of ubiquitous expression of the muscle isozyme of FBPase, thus far considered to merely regulate glycogen synthesis from carbohydrate precursors, and its nuclear localisation in several cell types has risen new interest in the protein, resulting in numerous publications revealing complex functions/properties of FBPase. This review summarises the current knowledge of FBPase in animal cells providing evidence that the enzyme merits the name of moonlighting protein.


Subject(s)
Fructose-Bisphosphatase/metabolism , Glucose/metabolism , Animals , Fructose-Bisphosphatase/chemistry , Glucose/chemistry , Glycogen/biosynthesis , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics
6.
Expert Opin Ther Targets ; 22(10): 833-848, 2018 10.
Article in English | MEDLINE | ID: mdl-30244615

ABSTRACT

INTRODUCTION: Glycogen synthase kinase 3 (GSK3) is at the center of cellular signaling and controls various aspects of brain functions, including development of the nervous system, neuronal plasticity and onset of neurodegenerative disorders. Areas covered: In this review, recent efforts in elucidating the roles of GSK3 in neuronal plasticity and development of brain pathologies; Alzheimer's and Parkinson's disease, schizophrenia, and age-related neurodegeneration are described. The effect of microglia and astrocytes on development of the pathological states is also discussed. Expert opinion: GSK3ß and its signaling pathway partners hold great promise as therapeutic target(s) for a multitude of neurological disorders. Activity of the kinase is often elevated in brain disorders. However, due to the wide range of GSK3 cellular targets, global inhibition of the kinase leads to severe side-effects and GSK3 inhibitors rarely reach Phase-2 clinical trials. Thus, a selective modulation of a specific cellular pool of GSK3 or specific down- or upstream partners of the kinase might provide more efficient anti-neurodegenerative therapies.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Molecular Targeted Therapy , Neurodegenerative Diseases/drug therapy , Aging/pathology , Animals , Astrocytes/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Microglia/metabolism , Neurodegenerative Diseases/physiopathology , Neuronal Plasticity/physiology , Signal Transduction/physiology
7.
Oncotarget ; 8(70): 115420-115433, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383170

ABSTRACT

Muscle fructose 1,6-bisphosphatase (FBP2), besides being a regulatory enzyme of glyconeogenesis also protects mitochondria against calcium stress and plays a key role in regulation of the cell cycle, promoting cardiomyocytes survival. However, in cancer cells, FBP2 acts as an anti-oncogenic/anti-proliferative protein. Here, we show that the physiological function of FBP2 depends both on its level of expression in a cell as well as its oligomerization state. Animal fructose-1,6-bisphosphatases are thought to function as tetramers. We present evidence that FBP2 exists in an equilibrium between tetramers and dimers. The dimeric form is fully active and insensitive to AMP, the main allosteric inhibitor of FBP2. Tetramerization induces the sensitivity of the protein to AMP, but it requires the presence of a hydrophobic central region in which leucine 190 plays a crucial role. Only the tetrameric form of FBP2 is retained in cardiomyocyte cell nucleus whereas only the dimeric form associates with mitochondria and protects them against stress stimuli, such as elevated calcium and H2O2 level. Remarkably, in hypoxic conditions, which are typical for many cancers, FBP2 ceases to interact with mitochondria and loses its pro-survival potential. Our results throw new light on the basis of the diverse role of FBP2 in cells.

8.
Acta Crystallogr D Struct Biol ; 72(Pt 4): 536-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27050133

ABSTRACT

Fructose-1,6-bisphosphatase (FBPase) catalyzes the hydrolysis of fructose 1,6-bisphosphate to fructose 6-phosphate and is a key enzyme of gluconeogenesis and glyconeogenesis and, more generally, of the control of energy metabolism and glucose homeostasis. Vertebrates, and notably Homo sapiens, express two FBPase isoforms. The liver isozyme is expressed mainly in gluconeogenic organs, where it functions as a regulator of glucose synthesis. The muscle isoform is expressed in all cells, and recent studies have demonstrated that its role goes far beyond the enzymatic function, as it can interact with various nuclear and mitochondrial proteins. Even in its enzymatic function, the muscle enzyme is different from the liver isoform, as it is 100-fold more susceptible to allosteric inhibition by AMP and this effect can be abrogated by complex formation with aldolase. All FBPases are homotetramers composed of two intimate dimers: the upper dimer and the lower dimer. They oscillate between two conformational states: the inactive T form when in complex with AMP, and the active R form. Parenthetically, it is noted that bacterial FBPases behave somewhat differently, and in the absence of allosteric activators exist in a tetramer-dimer equilibrium even at relatively high concentrations. [Hines et al. (2007), J. Biol. Chem. 282, 11696-11704]. The T-to-R transition is correlated with the conformation of the key loop L2, which in the T form becomes `disengaged' and unable to participate in the catalytic mechanism. The T states of both isoforms are very similar, with a small twist of the upper dimer relative to the lower dimer. It is shown that at variance with the well studied R form of the liver enzyme, which is flat, the R form of the muscle enzyme is diametrically different, with a perpendicular orientation of the upper and lower dimers. The crystal structure of the muscle-isozyme R form shows that in this arrangement of the tetramer completely new protein surfaces are exposed that are most likely targets for the interactions with various cellular and enzymatic partners. The cruciform R structure is stabilized by a novel `leucine lock', which prevents the key residue, Asp187, from locking loop L2 in the disengaged conformation. In addition, the crystal structures of muscle FBPase in the T conformation with and without AMP strongly suggest that the T-to-R transition is a discrete jump rather than a shift of an equilibrium smooth transition through multiple intermediate states. Finally, using snapshots from three crystal structures of human muscle FBPase, it is conclusively demonstrated that the AMP-binding event is correlated with a ß→α transition at the N-terminus of the protein and with the formation of a new helical structure.


Subject(s)
Fructose-Bisphosphatase/chemistry , Humans , Protein Structure, Quaternary , Protein Structure, Secondary
9.
Oncotarget ; 6(19): 17237-50, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26033454

ABSTRACT

Phosphoglycerate mutase (PGAM), a conserved, glycolytic enzyme has been found in nucleoli of cancer cells. Here, we present evidence that accumulation of PGAM in the nucleolus is a universal phenomenon concerning not only neoplastically transformed but also non-malignant cells. Nucleolar localization of the enzyme is dependent on the presence of the PGAM2 (muscle) subunit and is regulated by insulin/IGF-1-PI3K signaling pathway as well as drugs influencing ribosomal biogenesis. We document that PGAM interacts with several 40S and 60S ribosomal proteins and that silencing of PGAM2 expression results in disturbance of nucleolar structure, inhibition of RNA synthesis and decrease of the mitotic index of squamous cell carcinoma cells. We conclude that presence of PGAM in the nucleolus is a prerequisite for synthesis and initial assembly of new pre-ribosome subunits.


Subject(s)
Cell Nucleolus/metabolism , Phosphoglycerate Mutase/metabolism , Transcription, Genetic/physiology , Animals , Cell Line , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Mass Spectrometry , Mice , Microscopy, Confocal , Phosphatidylinositol 3-Kinases/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL