Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
1.
BMC Urol ; 24(1): 59, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38481182

RESUMO

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.


Assuntos
Neoplasias da Bexiga Urinária , Humanos , Linhagem Celular Tumoral , Movimento Celular , Neoplasias da Bexiga Urinária/patologia , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Lactatos , Glucose , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
2.
Cell Rep ; 42(10): 113246, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37831605

RESUMO

Metastasis is the leading cause of high ovarian-cancer-related mortality worldwide. Three major processes constitute the whole metastatic cascade: invasion, intravasation, and extravasation. Tumor cells often reprogram their metabolism to gain advantages in proliferation and survival. However, whether and how those metabolic alterations contribute to the invasiveness of tumor cells has yet to be fully understood. Here we performed a genome-wide CRISPR-Cas9 screening to identify genes participating in tumor cell dissemination and revealed that PTGES3 acts as an invasion suppressor in ovarian cancer. Mechanistically, PTGES3 binds to phosphofructokinase, liver type (PFKL) and generates a local source of prostaglandin E2 (PGE2) to allosterically inhibit the enzymatic activity of PFKL. Repressed PFKL leads to downgraded glycolysis and the subsequent TCA cycle for glucose metabolism. However, ovarian cancer suppresses the expression of PTGES3 and disrupts the PTGES3-PGE2-PFKL inhibitory axis, leading to hyperactivation of glucose oxidation, eventually facilitating ovarian cancer cell motility and invasiveness.


Assuntos
Dinoprostona , Neoplasias Ovarianas , Humanos , Feminino , Fosfofrutoquinases , Fosfofrutoquinase-1/genética , Fígado/metabolismo , Glucose/metabolismo , Neoplasias Ovarianas/patologia , Proliferação de Células , Linhagem Celular Tumoral , Invasividade Neoplásica
3.
BMB Rep ; 56(11): 618-623, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37605615

RESUMO

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].


Assuntos
Fosfofrutoquinase-1 , Fosfoproteínas Fosfatases , Fosforilação , Fosfoproteínas Fosfatases/metabolismo , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Metabolismo dos Carboidratos , Glicólise
4.
Biochem J ; 480(17): 1411-1427, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37622331

RESUMO

Metabolic reprogramming, including increased glucose uptake and lactic acid excretion, is a hallmark of cancer. The glycolytic 'gatekeeper' enzyme phosphofructokinase-1 (PFK1), which catalyzes the step committing glucose to breakdown, is dysregulated in cancers. While altered PFK1 activity and expression in tumors have been demonstrated, little is known about the effects of cancer-associated somatic mutations. Somatic mutations in PFK1 inform our understanding of allosteric regulation by identifying key amino acid residues involved in the regulation of enzyme activity. Here, we characterized mutations disrupting an evolutionarily conserved salt bridge between aspartic acid and arginine in human platelet (PFKP) and liver (PFKL) isoforms. Using purified recombinant proteins, we showed that disruption of the Asp-Arg pair in two PFK1 isoforms decreased enzyme activity and altered allosteric regulation. We determined the crystal structure of PFK1 to 3.6 Šresolution and used molecular dynamic simulations to understand molecular mechanisms of altered allosteric regulation. We showed that PFKP-D564N had a decreased total system energy and changes in the electrostatic surface potential of the effector site. Cells expressing PFKP-D564N demonstrated a decreased rate of glycolysis, while their ability to induce glycolytic flux under conditions of low cellular energy was enhanced compared with cells expressing wild-type PFKP. Taken together, these results suggest that mutations in Arg-Asp pair at the interface of the catalytic-regulatory domains stabilizes the t-state and presents novel mechanistic insight for therapeutic development in cancer.


Assuntos
Neoplasias , Fosfofrutoquinase-1 , Humanos , Regulação Alostérica , Eletricidade Estática , Fosfofrutoquinase-1/genética , Metabolismo dos Carboidratos , Neoplasias/genética
5.
Arch Biochem Biophys ; 743: 109676, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37380119

RESUMO

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.


Assuntos
Clostridium thermocellum , Fosfofrutoquinases , Fosfofrutoquinases/metabolismo , Clostridium thermocellum/metabolismo , Difosfatos , Sequência de Aminoácidos , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Bactérias/metabolismo , Trifosfato de Adenosina , Guanosina Trifosfato , Cinética
6.
Cancer Sci ; 114(4): 1663-1671, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36601784

RESUMO

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.


Assuntos
Adenosina Trifosfatases , Detecção Precoce de Câncer , Neoplasias , Fosfofrutoquinase-1 , Humanos , Glicólise/genética , Células HeLa , Neoplasias/genética , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Interferência de RNA , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo
7.
Anticancer Res ; 43(1): 75-84, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36585185

RESUMO

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.


Assuntos
Neoplasias da Mama , Subunidade alfa do Fator 1 Induzível por Hipóxia , Fosfofrutoquinase-1 , Fator A de Crescimento do Endotélio Vascular , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Receptores ErbB/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Isoformas de Proteínas/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Appl Environ Microbiol ; 88(22): e0125822, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36286488

RESUMO

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.


Assuntos
Clostridium thermocellum , Clostridium thermocellum/metabolismo , Difosfatos/metabolismo , Fosfofrutoquinases/genética , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Glicólise , Termodinâmica , Trifosfato de Adenosina/metabolismo
9.
J BUON ; 26(4): 1210-1218, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34564972

RESUMO

PURPOSE: Metformin has been widely used for the treatment of Type 2 Diabetes Mellitus (T2DM), hyperglycemia and polycystic ovarian syndrome. Recent studies have suggested the potential of this substance as a cancer chemopreventive agent. We evaluated the antitumoral effect of iRNA-PFK-1 and the combined therapy iRNA-PFK-1 + metformin in RKO p53-positive cells. METHODS: mRNA levels of tumor suppressor genes AMPK, APC, and c-MYC, KRAS oncogenes were measured by qRT-PCR in RKO cells treated with 25 µM metformin alone or combined with iRNA-PFK-1, to evaluate the effect of both treatments. RESULTS: At 72 h after treatment with either 25 µM metformin, 150 nM iRNA-PFK-1, or the combined treatment, the transcriptional levels of these biomarkers were decreased by ~73% (p˂0.05), ~99.9%, (p˂0.01), and ~76% (p˂0.05), respectively. CONCLUSION: These in vitro results support the potential therapeutic role of metformin and PFK-1 in the treatment of colon cancer via down-modulation of the expression of several important cancer biomarkers.


Assuntos
Biomarcadores Tumorais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Regulação para Baixo/efeitos dos fármacos , Metformina/administração & dosagem , Fosfofrutoquinase-1/administração & dosagem , Biomarcadores Tumorais/genética , Neoplasias do Colo/genética , Combinação de Medicamentos , Humanos , Fosfofrutoquinase-1/genética , RNA , Células Tumorais Cultivadas
10.
Trends Endocrinol Metab ; 32(8): 540-543, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34016523

RESUMO

We propose that fructose-1,6-bisphosphate (F-1,6-BP) promotes a feedback loop between phosphofructokinase-1 (PFK1), phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt), and PFK2/PFKFB3, which enhances aerobic glycolysis and sustains effector T (Teff) cell activation, while oxidative metabolism is concomitantly downregulated. This regulation, promoted by low citrate and mitochondrial ATP synthesis, also sustains the Warburg effect in cancer cells.


Assuntos
Frutosedifosfatos/metabolismo , Glicólise , Fosfofrutoquinase-1 , Linfócitos T , Trifosfato de Adenosina/biossíntese , Ácido Cítrico , Ativação Linfocitária , Mitocôndrias , Neoplasias , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T/metabolismo
11.
Am J Physiol Cell Physiol ; 321(1): C147-C157, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34038242

RESUMO

Many cancer cells, regardless of their tissue origin or genetic landscape, have increased expression or activity of the plasma membrane Na-H exchanger NHE1 and a higher intracellular pH (pHi) compared with untransformed cells. A current perspective that remains to be validated is that increased NHE1 activity and pHi enable a Warburg-like metabolic reprogramming of increased glycolysis and decreased mitochondrial oxidative phosphorylation. We tested this perspective and find it is not accurate for clonal pancreatic and breast cancer cells. Using the pharmacological reagent ethyl isopropyl amiloride (EIPA) to inhibit NHE1 activity and decrease pHi, we observe no change in glycolysis, as indicated by secreted lactate and intracellular pyruvate, despite confirming increased activity of the glycolytic enzyme phosphofructokinase-1 at higher pH. Also, in contrast to predictions, we find a significant decrease in oxidative phosphorylation with EIPA, as indicated by oxygen consumption rate (OCR). Decreased OCR with EIPA is not associated with changes in pathways that fuel oxidative phosphorylation or with mitochondrial membrane potential but occurs with a change in mitochondrial dynamics that includes a significant increase in elongated mitochondrial networks, suggesting increased fusion. These findings conflict with current paradigms on increased pHi inhibiting oxidative phosphorylation and increased oxidative phosphorylation being associated with mitochondrial fusion. Moreover, these findings raise questions on the suggested use of EIPA-like compounds to limit metabolic reprogramming in cancer cells.


Assuntos
Amilorida/análogos & derivados , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Dinâmica Mitocondrial/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Trocador 1 de Sódio-Hidrogênio/genética , Amilorida/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Células Clonais , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Expressão Gênica , Glicólise/genética , Humanos , Concentração de Íons de Hidrogênio , Ácido Láctico/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Ácido Pirúvico/metabolismo , Trocador 1 de Sódio-Hidrogênio/antagonistas & inibidores , Trocador 1 de Sódio-Hidrogênio/metabolismo
12.
Cell Cycle ; 20(2): 154-165, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33404290

RESUMO

Nasopharyngeal carcinoma (NPC) is the most prevailing malignancy of the head and neck with unique geographic distribution. Southern China has one of the highest incidence rates of NPC in the world. Although radiotherapy and chemotherapy are the most important treatment modalities for NPC, recurrence, and metastasis severely interfere with the survival quality of patients. It is much-needed to find an effective method of NPC treatment with a good prognosis such as gene therapy. PFK1, a key regulatory enzyme of glycolysis, is frequently shown to be amplified and overexpressed in a variety of human cancers. However, the function of PFK1 and molecular mechanism in NPC is elusive. Here, we knockdown PFK1 expression by utilizing DNA vector-based RNA Interference. Western blotting and real-time PCR show that the expression of PFK1 is efficiently down-regulated in both protein and mRNA levels by stable transfection with PFK1 siRNA expression vector. In addition, stable knockdown of PFK1 expression inhibits cell growth, induces apoptosis, decreases the invasive capability and metastasis in the CNE2 human NPC cell line. This present study finds the importance of PFK1 which can be worked as a novel target in NPC treatment and holds great potential to be extended to other malignant cancers.


Assuntos
Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/patologia , Metástase Neoplásica/patologia , Recidiva Local de Neoplasia/genética , Fosfofrutoquinase-1/genética , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/genética , Recidiva Local de Neoplasia/patologia , Interferência de RNA/fisiologia
13.
Nat Commun ; 11(1): 4113, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807791

RESUMO

The acidic pH of tumors profoundly inhibits effector functions of activated CD8 + T-cells. We hypothesize that this is a physiological process in immune regulation, and that it occurs within lymph nodes (LNs), which are likely acidic because of low convective flow and high glucose metabolism. Here we show by in vivo fluorescence and MR imaging, that LN paracortical zones are profoundly acidic. These acidic niches are absent in athymic Nu/Nu and lymphodepleted mice, implicating T-cells in the acidifying process. T-cell glycolysis is inhibited at the low pH observed in LNs. We show that this is due to acid inhibition of monocarboxylate transporters (MCTs), resulting in a negative feedback on glycolytic rate. Importantly, we demonstrate that this acid pH does not hinder initial activation of naïve T-cells by dendritic cells. Thus, we describe an acidic niche within the immune system, and demonstrate its physiological role in regulating T-cell activation.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfonodos/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Citometria de Fluxo , Concentração de Íons de Hidrogênio , Imunoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo
14.
Sci Rep ; 10(1): 9163, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32514127

RESUMO

In solid tumors, hypoxia can trigger aberrant expression of transcription factors and genes, resulting in abnormal biological functions such as altered energetic pathways in cancer cells. Glucose metabolism is an important part of this phenomenon, which is associated with changes in the functional expression of transporters and enzymes involved in the glycolysis pathway. The latter phenomenon can finally lead to the lactate accumulation and pH dysregulation in the tumor microenvironment and subsequently further invasion and metastasis of cancer cells. Having capitalized on the computational modeling, in this study, for the first time, we aimed to investigate the effects of hypoxia-induced factor-1 (HIF-1) mediated hypoxia on the magnitude of functional expression of all the enzymes and transporters involved in the glycolysis process. The main objective was to establish a quantitative relationship between the hypoxia intensity and the intracellular lactate levels and determine the key regulators of the glycolysis pathway. This model clearly showed an increase in the lactate concentration during the oxygen depletion. The proposed model also predicted that the phosphofructokinase-1 and phosphoglucomutase enzymes might play the most important roles in the regulation of the lactate production.


Assuntos
Glicólise/genética , Hipóxia/genética , Hipóxia/metabolismo , Ácido Láctico/metabolismo , Modelos Teóricos , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais/genética , Microambiente Tumoral , Expressão Gênica/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Neoplasias/patologia , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/fisiologia , Fosfoglucomutase/genética , Fosfoglucomutase/fisiologia
15.
Mol Med Rep ; 22(3): 1727-1736, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32582969

RESUMO

Hypopharyngeal squamous cell carcinoma (HSCC) is a malignant tumor found in the head and neck region. Lactate receptor 1, also known as G protein­coupled receptor81 (GPR81), has been reported to play a vital role in cancer growth and metabolism. However, the function of GPR81 in HSCC remains largely unknown. The present study investigated the effect of GPR81 on cell survival and GPR81­mediated energy metabolism under cisplatin treatment in HSCC. GPR81 knockdown reduced the proliferation and invasion of the human HSCC cell line FaDu. Furthermore, GPR81 silencing combined with cisplatin treatment increased the expression of translocase of outer mitochondrial membrane 20 at the mRNA and protein levels (P<0.05). mRNA and protein expression of phosphofructokinase 1 in mRNA appeared to be downregulated in GPR81 knockdown FaDu cells treated with cisplatin, although this was not statistically significant. GPR81 silencing and cisplatin challenge showed no significant upregulation compared with the control, but significant downregulation in mRNA and protein levels compared with the shRNA­scramble group. Apoptosis was measured by flow cytometry with annexin V and 7­aminoactinomycin D. GPR81 silencing and cisplatin led to an increased apoptotic rate. Moreover, absence of GPR81 combined with cisplatin exposure increased caspase­3 expression and decreased Bcl­2 levels. The results of the present study suggested that GPR81 and cisplatin sensitivity played an important role in HSCC growth and metabolism.


Assuntos
Cisplatino/farmacologia , Neoplasias Hipofaríngeas/genética , Receptores Acoplados a Proteínas G/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glicólise , Humanos , Neoplasias Hipofaríngeas/tratamento farmacológico , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Complexo de Importação de Proteína Precursora Mitocondrial , Fosforilação Oxidativa , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico
16.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914380

RESUMO

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Assuntos
Glicólise , Interações Hospedeiro-Patógeno , Interleucina-1beta/metabolismo , MicroRNAs/metabolismo , Mycobacterium tuberculosis/fisiologia , Fosfofrutoquinase-1/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Sequência de Bases , Proliferação de Células , Células HEK293 , Humanos , Interferon gama/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , MicroRNAs/genética , Fosfofrutoquinase-1/genética , Células RAW 264.7 , Tuberculose/microbiologia
17.
Sci Rep ; 9(1): 18859, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827136

RESUMO

Metabolism is a critical regulator of cell fate determination. Recently, the significance of metabolic reprogramming in environmental adaptation during tumorigenesis has attracted much attention in cancer research. Recurrent mutations in the isocitrate dehydrogenase (IDH) 1 or 2 genes have been identified in several cancers, including intrahepatic cholangiocarcinoma (ICC). Mutant IDHs convert α-ketoglutarate (α-KG) to 2-hydroxyglutarate (2-HG), which affects the activity of multiple α-KG-dependent dioxygenases including histone lysine demethylases. Although mutant IDH can be detected even in the early stages of neoplasia, how IDH mutations function as oncogenic drivers remains unclear. In this study, we aimed to address the biological effects of IDH1 mutation using intrahepatic biliary organoids (IBOs). We demonstrated that mutant IDH1 increased the formation of IBOs as well as accelerated glucose metabolism. Gene expression analysis and ChIP results revealed the upregulation of platelet isoform of phosphofructokinase-1 (PFKP), which is a rate-limiting glycolytic enzyme, through the alteration of histone modification. Knockdown of the Pfkp gene alleviated the mutant IDH1-induced increase in IBO formation. Notably, the high expression of PFKP was observed more frequently in patients with IDH-mutant ICC compared to in those with wild-type IDH (p < 0.01, 80.9% vs. 42.5%, respectively). Furthermore, IBOs expressing mutant IDH1 survived the suppression of ATP production caused by growth factor depletion and matrix detachment by retaining high ATP levels through 5' adenosine monophosphate-activated protein kinase (AMPK) activation. Our findings provide a systematic understanding as to how mutant IDH induces tumorigenic preconditioning by metabolic rewiring in intrahepatic cholangiocytes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Sistema Biliar/metabolismo , Isocitrato Desidrogenase/genética , Mutação , Fosfofrutoquinase-1/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Ciclo do Ácido Cítrico , Regulação da Expressão Gênica , Glutaratos/metabolismo , Humanos , Isocitrato Desidrogenase/metabolismo , Ácidos Cetoglutáricos/metabolismo , Camundongos , Fosfofrutoquinase-1/genética
18.
Arch Biochem Biophys ; 669: 39-49, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31128085

RESUMO

To enhance our understanding of the control of archaeal carbon central metabolism, a detailed analysis of the regulation mechanisms of both fructose1,6-bisphosphatase (FruBPase) and ADP-phosphofructokinase-1 (ADP-PFK1) was carried out in the methanogen Methanosarcina acetivorans. No correlations were found among the transcript levels of the MA_1152 and MA_3563 (frubpase type II and pfk1) genes, the FruBPase and ADP-PFK1 activities, and their protein contents. The kinetics of the recombinant FruBPase II and ADP-PFK1 were hyperbolic and showed simple mixed-type inhibition by AMP and ATP, respectively. Under physiological metabolite concentrations, the FruBPase II and ADP-PFK1 activities were strongly modulated by their inhibitors. To assess whether these enzymes were also regulated by a phosphorylation/dephosphorylation process, the recombinant enzymes and cytosolic-enriched fractions were incubated in the presence of commercial protein phosphatase or protein kinase. De-phosphorylation of ADP-PFK1 slightly decreased its activity (i.e. Vmax) and did not change its kinetic parameters and oligomeric state. Thus, the data indicated a predominant metabolic regulation of both FruBPase and ADP-PFK1 activities by adenine nucleotides and suggested high degrees of control on the respective pathway fluxes.


Assuntos
Proteínas Arqueais/metabolismo , Frutose-Bifosfatase/metabolismo , Methanosarcina/metabolismo , Fosfofrutoquinase-1/metabolismo , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Proteínas Arqueais/genética , Proteínas Arqueais/isolamento & purificação , Galinhas , Frutose-Bifosfatase/genética , Frutose-Bifosfatase/isolamento & purificação , Frutosefosfatos/metabolismo , Genes Arqueais , Cinética , Methanosarcina/genética , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1/isolamento & purificação , Fosforilação , Inibidores de Proteínas Quinases/metabolismo , Processamento de Proteína Pós-Traducional
19.
Cell Death Dis ; 10(3): 228, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850587

RESUMO

Citrate, a substance being related to de novo fatty acid synthesis and tricarboxylic acid (TCA) cycle, has a pivotal role in cell survival. However, the molecular mechanisms that regulate intracellular citrate in triple-negative breast cancer (TNBC), especially under hypoxic condition, remain poorly understood. Here we find that hypoxia (1% O2) induces DNA damage-independent ATM activation (oxidized ATM) and suppression of oxidized ATM reduces intracellular citrate via decreasing the levels of phosphofructokinase (PFKP) and citrate synthase (CS), two key glucose metabolism-associated enzymes. Mechanistically, PFKP is regulated by HIF1A at the translational level, whereas CS is of posttranscriptional regulation by UBR5-mediated ubiquitination. Interestingly, accumulation of citrate in cytoplasm or exogenous citrate significantly enhances cell migration, invasion, and metastasis of hypoxic TNBC cells in vitro and in mice xenografts. The underlying mechanism mainly involves citrate-stimulated activation of the AKT/ERK/MMP2/9 signaling axis. Our findings unravel a novel function of oxidized ATM in promoting migration, invasion, and metastasis of TNBC.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Citrato (si)-Sintase/metabolismo , Ácido Cítrico/metabolismo , Fosfofrutoquinase-1/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/química , Proteínas Mutadas de Ataxia Telangiectasia/genética , Hipóxia Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Respiração Celular , Sobrevivência Celular/genética , Citrato (si)-Sintase/genética , Metabolismo Energético , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Nus , Metástase Neoplásica , Fosfofrutoquinase-1/genética , Transdução de Sinais/genética , Transplante Heterólogo , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
20.
Nat Commun ; 9(1): 4683, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30409970

RESUMO

The Warburg effect is a prominent metabolic feature associated with neoplastic diseases; however, the underlying mechanism remains incompletely understood. TAp73, a structural homolog of the tumor suppressor p53, is frequently overexpressed in human tumors, indicating a proliferative advantage that it can confer to tumor cells. Here we show that TAp73 stimulates the expression of phosphofructokinase-1, liver type (PFKL), which catalyzes the committed step in glycolysis. Through this regulation, TAp73 enhances glucose consumption and lactate excretion, promoting the Warburg effect. By activating PFKL, TAp73 also increases ATP production and bolsters anti-oxidant defense. TAp73 deficiency results in a pronounced reduction in tumorigenic potential, which can be rescued by forced PFKL expression. These findings establish TAp73 as a critical regulator of glycolysis and reveal a mechanism by which tumor cells achieve the Warburg effect to enable oncogenic growth.


Assuntos
Glicólise , Fosfofrutoquinase-1/genética , Transcrição Gênica , Proteína Tumoral p73/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Sequência de Bases , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Fibroblastos/metabolismo , Glucose/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glicólise/genética , Homeostase , Humanos , Ácido Láctico/metabolismo , Fígado/enzimologia , Camundongos , Fosfofrutoquinase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA