Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.712
Filter
1.
Sci Rep ; 14(1): 21192, 2024 09 11.
Article in English | MEDLINE | ID: mdl-39261563

ABSTRACT

A key player in energy metabolism is phosphofructokinase-1 (PFK1) whose activity and behavior strongly influence glycolysis and thus have implications in many areas. In this research, PFK1 assays were performed to convert F6P and ATP into F-1,6-P and ADP for varied pH and ATP concentrations. PFK1 activity was assessed by evaluating F-1,6-P generation velocity in two ways: (1) directly calculating the time slope from the first two or more datapoints of measured product concentration (the initial-velocity method), and (2) by fitting all the datapoints with a differential equation explicitly representing the effects of ATP and pH (the modeling method). Similar general trends of inhibition were shown by both methods, but the former gives only a qualitative picture while the modeling method yields the degree of inhibition because the model can separate the two simultaneous roles of ATP as both a substrate of reaction and an inhibitor of PFK1. Analysis based on the model suggests that the ATP affinity is much greater to the PFK1 catalytic site than to the inhibitory site, but the inhibited ATP-PFK1-ATP complex is much slower than the uninhibited PFK1-ATP complex in product generation, leading to reduced overall reaction velocity when ATP concentration increases. The initial-velocity method is simple and useful for general observation of enzyme activity while the modeling method has advantages in quantifying the inhibition effects and providing insights into the process.


Subject(s)
Adenosine Triphosphate , Phosphofructokinase-1 , Adenosine Triphosphate/metabolism , Phosphofructokinase-1/metabolism , Hydrogen-Ion Concentration , Kinetics , Fructosephosphates/metabolism , Adenosine Diphosphate/metabolism , Glycolysis
2.
J Biol Chem ; 300(9): 107648, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39121998

ABSTRACT

Most cancer cells exhibit high glycolysis rates under conditions of abundant oxygen. Maintaining a stable glycolytic rate is critical for cancer cell growth as it ensures sufficient conversion of glucose carbons to energy, biosynthesis, and redox balance. Here we deciphered the interaction between PKM2 and the thermodynamic properties of the glycolytic pathway. Knocking down or knocking out PKM2 induced a thermodynamic equilibration in the glycolytic pathway, characterized by the reciprocal changes of the Gibbs free energy (ΔG) of the reactions catalyzed by PFK1 and PK, leading to a less exergonic PFK1-catalyzed reaction and a more exergonic PK-catalyzed reaction. The changes in the ΔGs of the two reactions cause the accumulation of intermediates, including the substrate PEP (the substrate of PK), in the segment between PFK1 and PK. The increased concentration of PEP in turn increased PK activity in the glycolytic pathway. Thus, the interaction between PKM2 and the thermodynamic properties of the glycolytic pathway maintains the reciprocal relationship between PK concentration and its substrate PEP concentration, by which, PK activity in the glycolytic pathway can be stabilized and effectively counteracts the effect of PKM2 KD or KO on glycolytic rate. In line with our previous reports, this study further validates the roles of the thermodynamics of the glycolytic pathway in stabilizing glycolysis in cancer cells. Deciphering the interaction between glycolytic enzymes and the thermodynamics of the glycolytic pathway will promote a better understanding of the flux control of glycolysis in cancer cells.


Subject(s)
Carrier Proteins , Glycolysis , Membrane Proteins , Thermodynamics , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Line, Tumor , Phosphofructokinase-1/metabolism , Phosphofructokinase-1/genetics , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics
3.
Nat Commun ; 15(1): 7323, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39183237

ABSTRACT

Phosphofructokinase-1 (PFK1) catalyzes the rate-limiting step of glycolysis, committing glucose to conversion into cellular energy. PFK1 is highly regulated to respond to the changing energy needs of the cell. In bacteria, the structural basis of PFK1 regulation is a textbook example of allostery; molecular signals of low and high cellular energy promote transition between an active R-state and inactive T-state conformation, respectively. Little is known, however, about the structural basis for regulation of eukaryotic PFK1. Here, we determine structures of the human liver isoform of PFK1 (PFKL) in the R- and T-state by cryoEM, providing insight into eukaryotic PFK1 allosteric regulatory mechanisms. The T-state structure reveals conformational differences between the bacterial and eukaryotic enzyme, the mechanisms of allosteric inhibition by ATP binding at multiple sites, and an autoinhibitory role of the C-terminus in stabilizing the T-state. We also determine structures of PFKL filaments that define the mechanism of higher-order assembly and demonstrate that these structures are necessary for higher-order assembly of PFKL in cells.


Subject(s)
Adenosine Triphosphate , Phosphofructokinase-1 , Humans , Adenosine Triphosphate/metabolism , Allosteric Regulation , Cryoelectron Microscopy , Glycolysis , Liver/enzymology , Liver/metabolism , Models, Molecular , Phosphofructokinase-1/metabolism , Phosphofructokinase-1/chemistry , Phosphofructokinase-1/genetics , Protein Conformation
4.
Biochem Biophys Res Commun ; 730: 150366, 2024 10 20.
Article in English | MEDLINE | ID: mdl-38991254

ABSTRACT

Laryngeal squamous cell carcinoma (LSCC) with a high incidence and mortality rate, has a serious impact worldwide. Phosphofructokinase-1 liver type (PFKL) is a major enzyme in glycolysis progress, but its role in modulating tumorigenesis and cisplatin (DDP) chemosensitivity of LSCC was still unclear. The mRNA and protein levels of PFKL were detected by qRT-PCR and immunohistochemical assay. Cell Counting Kit-8 assay and flow cytometry were carried out to observe cell viability, as well as apoptosis and mitochondrial reactive oxygen species (mito-ROS). Extracellular acidification rate and lactate content were measured using extracellular flux analysis and lactate assay kit. Immunofluorescent staining was used to evaluate the expression of γ-H2AX foci. DNA damage was detected via single-cell gel electrophoresis. Western blotting was introduced to evaluate the protein level of PFKL, LDHA, γ-H2AX, cleaved PARP, H3K27ac, and H3K9ac. Mice xenograft model of LSCC was built for in vivo validation. The PFKL expression was significantly increased in LSCC and associated with poor survival of LSCC patients. Knockdown of PFKL in LSCC cells significantly inhibited cell viability, ECAR, lactate content, and LDHA expression, but promoted mito-ROS level. Furthermore, knockdown of PFKL enhanced response of LSCC cells to DDP by increasing DDP-induced apoptosis, promoting DDP-induced mito-ROS level, γ-H2AX foci, tail DNA, and the expression of γ-H2AX and cleaved PARP. However, the overexpression of PFKL in LSCC cells had opposite experimental results. Nude mice tumor formation experiment proved that downregulation of PFKL significantly enhanced response of cells to DDP, demonstrated by reduced tumor volume, weight and increased TUNEL-positive cells. Suppression of CBP/EP300-mediated PFKL transcription inhibited cell viability and glycolysis and promoted mito-ROS in LSCC. PFKL promotes cell viability and DNA damage repair in DDP-treated LSCC through regulation of glycolysis pathway.


Subject(s)
Antineoplastic Agents , Cell Survival , Cisplatin , Glycolysis , Laryngeal Neoplasms , Mice, Nude , Cisplatin/pharmacology , Glycolysis/drug effects , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/pathology , Laryngeal Neoplasms/genetics , Animals , Cell Survival/drug effects , Mice , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Phosphofructokinase-1/metabolism , Phosphofructokinase-1/genetics , Drug Resistance, Neoplasm/drug effects , Xenograft Model Antitumor Assays , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/genetics , Mice, Inbred BALB C , DNA Damage/drug effects
5.
Nat Commun ; 15(1): 6438, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085210

ABSTRACT

Innate immune responses are linked to key metabolic pathways, yet the proximal signaling events that connect these systems remain poorly understood. Here we show that phosphofructokinase 1, liver type (PFKL), a rate-limiting enzyme of glycolysis, is phosphorylated at Ser775 in macrophages following several innate stimuli. This phosphorylation increases the catalytic activity of PFKL, as shown by biochemical assays and glycolysis monitoring in cells expressing phosphorylation-defective PFKL variants. Using a genetic mouse model in which PFKL Ser775 phosphorylation cannot take place, we observe that upon activation, glycolysis in macrophages is lower than in the same cell population of wild-type animals. Consistent with their higher glycolytic activity, wild-type cells have higher levels of HIF1α and IL-1ß than PfklS775A/S775A after LPS treatment. In an in vivo inflammation model, PfklS775A/S775A mice show reduced levels of MCP-1 and IL-1ß. Our study thus identifies a molecular link between innate immune activation and early induction of glycolysis.


Subject(s)
Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Immunity, Innate , Interleukin-1beta , Macrophages , Animals , Macrophages/metabolism , Macrophages/immunology , Mice , Phosphorylation , Interleukin-1beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/genetics , Phosphofructokinase-1/metabolism , Phosphofructokinase-1/genetics , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Humans , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Inflammation/metabolism , Male , Metabolic Reprogramming
6.
BMC Urol ; 24(1): 59, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38481182

ABSTRACT

BACKGROUND: Bladder cancer (BC) is one of the most common malignancies of the genitourinary system. Phosphofructokinase 1 (PFK-1) is one of member of PFK, which plays an important role in reprogramming cancer metabolism, such as lactylation modification. Zinc finger E-box-binding homeobox 1 (ZEB1) has been demonstrated to be a oncogene in many cancers. Therefore, this study was performed to explore the effects of PFK-1 on the lactylation of ZEB1 in BC development. METHODS: Cell viability was measured using the CCK-8 kit. The glucose assay kit and lactate assay kit were used to detect glucose utilization and lactate production. The DNA was purified and quantified by qRT-PCR. RESULTS: In the present study, we found that ZEB1 expression levels were significantly elevated in bladder cancer cells. Impaired PFK-1 expression inhibits proliferation, migration, and invasion of BC cells and suppresses tumour growth in vivo. We subsequently found that knockdown of PFK-1 decreases glycolysis, including reduced glucose consumption, lactate production and total extracellular acidification rate (ECAR). Mechanistically, PFK-1 inhibits histone lactylation of bladder cancer cells, and thus inhibits the transcription activity of ZEB1. CONCLUSION: Our results suggest that PFK-1 can inhibit the malignant phenotype of bladder cancer cells by mediating the lactylation of ZEB1. These findings suggested PFK-1 to be a new potential target for bladder cancer therapy.


Subject(s)
Urinary Bladder Neoplasms , Humans , Cell Line, Tumor , Cell Movement , Urinary Bladder Neoplasms/pathology , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Lactates , Glucose , Cell Proliferation , Gene Expression Regulation, Neoplastic , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
7.
J R Soc Interface ; 21(211): 20230588, 2024 02.
Article in English | MEDLINE | ID: mdl-38350614

ABSTRACT

In many living organisms displaying circadian rhythms, the intake of energy often occurs in a periodic manner. Glycolysis is a prototypical biochemical reaction that exhibits a self-sustained oscillation under continuous injection of glucose. Here we study the effect of periodic injection of glucose on the glycolytic oscillation from a dynamical systems perspective. In particular, we employ Goldbeter's allosteric model of phosphofructokinase as a model system for glycolytic oscillations, and explore the effect of periodic substrate influx of varying frequencies and amplitudes by building the phase diagrams of Lyapunov exponents and oscillatory periods. When the frequency of driving is tuned around the harmonic and sub/super-harmonic conditions of the natural frequency, the system is entrained to a frequency-locked state, forming an entrainment band that broadens with an increasing amplitude of driving. On the other hand, if the amplitude is substantial, the system may transition, albeit infrequent, to a chaotic state which defies prediction of dynamical behaviour. Our study offers in-depth understandings into the controllability of glycolytic oscillation as well as explaining physical underpinnings that enable the synchronous oscillations among a dense population of cells.


Subject(s)
Circadian Rhythm , Models, Biological , Phosphofructokinase-1/metabolism , Glycolysis , Glucose
8.
Cell Death Dis ; 14(12): 816, 2023 12 12.
Article in English | MEDLINE | ID: mdl-38086793

ABSTRACT

Metabolic reprogramming to glycolysis is closely associated with the development of chronic kidney disease (CKD). Although it has been reported that phosphofructokinase 1 (PFK) is a rate-limiting enzyme in glycolysis, the role of the platelet isoform of PFK (PFKP) in kidney fibrosis initiation and progression is as yet poorly understood. Here, we investigated whether PFKP could mediate the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells (PTECs). We induced PFKP overexpression or knockdown in renal tubules via an adeno-associated virus (AAV) vector in the kidneys of mice following unilateral ureteral occlusion. Our results show that the dilated tubules, the area of interstitial fibrosis, and renal glycolysis were promoted by proximal tubule-specific overexpression of PFKP, and repressed by knockdown of PFKP. Furthermore, knockdown of PFKP expression restrained, while PFKP overexpression promoted TGF-ß1-induced glycolysis in the human PTECs line. Mechanistically, Chip-qPCR revealed that TGF-ß1 recruited the small mothers against decapentaplegic (SMAD) family member 3-SP1 complex to the PFKP promoter to enhance its expression. Treatment of mice with isorhamnetin notably ameliorated PTEC-elevated glycolysis and kidney fibrosis. Hence, our results suggest that PFKP mediates the progression of kidney interstitial fibrosis by regulating glycolysis in PTECs.


Subject(s)
Renal Insufficiency, Chronic , Ureteral Obstruction , Animals , Humans , Mice , Epithelial Cells/metabolism , Fibrosis , Glycolysis , Kidney/pathology , Phosphofructokinase-1/metabolism , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/pathology
9.
Cell Rep ; 42(11): 113426, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37967006

ABSTRACT

Aerobic glycolysis is critical for cancer progression and can be exploited in cancer therapy. Here, we report that the human carboxymethylenebutenolidase homolog (carboxymethylenebutenolidase-like [CMBL]) acts as a tumor suppressor by reprogramming glycolysis in colorectal cancer (CRC). The anti-cancer action of CMBL is mediated through its interactions with the E3 ubiquitin ligase TRIM25 and the glycolytic enzyme phosphofructokinase-1 platelet type (PFKP). Ectopic CMBL enhances TRIM25 binding to PFKP, leading to the ubiquitination and proteasomal degradation of PFKP. Interestingly, CMBL is transcriptionally activated by p53 in response to genotoxic stress, and p53 activation represses glycolysis by promoting PFKP degradation. Remarkably, CMBL deficiency, which impairs p53's ability to inhibit glycolysis, makes tumors more sensitive to a combination therapy involving the glycolysis inhibitor 2-deoxyglucose. Taken together, our study demonstrates that CMBL suppresses CRC growth by inhibiting glycolysis and suggests a potential combination strategy for the treatment of CMBL-deficient CRC.


Subject(s)
Neoplasms , Phosphofructokinase-1, Type C , Humans , Cell Line, Tumor , Glucose/metabolism , Glycolysis , Phosphofructokinase-1/metabolism , Phosphofructokinase-1, Type C/metabolism , Phosphofructokinases/metabolism , Tumor Suppressor Protein p53/metabolism
10.
Am J Physiol Cell Physiol ; 325(5): C1354-C1368, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37781737

ABSTRACT

Glomerular angiogenesis is a characteristic feature of diabetic nephropathy (DN). Enhanced glycolysis plays a crucial role in angiogenesis. The present study was designed to investigate the role of glycolysis in glomerular endothelial cells (GECs) in a mouse model of DN. Mouse renal cortex and isolated glomerular cells were collected for single-cell and RNA sequencing. Cultured GECs were exposed to high glucose in the presence (proangiogenic) and absence of a vascular sprouting regimen. MicroRNA-590-3p was delivered by lipofectamine in vivo and in vitro. In the present study, a subgroup of GECs with proangiogenic features was identified in diabetic kidneys by using sequencing analyses. In cultured proangiogenic GECs, high glucose increased glycolysis and phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) protein expression, which were inhibited by overexpressing miRNA-590-3p. Mimics of miRNA-590-3p also increased receptor for sphingosine 1-phosphate (S1pR1) expression, an angiogenesis regulator, in proangiogenic GECs challenged with high glucose. Inhibition of PFKFB3 by pharmacological and genetic approaches upregulated S1pR1 protein in vitro. Mimics of miRNA-590-3p significantly reduced migration and angiogenic potential in proangiogenic GECs challenged with high glucose. Ten-week-old type 2 diabetic mice had elevated urinary albumin levels, reduced renal cortex miRNA-590-3p expression, and disarrangement of glomerular endothelial cell fenestration. Overexpressing miRNA-590-3p via perirenal adipose tissue injection restored endothelial cell fenestration and reduced urinary albumin levels in diabetic mice. Therefore, the present study identifies a subgroup of GECs with proangiogenic features in mice with DN. Local administration of miRNA-590-3p mimics reduces glycolytic rate and upregulates S1pR1 protein expression in proangiogenic GECs. The protective effects of miRNA-590-3p provide therapeutic potential in DN treatment.NEW & NOTEWORTHY Proangiogenetic glomerular endothelial cells (GECs) are activated in diabetic nephropathy. High glucose upregulates glycolytic enzyme phosphofructokinase/fructose bisphosphatase 3 (PFKFB3) in proangiogenetic cells. PFKFB3 protects the glomerular filtration barrier by targeting endothelial S1pR1. MiRNA-590-3p restores endothelial cell function and mitigates diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , MicroRNAs , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Fructose-Bisphosphatase/metabolism , Fructose-Bisphosphatase/pharmacology , Phosphofructokinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Phosphofructokinase-1/metabolism , Glucose/metabolism , MicroRNAs/metabolism , Albumins/metabolism , Albumins/pharmacology , Glycolysis
11.
Adv Biol Regul ; 90: 100987, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806136

ABSTRACT

Phosphofructokinase is the central enzyme in glycolysis and constitutes a highly regulated step. The liver isoform (PFKL) compartmentalizes during activation and inhibition in vitro and in vivo, respectively. Compartmentalized PFKL is hypothesized to modulate metabolic flux consistent with its central role as the rate limiting step in glycolysis. PFKL tetramers self-assemble at two interfaces in the monomer (interface 1 and 2), yet how these interfaces contribute to PFKL compartmentalization and drive protein interactions remains unclear. Here, we used site-specific incorporation of noncanonical photocrosslinking amino acids to identify PFKL interactors at interface 1, 2, and the active site. Tandem mass tag-based quantitative interactomics reveals interface 2 as a hotspot for PFKL interactions, particularly with cytoskeletal, glycolytic, and carbohydrate derivative metabolic proteins. Furthermore, PFKL compartmentalization into puncta was observed in human cells using citrate inhibition. Puncta formation attenuated crosslinked protein-protein interactions with the cytoskeleton at interface 2. This result suggests that PFKL compartmentalization sequesters interface 2, but not interface 1, and may modulate associated protein assemblies with the cytoskeleton.


Subject(s)
Phosphofructokinase-1 , Phosphofructokinases , Humans , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Liver/metabolism , Citrates , Citric Acid
12.
BMB Rep ; 56(11): 618-623, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37605615

ABSTRACT

Most cancer cells utilize glucose at a high rate to produce energyand precursors for the biosynthesis of macromolecules such as lipids, proteins, and nucleic acids. This phenomenon is called the Warburg effect or aerobic glycolysis- this distinct characteristic is an attractive target for developing anticancer drugs. Here, we found that Phosphofructokinase-1 (PFK-1) is a substrate of the Protein Phosphatase 4 catalytic subunit (PP4C)/PP4 regulatory subunit 1 (PP4R1) complex by using immunoprecipitation and in vitro assay. While manipulation of PP4C/PP4R1 does not have a critical impact on PFK-1 expression, the absence of the PP4C/PP4R1 complex increases PFK-1 activity. Although PP4C depletion or overexpression does not cause a dramatic change in the overall glycolytic rate, PP4R1 depletion induces a considerable increase in both basal and compensatory glycolytic rates, as well as the oxygen consumption rate, indicating oxidative phosphorylation. Collectively, the PP4C/PP4R1 complex regulates PFK-1 activity by reversing its phosphorylation and is a promising candidate for treating glycolytic disorders and cancers. Targeting PP4R1 could be a more efficient and safer strategy to avoid pleiotropic effects than targeting PP4C directly. [BMB Reports 2023; 56(11): 618-623].


Subject(s)
Phosphofructokinase-1 , Phosphoprotein Phosphatases , Phosphorylation , Phosphoprotein Phosphatases/metabolism , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Carbohydrate Metabolism , Glycolysis
13.
Arch Biochem Biophys ; 743: 109676, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37380119

ABSTRACT

The phosphofructokinase (Pfk) reaction represents one of the key regulatory points in glycolysis. While most organisms encode for Pfks that use ATP as phosphoryl donor, some organisms also encode for PPi-dependent Pfks. Despite this central role, the biochemical characteristics as well as the physiological role of both Pfks is often not known. Clostridium thermocellum is an example of a microorganism that encodes for both Pfks, however, only PPi-Pfk activity has been detected in cell-free extracts and little is known about the regulation and function of both enzymes. In this study, the ATP- and PPi-Pfk of C. thermocellum were purified and biochemically characterized. No allosteric regulators were found for PPi-Pfk amongst common effectors. With fructose-6-P, PPi, fructose-1,6-bisP, and Pi PPi-Pfk showed high specificity (KM < 0.62 mM) and maximum activity (Vmax > 156 U mg-1). In contrast, ATP-Pfk showed much lower affinity (K0.5 of 9.26 mM) and maximum activity (14.5 U mg-1) with fructose-6-P. In addition to ATP, also GTP, UTP and ITP could be used as phosphoryl donors. The catalytic efficiency with GTP was 7-fold higher than with ATP, suggesting that GTP is the preferred substrate. The enzyme was activated by NH4+, and pronounced inhibition was observed with GDP, FBP, PEP, and especially with PPi (Ki of 0.007 mM). Characterization of purified ATP-Pfks originating from eleven different bacteria, encoding for only ATP-Pfk or for both ATP- and PPi-Pfk, identified that PPi inhibition of ATP-Pfks could be a common phenomenon for organisms with a PPi-dependent glycolysis.


Subject(s)
Clostridium thermocellum , Phosphofructokinases , Phosphofructokinases/metabolism , Clostridium thermocellum/metabolism , Diphosphates , Amino Acid Sequence , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Bacteria/metabolism , Adenosine Triphosphate , Guanosine Triphosphate , Kinetics
14.
Cancer Sci ; 114(4): 1663-1671, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36601784

ABSTRACT

To meet cellular bioenergetic and biosynthetic demands, cancer cells remodel their metabolism to increase glycolytic flux, a phenomenon known as the Warburg effect and believed to contribute to cancer malignancy. Among glycolytic enzymes, phosphofructokinase-1 (PFK1) has been shown to act as a rate-limiting enzyme and to facilitate the Warburg effect in cancer cells. In this study, however, we found that decreased PFK1 activity did not affect cell survival or proliferation in cancer cells. This raised a question regarding the importance of PFK1 in malignancy. To gain insights into the role of PFK1 in cancer metabolism and the possibility of adopting it as a novel anticancer therapeutic target, we screened for genes that caused lethality when they were knocked down in the presence of tryptolinamide (TLAM), a PFK1 inhibitor. The screen revealed a synthetic chemical-genetic interaction between genes encoding subunits of ATP synthase (complex V) and TLAM. Indeed, after TLAM treatment, the sensitivity of HeLa cells to oligomycin A (OMA), an ATP synthase inhibitor, was 13,000 times higher than that of untreated cells. Furthermore, this sensitivity potentiation by TLAM treatment was recapitulated by genetic mutations of PFK1. By contrast, TLAM did not potentiate the sensitivity of normal fibroblast cell lines to OMA, possibly due to their reduced energy demands compared to cancer cells. We also showed that the PFK1-mediated glycolytic pathway can act as an energy reservoir. Selective potentiation of the efficacy of ATP synthase inhibitors by PFK1 inhibition may serve as a foundation for novel anticancer therapeutic strategies.


Subject(s)
Adenosine Triphosphatases , Early Detection of Cancer , Neoplasms , Phosphofructokinase-1 , Humans , Glycolysis/genetics , HeLa Cells , Neoplasms/genetics , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , RNA Interference , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism
15.
Pest Manag Sci ; 79(5): 1684-1691, 2023 May.
Article in English | MEDLINE | ID: mdl-36602054

ABSTRACT

BACKGROUND: Myzus persicae (Hemiptera: Aphididae) is one of the most notorious pests of many crops worldwide. Most Cry toxins produced by Bacillus thuringiensis show very low toxicity to M. persicae; however, a study showed that Cry41-related toxin had moderate toxic activity against M. persicae. In our previous work, potential Cry41-related toxin-binding proteins in M. persicae were identified, including cathepsin B, calcium-transporting ATPase, and Buchnera-derived ATP-dependent 6-phosphofructokinase (PFKA). Buchnera is an endosymbiont present in almost all aphids and it provides necessary nutrients for aphid growth. This study investigated the role of Buchnera-derived PFKA in Cry41-related toxicity against M. persicae. RESULTS: In this study, recombinant PFKA was expressed and purified, and in vitro assays revealed that PFKA bound to Cry41-related toxin, and Cry41-related toxin at 25 µg ml-1 significantly inhibited the activity of PFKA. In addition, when M. persicae was treated with 30 µg ml-1 of Cry41-related toxin for 24 h, the expression of dnak, a single-copy gene in Buchnera, was significantly decreased, indicating a decrease in the number of Buchnera. CONCLUSION: Our results suggest that Cry41-related toxin interacts with Buchnera-derived PFKA to inhibit its enzymatic activity and likely impair cell viability, resulting in a decrease in the number of Buchnera, and finally leading to M. persicae death. These findings open up new perspectives in our understanding of the mode of action of Cry toxins and are useful in helping improve Cry toxicity for aphid control. © 2023 Society of Chemical Industry.


Subject(s)
Aphids , Buchnera , Animals , Phosphofructokinases/metabolism , Phosphofructokinase-1/metabolism , Adenosine Triphosphate/metabolism
16.
Anticancer Res ; 43(1): 75-84, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36585185

ABSTRACT

BACKGROUND/AIM: Phosphofructokinase 1 platelet isoform (PFKP) catalyzes a rate-limiting reaction in glycolysis. It is highly expressed in several tumors, including breast cancer (BC). It can regulate tumor progression through metabolic reprogramming and gene transcription. In addition, overexpression of vascular endothelial growth factor (VEGF) is commonly observed in BC, which is associated with poor prognosis. However, whether PFKP regulates VEGF expression in BC remains unknown. Thus, the aim of this study was to investigate whether PFKP could regulate VEGF expression in BC. MATERIALS AND METHODS: We designed an in vitro study to investigate the role of PFKP in VEGF expression and angiogenesis using several experiments, including shRNA-mediated PFKP knock-down, RNAi-resistant PFKP restoration, qPCR, immunoblotting, luciferase reporter assay and tube formation assay. The clinical relationship between PFKP and VEGF was analyzed using The Cancer Genome Atlas (TCGA) database. RESULTS: PFKP expression was associated with VEGF expression in BC patients from the TCGA database. Importantly, PFKP played an essential role in the EGFR activation-induced VEGF expression in BC cells. Mechanistically, EGFR-phosphorylated PFKP Y64 played a critical role in AKT-mediated transcriptional expression of HIF-1α and subsequent VEGF transcription. Hence, PFKP expression played a role in human umbilical vein endothelial cells (HUVECs) tube formation by regulating VEGF expression in BC cells. CONCLUSION: These findings highlight a novel mechanism underlying the non-metabolic function of PFKP in VEGF expression in BC and provide a therapeutic potential of targeting PFKP in BC patients.


Subject(s)
Breast Neoplasms , Hypoxia-Inducible Factor 1, alpha Subunit , Phosphofructokinase-1 , Vascular Endothelial Growth Factor A , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelial Cells/metabolism , ErbB Receptors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Protein Isoforms/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Death Dis ; 13(11): 1002, 2022 11 26.
Article in English | MEDLINE | ID: mdl-36435833

ABSTRACT

Glioblastoma (GBM) is a highly vascular malignant brain tumor that overexpresses vascular endothelial growth factor (VEGF) and phosphofructokinase 1 platelet isoform (PFKP), which catalyzes a rate-limiting reaction in glycolysis. However, whether PFKP and VEGF are reciprocally regulated during GBM tumor growth remains unknown. Here, we show that PFKP can promote EGFR activation-induced VEGF expression in HIF-1α-dependent and -independent manners in GBM cells. Importantly, we demonstrate that EGFR-phosphorylated PFKP Y64 has critical roles in both AKT/SP1-mediated transcriptional expression of HIF-1α and in the AKT-mediated ß-catenin S552 phosphorylation, to fully enhance VEGF transcription, subsequently promoting blood vessel formation and brain tumor growth. Levels of PFKP Y64 phosphorylation in human GBM specimens are positively correlated with HIF-1α expression, ß-catenin S552 phosphorylation, and VEGF expression. Conversely, VEGF upregulates PFKP expression in a PFKP S386 phosphorylation-dependent manner, leading to increased PFK enzyme activity, aerobic glycolysis, and proliferation in GBM cells. These findings highlight a novel mechanism underlying the mutual regulation that occurs between PFKP and VEGF for promoting GBM tumor growth and also suggest that targeting the PFKP/VEGF regulatory loop might show therapeutic potential for treating GBM patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Phosphorylation , beta Catenin/genetics , beta Catenin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphofructokinase-1/metabolism , Vascular Endothelial Growth Factors/metabolism , Brain Neoplasms/genetics , Protein Isoforms/metabolism , ErbB Receptors/metabolism
18.
Appl Environ Microbiol ; 88(22): e0125822, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36286488

ABSTRACT

Glycolysis is an ancient, widespread, and highly conserved metabolic pathway that converts glucose into pyruvate. In the canonical pathway, the phosphofructokinase (PFK) reaction plays an important role in controlling flux through the pathway. Clostridium thermocellum has an atypical glycolysis and uses pyrophosphate (PPi) instead of ATP as the phosphate donor for the PFK reaction. The reduced thermodynamic driving force of the PPi-PFK reaction shifts the entire pathway closer to thermodynamic equilibrium, which has been predicted to limit product titers. Here, we replace the PPi-PFK reaction with an ATP-PFK reaction. We demonstrate that the local changes are consistent with thermodynamic predictions: the ratio of fructose 1,6-bisphosphate to fructose-6-phosphate increases, and the reverse flux through the reaction (determined by 13C labeling) decreases. The final titer and distribution of fermentation products, however, do not change, demonstrating that the thermodynamic constraints of the PPi-PFK reaction are not the sole factor limiting product titer. IMPORTANCE The ability to control the distribution of thermodynamic driving force throughout a metabolic pathway is likely to be an important tool for metabolic engineering. The phosphofructokinase reaction is a key enzyme in Embden-Mayerhof-Parnas glycolysis and therefore improving the thermodynamic driving force of this reaction in C. thermocellum is believed to enable higher product titers. Here, we demonstrate switching from pyrophosphate to ATP does in fact increases the thermodynamic driving force of the phosphofructokinase reaction in vivo. This study also identifies and overcomes a physiological hurdle toward expressing an ATP-dependent phosphofructokinase in an organism that utilizes an atypical glycolytic pathway. As such, the method described here to enable expression of ATP-dependent phosphofructokinase in an organism with an atypical glycolytic pathway will be informative toward engineering the glycolytic pathways of other industrial organism candidates with atypical glycolytic pathways.


Subject(s)
Clostridium thermocellum , Clostridium thermocellum/metabolism , Diphosphates/metabolism , Phosphofructokinases/genetics , Phosphofructokinase-1/genetics , Phosphofructokinase-1/metabolism , Glycolysis , Thermodynamics , Adenosine Triphosphate/metabolism
19.
Biomed Pharmacother ; 155: 113660, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36095960

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), often associated with obesity, is becoming one of the most common liver diseases worldwide. It is estimated to affect one billion individuals and may be present in approximately 25% of the population globally. NAFLD is viewed as a hepatic manifestation of metabolic syndrome, with humans and animal models presenting dyslipidemia, hypertension, and diabetes. The gut-liver axis has been considered the main pathogenesis branch for NAFLD development. Considering that foods or beverages could modulate the gastrointestinal tract, immune system, energy homeostasis regulation, and even the gut-liver axis, we conducted an exploratory study to analyze the effects of kombucha probiotic on hepatic steatosis, glucose tolerance, and hepatic enzymes involved in carbohydrate and fat metabolism using a pre-clinical model. The diet-induced obese mice presented glucose intolerance, hyperinsulinemia, hepatic steatosis, increased collagen fiber deposition in liver vascular spaces, and upregulated TNF-alpha and SREBP-1 gene expression. Mice receiving the kombucha supplement displayed improved glucose tolerance, reduced hyperinsulinemia, decreased citrate synthase and phosphofructokinase-1 enzyme activities, downregulated G-protein-coupled bile acid receptor, also known as TGR5, and farnesol X receptor gene expression, and attenuated steatosis and hepatic collagen fiber deposition. The improvement in glucose tolerance was accompanied by the recovery of acute insulin-induced liver AKT serine phosphorylation. Thus, it is possible to conclude that this probiotic drink has a beneficial effect in reducing the metabolic alterations associated with diet-induced obesity. This probiotic beverage deserves an extension of studies to confirm or refute its potentially beneficial effects.


Subject(s)
Insulin Resistance , Kombucha Tea , Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Citrate (si)-Synthase/metabolism , Farnesol/metabolism , Farnesol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Liver , Obesity/complications , Obesity/drug therapy , Obesity/metabolism , Insulin/metabolism , Glucose/metabolism , Bile Acids and Salts/metabolism , Carbohydrates/pharmacology , Serine/metabolism , Serine/pharmacology , Phosphofructokinase-1/metabolism , GTP-Binding Proteins/metabolism , Collagen/metabolism , Mice, Inbred C57BL , Diet, High-Fat
20.
Int J Mol Sci ; 23(17)2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36077431

ABSTRACT

Aerobic glycolysis is an emerging hallmark of many human cancers, as cancer cells are defined as a "metabolically abnormal system". Carbohydrates are metabolically reprogrammed by its metabolizing and catabolizing enzymes in such abnormal cancer cells. Normal cells acquire their energy from oxidative phosphorylation, while cancer cells acquire their energy from oxidative glycolysis, known as the "Warburg effect". Energy-metabolic differences are easily found in the growth, invasion, immune escape and anti-tumor drug resistance of cancer cells. The glycolysis pathway is carried out in multiple enzymatic steps and yields two pyruvate molecules from one glucose (Glc) molecule by orchestral reaction of enzymes. Uncontrolled glycolysis or abnormally activated glycolysis is easily observed in the metabolism of cancer cells with enhanced levels of glycolytic proteins and enzymatic activities. In the "Warburg effect", tumor cells utilize energy supplied from lactic acid-based fermentative glycolysis operated by glycolysis-specific enzymes of hexokinase (HK), keto-HK-A, Glc-6-phosphate isomerase, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, phosphofructokinase (PFK), phosphor-Glc isomerase (PGI), fructose-bisphosphate aldolase, phosphoglycerate (PG) kinase (PGK)1, triose phosphate isomerase, PG mutase (PGAM), glyceraldehyde-3-phosphate dehydrogenase, enolase, pyruvate kinase isozyme type M2 (PKM2), pyruvate dehydrogenase (PDH), PDH kinase and lactate dehydrogenase. They are related to glycolytic flux. The key enzymes involved in glycolysis are directly linked to oncogenesis and drug resistance. Among the metabolic enzymes, PKM2, PGK1, HK, keto-HK-A and nucleoside diphosphate kinase also have protein kinase activities. Because glycolysis-generated energy is not enough, the cancer cell-favored glycolysis to produce low ATP level seems to be non-efficient for cancer growth and self-protection. Thus, the Warburg effect is still an attractive phenomenon to understand the metabolic glycolysis favored in cancer. If the basic properties of the Warburg effect, including genetic mutations and signaling shifts are considered, anti-cancer therapeutic targets can be raised. Specific therapeutics targeting metabolic enzymes in aerobic glycolysis and hypoxic microenvironments have been developed to kill tumor cells. The present review deals with the tumor-specific Warburg effect with the revisited viewpoint of recent progress.


Subject(s)
Glycolysis , Neoplasms , Hexokinase/metabolism , Humans , Neoplasms/metabolism , Phosphofructokinase-1/metabolism , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Mutase/metabolism , Pyruvates , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL