Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
EMBO J ; 43(12): 2368-2396, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750259

ABSTRACT

Phosphoglycerate mutase 1 (PGAM1) is a key node enzyme that diverts the metabolic reactions from glycolysis into its shunts to support macromolecule biosynthesis for rapid and sustainable cell proliferation. It is prevalent that PGAM1 activity is upregulated in various tumors; however, the underlying mechanism remains unclear. Here, we unveil that pyruvate kinase M2 (PKM2) moonlights as a histidine kinase in a phosphoenolpyruvate (PEP)-dependent manner to catalyze PGAM1 H11 phosphorylation, that is essential for PGAM1 activity. Moreover, monomeric and dimeric but not tetrameric PKM2 are efficient to phosphorylate and activate PGAM1. In response to epidermal growth factor signaling, Src-catalyzed PGAM1 Y119 phosphorylation is a prerequisite for PKM2 binding and the subsequent PGAM1 H11 phosphorylation, which constitutes a discrepancy between tumor and normal cells. A PGAM1-derived pY119-containing cell-permeable peptide or Y119 mutation disrupts the interaction of PGAM1 with PKM2 and PGAM1 H11 phosphorylation, dampening the glycolysis shunts and tumor growth. Together, these results identify a function of PKM2 as a histidine kinase, and illustrate the importance of enzyme crosstalk as a regulatory mode during metabolic reprogramming and tumorigenesis.


Subject(s)
Glycolysis , Phosphoglycerate Mutase , Thyroid Hormones , Humans , Phosphoglycerate Mutase/metabolism , Phosphoglycerate Mutase/genetics , Phosphorylation , Animals , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Mice , Thyroid Hormone-Binding Proteins , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Carrier Proteins/metabolism , Carrier Proteins/genetics
2.
Int J Biol Macromol ; 268(Pt 2): 131547, 2024 May.
Article in English | MEDLINE | ID: mdl-38641281

ABSTRACT

Eicosapentaenoic acid regulates glucose uptake in skeletal muscle and significantly affects whole-body energy metabolism. However, the underlying molecular mechanism remains unclear. Here we report that eicosapentaenoic acid activates phosphoglycerate mutase 2, which mediates the conversion of 2-phosphoglycerate into 3-phosphoglycerate. This enzyme plays a pivotal role in glycerol degradation, thereby facilitating the proliferation and differentiation of satellite cells in skeletal muscle. Interestingly, phosphoglycerate mutase 2 inhibits mitochondrial metabolism, promoting the formation of fast-type muscle fibers. Treatment with eicosapentaenoic acid and phosphoglycerate mutase 2 knockdown induced opposite transcriptomic changes, most of which were enriched in the PI3K-AKT signaling pathway. Phosphoglycerate mutase 2 activated the PI3K-AKT signaling pathway, which inhibited the phosphorylation of FOXO1, and, in turn, inhibited mitochondrial function and promoted the formation of fast-type muscle fibers. Our results suggest that eicosapentaenoic acid promotes skeletal muscle growth and regulates glucose metabolism by targeting phosphoglycerate mutase 2 and activating the PI3K/AKT signaling pathway.


Subject(s)
Eicosapentaenoic Acid , Muscle, Skeletal , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Male , Mice , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Eicosapentaenoic Acid/pharmacology , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Muscle Development/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoglycerate Mutase/metabolism , Phosphoglycerate Mutase/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Swine
3.
J Parasitol ; 110(2): 96-105, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38466806

ABSTRACT

Schistosomiasis is a globally burdensome parasitic disease caused by flatworms (blood flukes) in the genus Schistosoma. The current standard treatment for schistosomiasis is the drug praziquantel, but there is an urgent need to advance novel interventions such as vaccines. Several glycolytic enzymes have been evaluated as vaccine targets for schistosomiasis, and data from these studies are reviewed here. Although these parasites are canonically considered to be intracellular, proteomic analysis has revealed that many schistosome glycolytic enzymes are additionally found at the host-interactive surface. We have recently found that the intravascular stage of Schistosoma mansoni (Sm) expresses the glycolytic enzyme phosphoglycerate mutase (PGM) on the tegumental surface. Live parasites display PGM activity, and suppression of PGM gene expression by RNA interference diminishes surface enzyme activity. Recombinant SmPGM (rSmPGM) can cleave its glycolytic substrate, 3-phosphoglycerate and can both bind to plasminogen and promote its conversion to an active form (plasmin) in vitro, suggesting a moonlighting role for this enzyme in regulating thrombosis in vivo. We found that antibodies in sera from chronically infected mice recognize rSmPGM. We also tested the protective efficacy of rSmPGM as a vaccine in the murine model. Although immunization generates high titers of anti-SmPGM antibodies (against both recombinant and native SmPGM), no significant differences in worm numbers were found between vaccinated and control animals.


Subject(s)
Schistosomiasis mansoni , Schistosomiasis , Vaccines , Animals , Mice , Schistosoma mansoni , Phosphoglycerate Mutase , Schistosomiasis mansoni/prevention & control , Schistosomiasis mansoni/parasitology , Proteomics , Schistosomiasis/prevention & control , Antigens, Helminth , Antibodies, Helminth
4.
Arch Microbiol ; 205(7): 263, 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37316743

ABSTRACT

Candida albicans colonizes oral tissues and causes infectious diseases. Colonization of C. albicans on the oral mucosa and tooth enamel surfaces is established via the interaction between C. albicans adhesins and salivary proteins, forming a film on the oral tissues. Deleted in malignant brain tumors 1 (DMBT1), also known as salivary agglutinin or gp-340, belongs to the scavenger receptor cysteine-rich (SRCR) superfamily. In the oral cavity, immobilized DMBT1 on oral tissues causes microbial adherence. Recently, we demonstrated that C. albicans binds to DMBT1 and isolated a 25-kDa C. albicans adhesin involved in the interaction with the binding domain of DMBT1, namely, SRCRP2. In the present study, we searched for additional DMBT1-binding adhesins in C. albicans. The component isolated here had a molecular mass of 29 kDa and was found to be phosphoglycerate mutase (Gpm1). Isolated Gpm1 inhibited C. albicans binding to SRCRP2 and directly bound to SRCRP2 in a dose-dependent manner. Gpm1 localization on the C. albicans cell wall surface was confirmed by immunostaining. These results suggest that surface-expressed Gpm1 functions as an adhesin for the establishment of C. albicans cells on the oral mucosa and tooth enamel by binding to DMBT1.


Subject(s)
Candida albicans , Phosphoglycerate Mutase , Phosphoglycerate Mutase/genetics , Adhesins, Bacterial , Cell Membrane , Cell Wall
5.
Biochem Biophys Res Commun ; 665: 88-97, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37149987

ABSTRACT

Bacillus anthracis Ser/Thr protein kinase PrkC is necessary for phenotypic memory and spore germination, and the loss of PrkC-dependent phosphorylation events affect the spore development. During sporulation, Bacillus sp. can store 3-Phosphoglycerate (3-PGA) that will be required at the onset of germination when ATP will be necessary. The Phosphoglycerate mutase (Pgm) catalyzes the isomerization of 2-PGA and 3-PGA and is important for spore germination as a key metabolic enzyme that maintains 3-PGA pool at later events. Therefore, regulation of Pgm is important for an efficient spore germination process and metabolic switching. While the increased expression of Pgm in B. anthracis decreases spore germination efficiency, it remains unexplored if PrkC could directly influence Pgm activity. Here, we report the phosphorylation and regulation of Pgm by PrkC and its impact on Pgm stability and catalytic activity. Mass spectrometry revealed Pgm phosphorylation on seven threonine residues. In silico mutational analysis highlighted the role of Thr459 residue towards metal and substrate binding. Altogether, we demonstrated that PrkC-mediated Pgm phosphorylation negatively regulates its activity that is essential to maintain Pgm in its apo-like isoform before germination. This study advances the role of Pgm regulation that represents an important switch for B. anthracis resumption of metabolism and spore germination.


Subject(s)
Bacillus anthracis , Protein Kinases , Phosphorylation , Protein Kinases/metabolism , Bacillus anthracis/metabolism , Phosphoglycerate Mutase/metabolism , Threonine/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/metabolism , Bacterial Proteins/metabolism
6.
Drug Resist Updat ; 68: 100957, 2023 05.
Article in English | MEDLINE | ID: mdl-36990047

ABSTRACT

Resistance to epidermal growth factor receptor (EGFR) inhibitors, from the first-generation erlotinib to the third generation osimertinib, is a clinical challenge in the treatment of patients with EGFR-mutant lung adenocarcinoma. Our previous work found that a novel allosteric inhibitor of phosphoglycerate mutase 1 (PGAM1), HKB99, restrains erlotinib resistance in lung adenocarcinoma cells. However, the role of HKB99 in osimertinib resistance and its underlying molecular mechanism remains to be elucidated. Herein, we found that IL-6/JAK2/STAT3 signaling pathway is aberrantly activated in both erlotinib and osimertinib resistant cells. Importantly, HKB99 significantly blocks the interaction of PGAM1 with JAK2 and STAT3 via the allosteric sites of PGAM1, which leads to inactivation of JAK2/STAT3 and thereby disrupts IL-6/JAK2/STAT3 signaling pathway. Consequently, HKB99 remarkably restores EGFR inhibitor sensitivity and exerts synergistic tumoricidal effect. Additionally, HKB99 alone or in combination with osimertinib down-regulated the level of p-STAT3 in xenograft tumor models. Collectively, this study identifies PGAM1 as a key regulator in IL-6/JAK2/STAT3 axis in the development of resistance to EGFR inhibitors, which could serve as a therapeutic target in lung adenocarcinoma with acquired resistance to EGFR inhibitors.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Interleukin-6/genetics , Interleukin-6/pharmacology , Interleukin-6/therapeutic use , Phosphoglycerate Mutase/metabolism , Phosphoglycerate Mutase/pharmacology , Drug Resistance, Neoplasm , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , ErbB Receptors , Signal Transduction , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation , Cell Line, Tumor , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology
7.
Int Immunopharmacol ; 116: 109773, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36773566

ABSTRACT

Spinal cord injury (SCI) is a high incidence worldwide that causes a heavy physical and psychological burden to patients. It is urgent to further reveal the pathological mechanism and effective treatment of SCI. Mitochondrial dysfunction plays an important role in the disease progression of SCI. As a mitochondrial membrane protein, phosphoglycerate mutase 5 (PGAM5) is mainly involved in mitochondrial function and mitosis to modulate cellular physiological functions, but the roles of PGAM5 in spinal tissues remain to be unreported after SCI. The purpose of this study was to evaluate the role of PGAM5 in SCI mice and its relationship with neuroinflammation. The results showed that the mitochondrial membrane protein PGAM5 was involved in microglia activation after SCI, and PGAM5 deletion could improve mitochondrial dysfunction (including abnormal mtDNA, ATP synthases, and ATP levels, Cyt C expression, and ROS and rGSH levels) in spinal cord tissue after SCI, Arg1/iNOS mRNA level, iNOS expression, and pro-inflammatory cytokines TNF-α, IL-1ß, and IL-18 levels. In vitro, H2O2 increased TNF-α, IL-1ß, and IL-18 levels in BV2 cells, and PGAM5-sh and Nrf2 activators significantly reversed H2O2-induced iNOS expression and proinflammatory cytokine production. Furthermore, IP/Western blotting results revealed that PGAM5-sh treatment significantly reduced the interaction of PGAM5 with Nrf2 and enhanced the nuclear translocation of Nrf2 in BV2 cells. The data suggested that PGAM5 was involved in the cascade of oxidative stress and inflammatory response in microglia via facilitating the expression level of Nrf2 in the nucleus after SCI. It provided a reference for clarifying the pathological mechanism and therapeutic target of SCI.


Subject(s)
Mitochondria , Neuroinflammatory Diseases , Phosphoglycerate Mutase , Spinal Cord Injuries , Animals , Mice , Adenosine Triphosphate/metabolism , Hydrogen Peroxide/metabolism , Interleukin-18/metabolism , Mitochondria/metabolism , Mitochondria/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy , Tumor Necrosis Factor-alpha/metabolism
8.
Toxicol Lett ; 372: 14-24, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36273635

ABSTRACT

The pathophysiology of renal lipid toxicity caused by excess adiposity is not well-understood. Necroptosis, a regulated form of cell death, is involved in injuring renal tubular epithelial cells (RTECs). Phosphoglycerate mutase 5 (PGAM5) is a key downstream effector of necroptosis. This study investigated the underlying mechanism of PGAM5 in promoting lipid-induced necroptosis in RTECs. HK2 cells (an immortalized proximal tubule epithelial cell line) were exposed to oleic acid (OA) to mimic the lipid overload environment in vitro. We found that OA suppressed HK2 cell proliferation, triggered cytoskeleton rupture and cell death. In OA-treated cells, upregulated expression of necroptosis pathway proteins, phosphorylated receptor-interacting protein-1/3 (pRIPK1/3), phosphorylated mixed lineage kinase domain-like protein (pMLKL), PGAM5, phosphorylated dynamin-related protein 1 (pDRP1S616), and downregulated pDRP1S637 expression were observed. This was accompanied by mitochondrial dysfunction (mitochondrial ROS overproduction and decreased mitochondrial membrane potential) and increased cellular necrosis, as reflected by Annexin V/ Propidium Iodide (PI) labeling. OA also induced the accumulation of LC3II and P62, blocking autophagosome fusion with lysosomes. Knockdown of PGAM5 could prevent these OA-induced changes. We propose inhibition of PGAM5 protects lipid-induced RTECs from necroptosis by reducing DRP1-mediated mitochondrial fission and improving mitophagy flux.


Subject(s)
Mitochondrial Dynamics , Mitophagy , Necroptosis , Phosphoglycerate Mutase/metabolism , Epithelial Cells/metabolism , Lipids , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
9.
Cell Biol Int ; 47(4): 776-786, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36576012

ABSTRACT

Gliomas account for about 80% of malignant brain tumors. The incidence of a new brain tumor is 6.4 per 100,000 persons per year with an overall 5-year survival rate of 33.4%. Regardless of the great advances that have been made in recent years, the causes and pathogenesis of glioma remain unclear. Here we study how phosphoglycerate mutase 4 (PGAM4) contributes to glioma. Using a variety of methods to examine glioma cell viability, proliferation, apoptosis, glycolysis, as well as ChIP coanalysis with modified histone H3, we showed that PGAM4 was significantly upregulated in patients with glioma and associated with poor survival. Silencing PGAM4 attenuated cell viability, proliferation, and glycolysis in T98G cells and suppressed tumor growth in vivo, while overexpressing PGAM4 promoted cell viability, proliferation, and glycolysis in U251 cells via regulating glycolysis pathway. Study also revealed that PGAM4 was regulated by EP300-mediated modifications of H3K27ac. PGAM4 silencing inhibited cell viability and proliferation, suppressed tumor growth, and decreased chemoresistance to temozolomide in glioma cells through suppressing glycolysis.


Subject(s)
Brain Neoplasms , Glioma , Humans , Temozolomide/pharmacology , Phosphoglycerate Mutase/metabolism , Drug Resistance, Neoplasm , Glioma/metabolism , Brain Neoplasms/metabolism , Apoptosis , Glycolysis , Cell Line, Tumor , Cell Proliferation
10.
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
11.
Front Plant Sci ; 13: 1039420, 2022.
Article in English | MEDLINE | ID: mdl-36438092

ABSTRACT

Acidovorax citrulli (Ac) is a gram-negative bacterium that causes bacterial fruit blotch (BFB) disease in cucurbit crops including watermelon. However, despite the great economic losses caused by this disease worldwide, Ac-resistant watermelon cultivars have not been developed. Therefore, characterizing the virulence factors/mechanisms of Ac would enable the development of effective control strategies against BFB disease. The 2,3-bisphosphoglycerate-dependent phosphoglycerate mutase (BdpM) is known to participate in the glycolysis and gluconeogenesis pathways. However, the roles of the protein have not been characterized in Ac. To elucidate the functions of BdpmAc (Bdpm in Ac), comparative proteomic analysis and diverse phenotypic assays were conducted using a bdpmAc knockout mutant (bdpmAc:Tn) and a wild-type strain. The virulence of the mutant to watermelon was remarkably reduced in both germinated seed inoculation and leaf infiltration assays. Moreover, the mutant could not grow with fructose or pyruvate as a sole carbon source. However, the growth of the mutant was restored to levels similar to those of the wild-type strain in the presence of both fructose and pyruvate. Comparative proteomic analyses revealed that diverse proteins involved in motility and wall/membrane/envelop biogenesis were differentially abundant. Furthermore, the mutant exhibited decreased biofilm formation and twitching halo size. Interestingly, the mutant exhibited a higher tolerance against osmotic stress. Overall, our findings suggest that BdpmAc affects the virulence, glycolysis/gluconeogenesis, biofilm formation, twitching halo size, and osmotic tolerance of Ac, suggesting that this protein has pleiotropic properties. Collectively, our findings provide fundamental insights into the functions of a previously uncharacterized phosphoglycerate mutase in Ac.

12.
Front Biosci (Landmark Ed) ; 27(9): 262, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36224008

ABSTRACT

BACKGROUND: Enhanced glycolysis occurs in most human cancer cells and is related to chemoresistance. However, detailed mechanisms remain vague. METHODS: Using proteinomics analysis, we found that the glycolytic enzyme Phosphoglycerate mutase 1 (PGAM1) was highly expressed in the paclitaxel-resistant ovarian cancer cell line SKOV3-TR30, as compared to its parental cell line SKOV3. Cell Counting Kit-8 proliferation experiment, plasmids and siRNA transfection, pyruvic acid and lactic acid production detection, immunofluorescence staining of functional mitochondria and oxygen consumption rate and extracellular acidification rate measurement were uesd to assess the glycolytic metabolism and paclitaxel resistance in ovarian cancer cells. The expression and prognostic effect of PGAM1 in 180 ovarian cancer patients were analyzed. RESULTS: SKOV3-TR30 cells display higher glycolytic flux and lower mitochondrial function than SKOV3 cells. Down-regulation of PGAM1 in SKOV3-TR30 cells resulted in decreased paclitaxel resistance. Up-regulation of PGAM1 in SKOV3 cells led to enhanced paclitaxel resistance. Analysis of the glycolytic flux revealed that PGAM1-mediated pyruvic acid or lactic acid production could modulate the capabilities of ovarian cancer cell resistance to paclitaxel. Our data also show high expression of PGAM1 as significantly correlated with reduced overall survival and reduced progression free survival in ovarian cancer patients. CONCLUSIONS: PGAM1 acts to promote paclitaxel resistance via pyruvic acid and/or lactate production in ovarian cancer cells. Inhibiting PGAM1 may provide a new approach to favorably alter paclitaxel resistance in ovarian cancer.


Subject(s)
Ovarian Neoplasms , Paclitaxel , Phosphoglycerate Mutase/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Glycolysis , Humans , Lactic Acid , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Phosphoglycerate Mutase/genetics , Pyruvic Acid , RNA, Small Interfering/metabolism
13.
Eur J Med Chem ; 244: 114798, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36215859

ABSTRACT

Glycolysis is a preferred metabolic pattern of cancer cells. Phosphoglycerate mutase 1 (PGAM1) is a pivotal glycolytic enzyme that catalyzes the reciprocal conversion between 2-phosphoglycerate and 3-phosphoglycerate. It also stimulates anabolic pathways, generates adenosine triphosphate, and keeps redox balance under hypoxic conditions. Mounting evidence supports that PGAM1 is overexpressed in many cancers and promotes their progression. The critical roles of PGAM1 in tumorigenesis make it a promising theranostical target for cancer. The aberrant expression of PGAM1 enables it to become a potential diagnosis gene for several cancers, and its heterogeneous regulations via interacting with its different ligands increase the possibility of it as a target for cancer therapy and discovery of tens of PGAM1 inhibitors, which can provide the potential feasibility for cancer treatment. This review provides insights into structure, function, and regulation of PGAM1, summarizes its mechanism in tumorigenesis, reviews the advanced status of PGAM1 inhibitors in cancer diagnosis and treatment, and finally emphasizes PGAM1 as an appealing theranostical target for cancer.


Subject(s)
Neoplasms , Phosphoglycerate Mutase , Humans , Neoplasms/metabolism , Glycolysis , Carcinogenesis
14.
J Clin Lab Anal ; 36(11): e24718, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36181311

ABSTRACT

BACKGROUND: To identify a novel marker for gastric cancer, we examined the usefulness of phosphoglycerate mutase 1 (PGAM1) as a potential diagnostic marker using isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics and evaluated its clinical significance. METHODS: Proteins from a discovery group of four paired gastric cancer tissues and adjacent gastric tissues were labeled with iTRAQ reagents and then identified and quantified using LC-MS/MS. The expression of PGAM1 was further validated in 139 gastric cancer patients using immunohistochemistry. Furthermore, the correlation of PGAM1 expression with clinical parameters was analyzed. Gene set enrichment analysis (GSEA) was performed to identify gene sets that were activated in PGAM1-overexpressing patients with gastric cancer. RESULTS: PGAM1 was significantly overexpressed in most cancers but particularly so in gastric cancer, with a sensitivity of 82.01% (95% confidence interval [CI]: 75.5%-88.5%) and specificity of 79.13% (95% CI: 72.3%-86%). Its expression was significantly associated with histological grade II and III tumors (p = 0.033), lymph node metastasis (p = 0.031), and TNM III-IV staging (p = 0.025). The area under the receiver operating characteristic (ROC) curve for the detection of PGAM1 overexpression in gastric cancer was 0.718 (p < 0.01). Furthermore, GSEA revealed that several important pathways such as glycolysis pathway and immune pathways were significantly enriched in patients with gastric cancer with PGAM1 overexpression. CONCLUSIONS: This study provided a sensitive method for detecting PGAM1, which may serve as a novel indicator for poor prognosis of gastric cancer, as well as a potent drug target for gastric cancer.


Subject(s)
Phosphoglycerate Mutase , Stomach Neoplasms , Humans , Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism , Stomach Neoplasms/diagnosis , Stomach Neoplasms/metabolism , Chromatography, Liquid , Tandem Mass Spectrometry , Glycolysis , Neoplasm Staging
15.
World J Clin Cases ; 10(14): 4368-4379, 2022 May 16.
Article in English | MEDLINE | ID: mdl-35663086

ABSTRACT

BACKGROUND: Drugs targeting mitochondria can induce mitophagy and restrain proliferation in colorectal cancer (CRC) cells. Phosphoglycerate mutase family member 5 (PGAM5) activates serine/threonine PTEN-induced putative kinase 1/Parkin pathway-mediated mitophagy. However, there are few studies on the clinical and prognostic significance of expression of PGAM5 protein and mitophagy-related protein Parkin in patients. AIM: To assess the clinical significance of PGAM5 and Parkin proteins, as biomarkers for diagnosis and prognosis of CRC, by studying their expression in advanced CRC tissues and their association with clinicopathological parameters. METHODS: The expression of PGAM5 and Parkin in CRC tissues from 100 patients was determined by immunohistochemistry. Each case was evaluated by using a combined scoring method based on signal intensity staining (scored 0-3) and the proportion of positively stained cancer cells (scored 0-4). The final staining score was calculated as the intensity score multiplied by the proportion score. Specimens were categorized as either high or low expression according to the Youden index, and the association between the expression of PGAM5 or Parkin and clinicopathological factors was ascertained. Additionally, we employed western blot to measure PGAM5 and Parkin protein expression in six matched pairs of CRC and adjacent non-tumor tissues. RESULTS: Immunohistochemical and western blot findings showed that both PGAM5 and Parkin protein expression in tumor tissues was significantly higher than that in the adjacent tissues: PGAM5 and Parkin were mainly expressed in the cytoplasm of colonic epithelial cells. PGAM5 and Parkin protein levels were significantly positively correlated in advanced CRC tissues. Moreover, reduced Parkin protein expression was an independent prognostic factor for overall survival and progression-free survival in CRC patients as evinced by multivariate analysis. CONCLUSION: The expression of PGAM5 protein and mitophagy-related protein Parkin has diagnostic significance for CRC and may become new biomarkers. Parkin may be a potential marker for the survival of CRC patients.

16.
Neurochem Int ; 157: 105346, 2022 07.
Article in English | MEDLINE | ID: mdl-35513204

ABSTRACT

Phosphoglycerate mutase 5 (PGAM5), a glycolytic enzyme, plays an important role in cell death and regulation of mitochondrial dynamics. In this study, we investigated the effects of PGAM5 on oxidative stress in HT22 hippocampal cells and ischemic damage in the gerbil hippocampus to elucidate the role of PGAM5 in oxidative and ischemic stress. Constructs were designed with a PEP-1 expression vector to facilitate the intracellular delivery of PGAM5 proteins. We observed time- and concentration-dependent increases in the intracellular delivery of the PEP-1-PGAM5 protein, but not its control protein (PGAM5), in HT22 cells, and morphologically demonstrated the localization of the transduced protein, which was stably expressed in the cytoplasm after 12 h of PEP-1-PGAM5 treatment. PEP-1-PGAM5 treatment significantly ameliorated cell death, reactive oxygen species formation, DNA fragmentation, and the reduction of cell proliferation induced by H2O2 treatment in HT22 cells. In addition, PEP-1-PGAM5 was effectively delivered to the gerbil hippocampus 8 h after treatment, and ischemia-induced hyperlocomotion and neuronal death in the hippocampal CA1 region were significantly alleviated 1 and 4 days after ischemia, respectively. Ischemia-induced microglial activation was also mitigated by treatment with 1.0 mg/kg PEP-1-PGAM5. At 3 h after ischemia, PEP-1-PGAM5 treatment significantly ameliorated the increase in lipid peroxidation, as assessed by malondialdehyde and hydroperoxide levels, and decreased glutathione levels (increases in glutathione disulfide, the oxidized form of glutathione) in the hippocampus. Two days after ischemia, treatment with PEP-1-PGAM5 significantly alleviated the ischemia-induced reduction in glutathione peroxidase activity and further increased superoxide dismutase activity in the hippocampus. The neuroprotective effects of PEP-1-PGAM5 are partially mediated by a reduction in oxidative stress, such as the formation of reactive oxygen species, and increases in the activity of antioxidants such as glutathione peroxidase and superoxide dismutase.


Subject(s)
Neuroprotective Agents , Animals , Antioxidants/pharmacology , Gerbillinae/metabolism , Glutathione/metabolism , Glutathione Peroxidase , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Ischemia/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress , Phosphoglycerate Mutase/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
17.
Acta Pharm Sin B ; 12(2): 558-580, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35256934

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive human cancer with increasing incidence worldwide. Multiple efforts have been made to explore pharmaceutical therapies to treat HCC, such as targeted tyrosine kinase inhibitors, immune based therapies and combination of chemotherapy. However, limitations exist in current strategies including chemoresistance for instance. Tumor initiation and progression is driven by reprogramming of metabolism, in particular during HCC development. Recently, metabolic associated fatty liver disease (MAFLD), a reappraisal of new nomenclature for non-alcoholic fatty liver disease (NAFLD), indicates growing appreciation of metabolism in the pathogenesis of liver disease, including HCC, thereby suggesting new strategies by targeting abnormal metabolism for HCC treatment. In this review, we introduce directions by highlighting the metabolic targets in glucose, fatty acid, amino acid and glutamine metabolism, which are suitable for HCC pharmaceutical intervention. We also summarize and discuss current pharmaceutical agents and studies targeting deregulated metabolism during HCC treatment. Furthermore, opportunities and challenges in the discovery and development of HCC therapy targeting metabolism are discussed.

18.
Am J Reprod Immunol ; 87(6): e13539, 2022 06.
Article in English | MEDLINE | ID: mdl-35304783

ABSTRACT

OBJECTIVES: Placentae from patients with preeclampsia have increased susceptibility to necroptosis and phosphoglycerate mutase 5 (PGAM5) plays a role in many necrosis pathways. We determined whether PGAM5 promotes necroptosis of trophoblast cells and the underlying mechanisms in this study. METHODS: The injury model was established by treating JEG3 cells with hypoxia for 24 h. The functional measurements were assessed by the cell counting kit-8, propidium iodide (PI)/Annexin V staining, JC-1 staining and firefly luciferase ATP assay. The expression of proteins in human placentae and JEG3 cells was measured Western blot. PGAM5 was knocked down to study its role in hypoxia-induced necroptosis. RESULTS: The placentae from patients with preeclampsia showed up-regulation of PGAM5 and decreased levels of p-Drp1-S637, accompanied by increased necroptosis-relevant proteins expression. The expression of PGAM5 in JEG3 cells was up-regulated under hypoxia, which promoted dephosphorylation of Drp1 at Serine 637 residue, mitochondrial dysfunction (elevated ROS level and reduced mitochondrial membrane potential and ATP content) and cellular necroptosis (increased PI+ /Annexin V+ cells and decreased cell viability), accompanied by increased expression of necroptosis-relevant proteins; knockdown of PGAM5 attenuated these phenomena. CONCLUSIONS: Our results indicate that PGAM5 can promote necroptosis in trophoblast cells through, at least in part, activation of Drp1. It may be used as a new therapeutic target to prevent trophoblast dysfunction in preeclampsia.


Subject(s)
Necroptosis , Pre-Eclampsia , Adenosine Triphosphate , Annexin A5 , Cell Line, Tumor , Dynamins/metabolism , Female , Humans , Hypoxia , Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism , Phosphoprotein Phosphatases/metabolism , Pregnancy , Trophoblasts/metabolism
19.
J Biol Chem ; 298(3): 101713, 2022 03.
Article in English | MEDLINE | ID: mdl-35150741

ABSTRACT

The glycolytic enzyme phosphoglycerate mutase (PGM) is of utmost importance for overall cellular metabolism and has emerged as a novel therapeutic target in cancer cells. This enzyme is also conserved in the rapidly proliferating malarial parasite Plasmodium falciparum, which have a similar metabolic framework as cancer cells and rely on glycolysis as the sole energy-yielding process during intraerythrocytic development. There is no redundancy among the annotated PGM enzymes in Plasmodium, and PfPGM1 is absolutely required for the parasite survival as evidenced by conditional knockdown in our study. A detailed comparison of PfPGM1 with its counterparts followed by in-depth structure-function analysis revealed unique attributes of this parasitic protein. Here, we report for the first time the importance of oligomerization for the optimal functioning of the enzyme in vivo, as earlier studies in eukaryotes only focused on the effects in vitro. We show that single point mutation of the amino acid residue W68 led to complete loss of tetramerization and diminished catalytic activity in vitro. Additionally, ectopic expression of the WT PfPGM1 protein enhanced parasite growth, whereas the monomeric form of PfPGM1 failed to provide growth advantage. Furthermore, mutation of the evolutionarily conserved residue K100 led to a drastic reduction in enzymatic activity. The indispensable nature of this parasite enzyme highlights the potential of PfPGM1 as a therapeutic target against malaria, and targeting the interfacial residues critical for oligomerization can serve as a focal point for promising drug development strategies that may not be restricted to malaria only.


Subject(s)
Phosphoglycerate Mutase , Plasmodium falciparum , Humans , Malaria/parasitology , Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism , Plasmodium falciparum/enzymology
20.
Front Cell Dev Biol ; 10: 1052363, 2022.
Article in English | MEDLINE | ID: mdl-36589741

ABSTRACT

Phosphoglycerate mutase 2 (PGAM2) is a critical glycolytic enzyme that is highly expressed in skeletal muscle. In humans, naturally occurring mutations in Phosphoglycerate mutase 2 have been etiologically linked to glycogen storage disease X (GSDX). Phosphoglycerate mutase 2 activity is regulated by several posttranslational modifications such as ubiquitination and acetylation. Here, we report that Phosphoglycerate mutase 2 activity is regulated by sumoylation-a covalent conjugation involved in a wide spectrum of cellular events. We found that Phosphoglycerate mutase 2 contains two primary SUMO acceptor sites, lysine (K)49 and K176, and that the mutation of either K to arginine (R) abolished Phosphoglycerate mutase 2 sumoylation. Given that K176 is more highly evolutionarily conserved across paralogs and orthologs than K49 is, we used the CRISPR-mediated homologous recombination technique in myogenic C2C12 cells to generate homozygous K176R knock-in cells (PGAM2K176R/K176R). Compared with wild-type (WT) C2C12 cells, PGAM2K176R/K176R C2C12 cells exhibited impaired myogenic differentiation, as indicated by decreased differentiation and fusion indexes. Furthermore, the results of glycolytic and mitochondrial stress assays with the XF96 Extracellular Flux analyzer revealed a reduced proton efflux rate (PER), glycolytic PER (glycoPER), extracellular acidification rate (ECAR), and oxygen consumption rate (OCR) in PGAM2K176R/K176R C2C12 cells, both at baseline and in response to stress. Impaired mitochondrial function was also observed in PGAM2K176R/K176R P19 cells, a carcinoma cell line. These findings indicate that the PGAM2-K176R mutation impaired glycolysis and mitochondrial function. Gene ontology term analysis of RNA sequencing data further revealed that several downregulated genes in PGAM2K176R/K176R C2C12 cells were associated with muscle differentiation/development/contraction programs. Finally, PGAM2 with either of two naturally occurring missense mutations linked to GSDX, E89A (conversion of glutamic acid 89 to alanine) or R90W (conversion of arginine 90 to tryptophan), exhibited reduced Phosphoglycerate mutase 2 sumoylation. Thus, sumoylation is an important mechanism that mediates Phosphoglycerate mutase 2 activity and is potentially implicated in Phosphoglycerate mutase 2 mutation-linked disease in humans.

SELECTION OF CITATIONS
SEARCH DETAIL
...