Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Acta Pharmacol Sin ; 44(3): 680-692, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36114272

ABSTRACT

The growth of solid tumors depends on tumor vascularization and the endothelial cells (ECs) that line the lumen of blood vessels. ECs generate a large fraction of ATP through glycolysis, and elevation of their glycolytic activity is associated with angiogenic behavior in solid tumors. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) positively regulates glycolysis via fructose-2/6-bisphosphate, the product of its kinase activity. Partial inhibition of glycolysis in tumor ECs by targeting PFKFB3 normalizes the otherwise abnormal tumor vessels, thereby reducing metastasis and improving the outcome of chemotherapy. Although a limited number of tool compounds exist, orally available PFKFB3 inhibitors are unavailable. In this study we conducted a high-throughput screening campaign against the kinase activity of PFKFB3, involving 250,240 chemical compounds. A total of 507 initial hits showing >50% inhibition at 20 µM were identified, 66 of them plus 1 analog from a similarity search consistently displayed low IC50 values (<10 µM). In vitro experiments yielded 22 nontoxic hits that suppressed the tube formation of primary human umbilical vein ECs at 10 µM. Of them, 15 exhibited binding affinity to PFKFB3 in surface plasmon resonance assays, including 3 (WNN0403-E003, WNN1352-H007 and WNN1542-F004) that passed the pan-assay interference compounds screening without warning flags. This study provides potential leads to the development of new PFKFB3 inhibitors.


Subject(s)
High-Throughput Screening Assays , Neoplasms , Phosphofructokinase-2 , Humans , Glycolysis , Human Umbilical Vein Endothelial Cells/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism
2.
Oxid Med Cell Longev ; 2022: 7548145, 2022.
Article in English | MEDLINE | ID: mdl-36187335

ABSTRACT

Intervertebral disc (IVD) degeneration (IVDD) is a characteristic of the dominating pathological processes of nucleus pulposus (NP) cell senescence, abnormal synthesis and irregular distribution of extracellular matrix (ECM), and tumor necrosis factor-α (TNF-α) induced inflammation. Nowadays, IVD acid environment variation which accelerates the pathological processes mentioned above arouses researchers' attention. KAN0438757 (KAN) is an effective inhibitor of selective metabolic kinase phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) that has both energy metabolism reprogramming and anti-inflammatory effects. Therefore, a potential therapeutic benefit of KAN lies in its ability to inhibit the development of IVDD. This study examined in vitro KAN toxicity in NP primary cells (NPPs). Moreover, KAN influenced tumor necrosis factor-α (TNF-α) induced ECM anabolism and catabolism; the inflammatory signaling pathway activation and the energy metabolism phenotype were also examined in NPPs. Furthermore, KAN's therapeutic effect was investigated in vivo using the rat tail disc puncture model. Phenotypically speaking, the KAN treatment partially rescued the ECM degradation and glycolysis energy metabolism phenotypes of NPPs induced by TNF-α. In terms of mechanism, KAN inhibited the activation of MAPK and NF-κB inflammatory signaling pathways induced by TNF-α and reprogramed the energy metabolism. For the therapeutic aspect, the rat tail disc puncture model demonstrated that KAN has a significant ameliorated effect on the progression of IVDD. To sum up, our research successfully authenticated the potential therapeutic effect of KAN on IVDD and declaimed its mechanisms of both novel energy metabolism reprogramming and conventional anti-inflammation effect.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Nucleus Pulposus , Phosphofructokinase-2/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Energy Metabolism , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/pathology , NF-kappa B/metabolism , Nucleus Pulposus/pathology , Phosphofructokinase-2/metabolism , Phosphofructokinase-2/pharmacology , Rats , Signal Transduction , Succinimides , Tumor Necrosis Factor-alpha/metabolism
3.
Br J Cancer ; 127(5): 811-823, 2022 09.
Article in English | MEDLINE | ID: mdl-35637411

ABSTRACT

BACKGROUND: Multiple mechanisms have been proposed that lead to reduced effectiveness of trastuzumab in HER2-positive gastric cancer (GC), yet resistance to trastuzumab remains a challenge in clinics. METHODS: We established trastuzumab-resistant cells and patient-derived xenografts models to measure metabolic levels and vascular density and shape. The HER2-positive GC patient samples were used to determine clinical significance. We also measured protein expression and phosphorylation modifications to determine those alterations related to resistance. In vivo studies combining inhibitor of PFKFB3 with trastuzumab corroborated the in vitro findings. RESULTS: The 6-phosphofructo-2-kinase (PFKFB3)-mediated trastuzumab resistance pathways in HER2-positive GC by activating the glycolytic pathway. We also found vessels are chaotic and destabilised in the tumour during the trastuzumab resistance process. Inhibition of PFKFB3 significantly diminished tumour proliferation and promoted vessel normalisation in the patient-derived xenograft model. Mechanistically, PFKFB3 promoted the secretion of CXCL8 into the tumour microenvironment, and phosphorylated Ser1151 of ERBB2, enhancing the transcription of CXCL8 by activating the PI3K/AKT/NFκB p65 pathway. CONCLUSIONS: Our current findings discover that PFKFB3 inhibitors might be effective tools to overcome adjuvant therapy resistance in HER2-positive GC and reshaping the microenvironment by normalising tumour vessels is a novel strategy to overcome trastuzumab resistance.


Subject(s)
Phosphofructokinase-2 , Stomach Neoplasms , Trastuzumab , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/genetics , Phosphofructokinase-2/pharmacology , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Tumor Microenvironment
4.
Cell Immunol ; 371: 104468, 2022 01.
Article in English | MEDLINE | ID: mdl-34968772

ABSTRACT

Dendritic cells (DCs) play central role in innate as well as adaptive immune responses regulated by diverse DC subtypes that vary in terms of surface markers, transcriptional profile and functional responses. Generation of DC diversity from progenitor stage is tightly regulated by complex molecular inter-play between transcription factors. We earlier demonstrated that Batf3 and Id2 expression have a synergistic effect on the Irf8 directed classical cDC1 development. In present study, Bi-molecular fluorescence complementation assay suggested that IRF8 interacts with BATF3, and ID2 may aid cDC1 development independently. Genome wide recruitment analysis of IRF8 and BATF3 from different DC subtypes led to identification of the overlapping regions of occupancy by these two transcription factors. Further analysis of overlapping peaks of IRF8 and BATF3 occupancy in promoter region within the cDC1 subtype specific transcriptional pattern identified a metabolically important Pfkfb3 gene. Among various immune cell types; splenic cDC1 subtype displayed enhanced expression of Pfkfb3. Analysis of Irf8-/-, Irf8R294C and Batf3DCKO DC confirmed direct regulation of Pfkfb3 enhanced expression specifically in cDC1 subtype. Further we show that inhibition of PFKFB3 enzymatic activity by a chemical agent PFK15 led to reduction in cDC1 subtype in both in vitro FLDC cultures as well as in vivo mouse spleens. Together, our study identified the direct regulation of cDC1 specific enhanced expression of Pfkfb3 in glycolysis and cDC1 biology.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/metabolism , Phosphofructokinase-2/biosynthesis , Repressor Proteins/metabolism , Animals , Cell Line , Female , Gene Expression Regulation/genetics , Glycolysis/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/genetics , Promoter Regions, Genetic/genetics , Pyridines/pharmacology , Quinolines/pharmacology
5.
Front Immunol ; 12: 779787, 2021.
Article in English | MEDLINE | ID: mdl-34899740

ABSTRACT

Rheumatoid arthritis (RA) is a classic autoimmune disease characterized by uncontrolled synovial proliferation, pannus formation, cartilage injury, and bone destruction. The specific pathogenesis of RA, a chronic inflammatory disease, remains unclear. However, both key glycolysis rate-limiting enzymes, hexokinase-II (HK-II), phosphofructokinase-1 (PFK-1), and pyruvate kinase M2 (PKM2), as well as indirect rate-limiting enzymes, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), are thought to participate in the pathogenesis of RA. In here, we review the latest literature on the pathogenesis of RA, introduce the pathophysiological characteristics of HK-II, PFK-1/PFKFB3, and PKM2 and their expression characteristics in this autoimmune disease, and systematically assess the association between the glycolytic rate-limiting enzymes and RA from a molecular level. Moreover, we highlight HK-II, PFK-1/PFKFB3, and PKM2 as potential targets for the clinical treatment of RA. There is great potential to develop new anti-rheumatic therapies through safe inhibition or overexpression of glycolysis rate-limiting enzymes.


Subject(s)
Arthritis, Rheumatoid/enzymology , Enzymes/metabolism , Glucose/metabolism , Glycolysis , Joints/enzymology , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Enzyme Inhibitors/therapeutic use , Glycolysis/drug effects , Hexokinase/antagonists & inhibitors , Hexokinase/metabolism , Humans , Joints/drug effects , Joints/immunology , Kinetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Phosphofructokinase-1/antagonists & inhibitors , Phosphofructokinase-1/metabolism , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
6.
Med Oncol ; 39(1): 10, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34761330

ABSTRACT

Numerous studies have shown that 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3), a pivotal enzyme in modulating glycolysis, plays vital roles in various physiological processes. PFKFB3 activity could be regulated by several factors, such as hypoxia and AMPK signaling; however, it could also function as upstream of AMPK signaling. Here, we showed that PFKFB3 inhibitor PFK-15 induced cell viability loss and apoptosis. Deprivation of PFKFB3 inhibited autophagy, while enhanced the ubiquitin-proteasome degradation pathway. Furthermore, PFK-15 reduced both the AMPK and AKT-mTORC1 signaling pathways, as the attenuated phosphorylation level of kinases themselves and their substrates. The addition of AICAR rescued the AMPK activity and autophagy, but enhanced PFK-15-induced cell viability loss. In fact, AICAR promoted the cytotoxicity of PFK-15 even in the AMPKα1/2-silenced cells, indicating AICAR might function in an AMPK-independent manner. Nevertheless, AICAR further reduced the AKT-mTORC1 activity down-regulated by PFK-15. Moreover, it failed to enhance PFK-15's cytotoxicity in the AKT1/2-silenced cells, indicating AKT-mTORC1 participated during these processes. Collectively, the presented data demonstrated that PFK-15 inhibited cell viability, AMPK, and AKT-mTORC1 signaling, and AICAR probably enhanced the cell viability loss aroused by PFK-15 in an AKT-dependent and AMPK-independent manner, thereby revealing a more intimate relationship among PFKFB3, AMPK, and AKT-mTORC1 signaling pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Phosphofructokinase-2/antagonists & inhibitors , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Humans , Signal Transduction/drug effects
7.
Cells ; 10(7)2021 07 03.
Article in English | MEDLINE | ID: mdl-34359849

ABSTRACT

Tyrosine kinase inhibitors (TKIs) targeting the kinase domain of the epidermal growth factor receptor (EGFR), such as erlotinib, have dramatically improved clinical outcomes of patients with EGFR-driven non-small cell lung carcinomas (NSCLCs). However, intrinsic or acquired resistance remains a clinical barrier to the success of FDA-approved EGFR TKIs. Multiple mechanisms of resistance have been identified, including the activation of prosurvival autophagy. We have previously shown that the expression and activity of PFKFB3-a known driver of glycolysis-is associated with resistance to erlotinib and that PFKFB3 inhibition improves the response of NSCLC cells to erlotinib. This study focuses on investigating the role of PFKFB3 in regulating erlotinib-driven autophagy to escape resistance to erlotinib. We evaluated the consequence of pharmacological inhibition of PFKFB3 on erlotinib-driven autophagy in NSCLC cells with different mutation statuses. Here, we identify PFKFB3 as a mediator of erlotinib-induced autophagy in NSCLCs. We demonstrate that PFKFB3 inhibition sensitizes NCSLCs to erlotinib via impairing autophagy flux. In summary, our studies uncovered a novel crosstalk between PFKFB3 and EGFR that regulates erlotinib-induced autophagy, thus contributing to erlotinib sensitivity in NSCLCs.


Subject(s)
Autophagy , Carcinoma, Non-Small-Cell Lung/pathology , Erlotinib Hydrochloride/pharmacology , Lung Neoplasms/pathology , Phosphofructokinase-2/antagonists & inhibitors , Adenylate Kinase/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Mutant Proteins/metabolism , Phosphofructokinase-2/metabolism , Protein Kinase Inhibitors/pharmacology , Sequestosome-1 Protein/metabolism
8.
Clin Transl Gastroenterol ; 12(7): e00377, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34193800

ABSTRACT

OBJECTIVES: PFKFB3 regulates glycolysis in tumor cells, might function as an oncogene, and is associated with cancer metastasis. However, its role in gastric cancer (GC) remains largely unknown. METHODS: PFKFB3 expression was assessed by immunohistochemistry (IHC) in GC tissues and paired paracancerous histological normal tissues (PCHNTs). The associations of PFKFB3 expression with clinical features and HIF-1α, Ki-67, E-cadherin, Snail, and Vimentin expression levels were assessed. A series of in vivo and in vitro experiments were performed to investigate the effects of PFKFB3 on the growth, migration, and invasion of GC cells. RESULTS: We found that PFKFB3 expression was significantly higher in GC tissues compared with PCHNTs (P = 0.000). PFKFB3 expression was positively correlated with tumor size (P = 0.000), differentiation (P = 0.025), venous invasion (P = 0.084), nerve invasion (P = 0.014), lymphatic invasion (P = 0.000), local invasion (P = 0.000), invasive depth (P = 0.000), nodal metastasis (P = 0.000), tumor-node-metastasis stage (P = 0.000), and patient survival (P = 0.000). Notably, PFKFB3 upregulation was highly correlated with increased epithelial-mesenchymal transition (EMT) in GC samples. PFKFB3 overexpression positively modulated cell proliferation, migration, and EMT in GC cells in vitro, with concomitant activation of NF-κB signaling. Administration of an NF-κB inhibitor attenuated PFKFB3-induced EMT in GC cells. PFKFB3 overexpression promoted tumor development and EMT in nude mice, which were attenuated by PFK-15, a PFKFB3 inhibitor. DISCUSSION: PFKFB3 could potentiate malignancy in GC cells through NF-κB pathway-mediated EMT, suggesting PFKFB3 represents a potential target for GC therapy.


Subject(s)
Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition , Female , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphofructokinase-2/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Tumor Burden , Vimentin/genetics , Vimentin/metabolism
9.
Biochem Biophys Res Commun ; 571: 118-124, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34325126

ABSTRACT

Activating mutations of the oncogenic KRAS in pancreatic ductal adenocarcinoma (PDAC) are associated with an aberrant metabolic phenotype that may be therapeutically exploited. Increased glutamine utilization via glutaminase-1 (GLS1) is one such feature of the activated KRAS signaling that is essential to cell survival and proliferation; however, metabolic plasticity of PDAC cells allow them to adapt to GLS1 inhibition via various mechanisms including activation of glycolysis, suggesting a requirement for combinatorial anti-metabolic approaches to combat PDAC. We investigated whether targeting the glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) in combination with GLS1 can selectively prevent the growth of KRAS-transformed cells. We show that KRAS-transformation of pancreatic duct cells robustly sensitizes them to the dual targeting of GLS1 and PFKFB3. We also report that this sensitivity is preserved in the PDAC cell line PANC-1 which harbors an activating KRAS mutation. We then demonstrate that GLS1 inhibition reduced fructose-2,6-bisphosphate levels, the product of PFKFB3, whereas PFKFB3 inhibition increased glutamine consumption, and these effects were augmented by the co-inhibition of GLS1 and PFKFB3, suggesting a reciprocal regulation between PFKFB3 and GLS1. In conclusion, this study identifies a novel mutant KRAS-induced metabolic vulnerability that may be targeted via combinatorial inhibition of GLS1 and PFKFB3 to suppress PDAC cell growth.


Subject(s)
Antineoplastic Agents/pharmacology , Benzeneacetamides/pharmacology , Glutaminase/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Phosphofructokinase-2/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Thiadiazoles/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Glutaminase/genetics , Glutaminase/metabolism , Humans , Mutation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
10.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073144

ABSTRACT

Angiogenesis is the process of new blood vessel formation. In this complex orchestrated growth, many factors are included. Lately, focus has shifted to endothelial cell metabolism, particularly to the PFKFB3 protein, a key regulatory enzyme of the glycolytic pathway. A variety of inhibitors of this important target have been studied, and a plethora of biological effects related to the process of angiogenesis have been reported. However, recent studies have disputed their mechanism of action, questioning whether all the effects are indeed due to PFKFB3 inhibition. Remarkably, the most well-studied inhibitor, 3PO, does not bind to PFKFB3, raising questions about this target. In our study, we aimed to elucidate the effects of PFKFB3 inhibition in angiogenesis by using the small molecule AZ67. We used isothermal titration calorimetry and confirmed binding to PFKFB3. In vitro, AZ67 did not decrease lactate production in endothelial cells (ECs), nor ATP levels, but exhibited good inhibitory efficacy in the tube-formation assay. Surprisingly, this was independent of EC migratory and proliferative abilities, as this was not diminished upon treatment. Strikingly however, even the lowest dose of AZ67 demonstrated significant inhibition of angiogenesis in vivo. To our knowledge, this is the first study to demonstrate that the process of angiogenesis can be disrupted by targeting PFKFB3 independently of glycolysis inhibition.


Subject(s)
Cell Proliferation/drug effects , Enzyme Inhibitors , Glycolysis/drug effects , Neovascularization, Physiologic/drug effects , Phosphofructokinase-2 , Animals , Cell Line , Endothelial Cells , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Inbred C57BL , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism , Protein Binding
11.
Front Immunol ; 12: 669456, 2021.
Article in English | MEDLINE | ID: mdl-34163475

ABSTRACT

In Type 1 Diabetes (T1D), CD4+ T cells initiate autoimmune attack of pancreatic islet ß cells. Importantly, bioenergetic programs dictate T cell function, with specific pathways required for progression through the T cell lifecycle. During activation, CD4+ T cells undergo metabolic reprogramming to the less efficient aerobic glycolysis, similarly to highly proliferative cancer cells. In an effort to limit tumor growth in cancer, use of glycolytic inhibitors have been successfully employed in preclinical and clinical studies. This strategy has also been utilized to suppress T cell responses in autoimmune diseases like Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), and Rheumatoid Arthritis (RA). However, modulating T cell metabolism in the context of T1D has remained an understudied therapeutic opportunity. In this study, we utilized the small molecule PFK15, a competitive inhibitor of the rate limiting glycolysis enzyme 6-phosphofructo-2-kinase/fructose-2,6- biphosphatase 3 (PFKFB3). Our results confirmed PFK15 inhibited glycolysis utilization by diabetogenic CD4+ T cells and reduced T cell responses to ß cell antigen in vitro. In an adoptive transfer model of T1D, PFK15 treatment delayed diabetes onset, with 57% of animals remaining euglycemic at the end of the study period. Protection was due to induction of a hyporesponsive T cell phenotype, characterized by increased and sustained expression of the checkpoint molecules PD-1 and LAG-3 and downstream functional and metabolic exhaustion. Glycolysis inhibition terminally exhausted diabetogenic CD4+ T cells, which was irreversible through restimulation or checkpoint blockade in vitro and in vivo. In sum, our results demonstrate a novel therapeutic strategy to control aberrant T cell responses by exploiting the metabolic reprogramming of these cells during T1D. Moreover, the data presented here highlight a key role for nutrient availability in fueling T cell function and has implications in our understanding of T cell biology in chronic infection, cancer, and autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/drug therapy , Enzyme Inhibitors/pharmacology , Glycolysis/drug effects , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Quinolines/pharmacology , Adoptive Transfer , Animals , Antigens, CD/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Cellular Reprogramming/drug effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Female , Male , Mice, Inbred NOD , Mice, SCID , Phosphofructokinase-2/metabolism , Programmed Cell Death 1 Receptor/metabolism , Time Factors , Lymphocyte Activation Gene 3 Protein
12.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922320

ABSTRACT

A high rate of glycolysis is considered a hallmark of tumor progression and is caused by overexpression of the enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3). Therefore, we analyzed the possibility of inhibiting tumor and endothelial cell metabolism through the inhibition of PFKFB3 by a small molecule, (E)-1-(pyridin-4-yl)-3-(quinolin-2-yl)prop-2-en-1-one (PFK15), as a promising therapy. The effects of PFK15 on cell proliferation and apoptosis were analyzed on human umbilical vein endothelial cells (HUVEC) and the human colorectal adenocarcinoma cell line DLD1 through cytotoxicity and proliferation assays, flow cytometry, and western blotting. The results showed that PFK15 inhibited the proliferation of both cell types and induced apoptosis with decreasing the Bcl-2/Bax ratio. On the basis of the results obtained from in vitro experiments, we performed a study on immunodeficient mice implanted with DLD1 cells. We found a reduced tumor mass after morning PFK15 treatment but not after evening treatment, suggesting circadian control of underlying processes. The reduction in tumor size was related to decreased expression of Ki-67, a marker of cell proliferation. We conclude that inhibition of glycolysis can represent a promising therapeutic strategy for cancer treatment and its efficiency is circadian dependent.


Subject(s)
Chronotherapy/methods , Colonic Neoplasms/drug therapy , Glucose/metabolism , Glycolysis , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Quinolines/pharmacology , Animals , Apoptosis , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Male , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Pharmacol Res ; 168: 105592, 2021 06.
Article in English | MEDLINE | ID: mdl-33813027

ABSTRACT

Abnormal glycolytic metabolism contributes to angiogenic sprouting involved in atherogenesis. We investigated the potential anti-angiogenic properties of specific 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) inhibitors in endothelial cells (ECs). ECs were treated with PFKFB3 inhibitors (named PA-1 and PA-2) and their effects on metabolic and functional characteristics of ECs were investigated. The anti-glycolytic compound 3-(pyridinyl)- 1-(4-pyridinyl)- 2-propen-1-one (3PO) was used as reference compound. PFKFB3 expression and activity (IC50 about 3-21 nM) was inhibited upon treatment with both compounds. Glucose uptake and lactate export were measured using commercial assays and showed a partial reduction up to 40%. PFKFB3 inhibition increased intracellular lactate accumulation, and reduced expression of monocarboxylate transporters-1 (MCT1) and MCT4. Furthermore, endothelial cell migration and proliferation assays demonstrated significant reduction upon treatment with both compounds. Matrix- metalloproteinase (MMP) activity, measured by gelatin zymography, and expression was significantly reduced (up to 25%). In addition, PA compounds downregulated the expression of VCAM-1, VE-cadherin, VEGFa, VEGFR2, TGF-ß, and IL-1ß, in inflamed ECs. Finally, PA-1 and PA-2 treatment impaired the formation of angiogenic sprouts measured by both morphogenesis and spheroid-based angiogenesis assays. Our data demonstrate that the anti-glycolytic PA compounds may affect several steps involved in angiogenesis. Targeting the key glycolytic enzyme PFKFB3 might represent an attractive therapeutic strategy to improve the efficacy of cancer treatments, or to be applied in other pathologies where angiogenesis is a detrimental factor.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Phosphofructokinase-2/antagonists & inhibitors , Cells, Cultured , Humans , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/physiology , Muscle Proteins/physiology , NAD/metabolism , Neovascularization, Pathologic/drug therapy , Symporters/physiology
14.
Oncogene ; 40(8): 1409-1424, 2021 02.
Article in English | MEDLINE | ID: mdl-33420377

ABSTRACT

The advanced or recurrent endometrial cancer (EC) has a poor prognosis because of chemoresistance. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a glycolytic enzyme, is overexpressed in a variety of human cancers and plays important roles in promoting tumor cell growth. Here, we showed that high expression of PFKFB3 in EC cell lines is associated with chemoresistance. Pharmacological inhibition of PFKFB3 with PFK158 and or genetic downregulation of PFKFB3 dramatically suppressed cell proliferation and enhanced the sensitivity of EC cells to carboplatin (CBPt) and cisplatin (Cis). Moreover, PFKFB3 inhibition resulted in reduced glucose uptake, ATP production, and lactate release. Notably, we found that PFK158 with CBPt or Cis exerted strong synergistic antitumor activity in chemoresistant EC cell lines, HEC-1B and ARK-2 cells. We also found that the combination of PFK158 and CBPt/Cis induced apoptosis- and autophagy-mediated cell death through inhibition of the Akt/mTOR signaling pathway. Mechanistically, we found that PFK158 downregulated the CBPt/Cis-induced upregulation of RAD51 expression and enhanced CBPt/Cis-induced DNA damage as demonstrated by an increase in γ-H2AX levels in HEC-1B and ARK-2 cells, potentially revealing a means to enhance PFK158-induced chemosensitivity. More importantly, PFK158 treatment, either as monotherapy or in combination with CBPt, led to a marked reduction in tumor growth in two chemoresistant EC mouse xenograft models. These data suggest that PFKFB3 inhibition alone or in combination with standard chemotherapy may be used as a novel therapeutic strategy for improved therapeutic efficacy and outcomes of advanced and recurrent EC patients.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Phosphofructokinase-2/genetics , Apoptosis/drug effects , Carboplatin/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
15.
Cancer Lett ; 500: 29-40, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33307155

ABSTRACT

Glycolysis emerges as a new therapeutic target for malignancies. The inhibition of glycolytic activator, PFKFB3, repairs tumor endothelial cell function, and normalizing the tumor microenvironment. We aimed to investigate the significance of PFKFB3 in HCC, and the effects of the PFKFB3 inhibitor, PFK15, in HCC tumor cells and tumor endothelial cells. Double immunofluorescent staining of PFKFB3 and CD31 in HCC tissues revealed that high PFKFB3 expression in both tumor cells and tumor endothelial cells was significantly correlated with poor prognosis. Multivariate analysis identified PFKFB3 expression as an independent prognostic factor. PFK15 suppressed proliferation of HCC cell line and tumor endothelial cells in vitro. In a subcutaneous tumor model of the HCC cell line with tumor endothelial cells, PFK15 suppressed tumor growth and induced apoptosis. Moreover, PFK15 treatment induced tumor vessel normalization, decreasing vessel diameter with pericyte attachment and improving vessel perfusion. High PFKFB3 expression in both tumor cells and tumor endothelial cells was identified as a novel prognostic marker in HCC. Targeting PFKFB3 via PFK15 might be a promising strategy for suppressing tumor growth and inducing tumor vessel normalization.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phosphofructokinase-2/genetics , Pyridines/pharmacology , Quinolines/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Blood Vessels/drug effects , Blood Vessels/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Phosphofructokinase-2/antagonists & inhibitors , Prognosis , Tumor Microenvironment/drug effects
16.
Theranostics ; 10(16): 7245-7259, 2020.
Article in English | MEDLINE | ID: mdl-32641990

ABSTRACT

Rationale: Tumor vascular normalization (TVN) is emerging to enhance the efficacy of anticancer treatment in many cancers including glioblastoma (GBM). However, a common and severe challenge being currently faced is the transient TVN effect, hampering the sustained administration of anticancer therapy during TVN window. Additionally, the lack of non-contrast agent-based imaging biomarkers to monitor TVN process postpones the clinical translation of TVN strategy. In this study, we investigated whether dual inhibition of VEGF and the glycolytic activator PFKFB3 could reinforce the TVN effect in GBM. Dynamic contrast-enhanced-magnetic resonance imaging (DCE-MRI) and intravoxel incoherent motion (IVIM)-MRI were performed to monitor TVN process and to identify whether IVIM-MRI is a candidate or complementary imaging biomarker for monitoring TVN window without exogenous contrast agent administration. Methods: Patient-derived orthotopic GBM xenografts in mice were established and treated with bevacizumab (BEV), 3PO (PFKFB3 inhibitor), BEV+3PO dual therapy, or saline. The vascular morphology, tumor hypoxia, and lactate level were evaluated before and at different time points after treatments. Doxorubicin was used to evaluate chemotherapeutic efficacy and drug delivery. Microarray of angiogenesis cytokines and western blotting were conducted to characterize post-treatment molecular profiling. TVN process was monitored by DCE- and IVIM-MRI. Correlation analysis of pathological indicators and MRI parameters was further analyzed. Results: Dual therapy extended survival and delayed tumor growth over each therapy alone, concomitant with a decrease of cell proliferation and an increase of cell apoptosis. The dual therapy reinforces TVN effect, thereby alleviating tumor hypoxia, reducing lactate production, and improving the efficacy and delivery of doxorubicin. Mechanistically, several angiogenic cytokines and pathways were downregulated after dual therapy. Notably, dual therapy inhibited Tie1 expression, the key regulator of TVN, in both endothelial cells and tumor cells. DCE- and IVIM-MRI data showed that dual therapy induced a more homogenous and prominent TVN effect characterized by improved vascular function in tumor core and tumor rim. Correlation analysis revealed that IVIM-MRI parameter D* had better correlations with TVN pathological indicators compared with the DCE-MRI parameter Ktrans. Conclusions: Our results propose a rationale to overcome the current limitation of BEV monotherapy by integrating the synergistic effects of VEGF and PFKFB3 blockade to enhance chemotherapy efficacy through a sustained TVN effect. Moreover, we unveil IVIM-MRI parameter D* has much potential as a complementary imaging biomarker to monitor TVN window more precisely without exogenous contrast agent injection.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Neovascularization, Pathologic/diagnosis , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Brain/blood supply , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Glioblastoma/blood supply , Glioblastoma/diagnosis , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells , Humans , Lactic Acid/analysis , Lactic Acid/metabolism , Male , Mice , Multiparametric Magnetic Resonance Imaging , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
17.
Eur J Med Chem ; 203: 112612, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32679452

ABSTRACT

Cancer cells adopt aerobic glycolysis as the major source of energy and biomass production for fast cell proliferation. The bifunctional enzyme, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), plays a crucial role in the regulation of glycolysis by controlling the steady-state cytoplasmic levels of fructose-2,6-bisphosphate (F2,6BP), which is the most potent allosteric activator of 6-phosphofructo-1-kinase (PFK-1), a key rate-limiting enzyme of glycolysis. Therefore, selective inhibition of PFKFB3 has gained substantial interest as an attractive strategy for cancer therapy. In recent years, numerous class PFKFB3 inhibitors have been disclosed, and emerging trends such as the availability of PFKFB3 crystal structures, structure-based screening strategies and diverse functional assays are improving optimization and development of original leads. Herein, we review the structure and function of PFKFB3 as well as the representative small-molecule inhibitors, in particular emphasis on their chemical structures, pharmacological properties, selectivity, binding modes and structure-activity relationships (SARs).


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Phosphofructokinase-2/antagonists & inhibitors , Animals , Humans , Structure-Activity Relationship
18.
Cancer Res ; 80(16): 3251-3264, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32580961

ABSTRACT

KRAS is mutated in most pancreatic ductal adenocarcinomas (PDAC) and yet remains undruggable. Here, we report that p38γ MAPK, which promotes PDAC tumorigenesis by linking KRAS signaling and aerobic glycolysis (also called the Warburg effect), is a novel therapeutic target. p38γ interacted with a glycolytic activator PFKFB3 that was dependent on mutated KRAS. KRAS transformation and overexpression of p38γ increased expression of PFKFB3 and glucose transporter GLUT2, conversely, silencing mutant KRAS, and p38γ decreased PFKFB3 and GLUT2 expression. p38γ phosphorylated PFKFB3 at S467, stabilized PFKFB3, and promoted their interaction with GLUT2. Pancreatic knockout of p38γ decreased p-PFKFB3/PFKFB3/GLUT2 protein levels, reduced aerobic glycolysis, and inhibited PDAC tumorigenesis in KPC mice. PFKFB3 and GLUT2 depended on p38γ to stimulate glycolysis and PDAC growth and p38γ required PFKFB3/S467 to promote these activities. A p38γ inhibitor cooperated with a PFKFB3 inhibitor to blunt aerobic glycolysis and PDAC growth, which was dependent on p38γ. Moreover, overexpression of p38γ, p-PFKFB3, PFKFB3, and GLUT2 in PDAC predicted poor clinical prognosis. These results indicate that p38γ links KRAS oncogene signaling and aerobic glycolysis to promote pancreatic tumorigenesis through PFKFB3 and GLUT2, and that p38γ and PFKFB3 may be targeted for therapeutic intervention in PDAC. SIGNIFICANCE: These findings show that p38γ links KRAS oncogene signaling and the Warburg effect through PFKBF3 and Glut2 to promote pancreatic tumorigenesis, which can be disrupted via inhibition of p38γ and PFKFB3.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Glucose Transporter Type 2/metabolism , Glycolysis , Mitogen-Activated Protein Kinase 12/metabolism , Pancreatic Neoplasms/etiology , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Aerobiosis , Animals , Carcinoma, Pancreatic Ductal/prevention & control , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Collagen , Drug Combinations , Female , Gene Knockout Techniques , Gene Silencing , Genes, ras , Genotyping Techniques , Humans , Laminin , Male , Mice , Mitogen-Activated Protein Kinase 12/antagonists & inhibitors , Mitogen-Activated Protein Kinase 12/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/prevention & control , Phosphorylation , Prognosis , Proteoglycans , Proto-Oncogene Proteins p21(ras)/genetics
19.
Anticancer Res ; 40(5): 2613-2625, 2020 May.
Article in English | MEDLINE | ID: mdl-32366406

ABSTRACT

BACKGROUND/AIM: The occurrence of BRAFV600E mutation causes an up-regulation of the B-raf kinase activity leading to the stabilization of hypoxia-inducible factor 1-alpha (HIF-1α) - the promoter of the 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) enzyme. The aim of the study was to examine the effect of the (2E)-3-(3-Pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO), as an inhibitor of PFKFB3, on human melanoma cells (A375) with endogenous BRAFV600E mutation. MATERIALS AND METHODS: A375 cells were exposed to different concentrations of 3PO and the following tests were performed: docking, cytotoxicity assay, immunocytochemistry staining glucose uptake, clonogenic assay, holotomography imaging, and flow cytometry. RESULTS: Our studies revealed that 3PO presents a dose-dependent and time-independent cytotoxic effect and promotes apoptosis of A375 cells. Furthermore, the obtained data indicate that 3PO induces cell cycle arrest in G1/0 and glucose uptake reduction. CONCLUSION: Taking all together, our research demonstrated a here should be proapoptotic and antiproliferative effect of 3PO on A375 human melanoma cells.


Subject(s)
Enzyme Inhibitors/pharmacology , Melanoma/enzymology , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Catalytic Domain , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Glucose/metabolism , Humans , Melanoma/pathology , Molecular Docking Simulation , Molecular Targeted Therapy , Phosphofructokinase-2/metabolism , Pyridines/chemistry , Tumor Stem Cell Assay
20.
Biochem Biophys Res Commun ; 526(4): 978-985, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32299611

ABSTRACT

Acute myeloid leukemia (AML), which is characterized by an overproliferation of blood cells, is divided into several subtypes in adults and children. Of those subtypes, acute monocytic leukemia (M4/M5, AMoL) is reported to be associated with abnormal gene fusions that result in monocytic cell differentiation being blocked. However, few studies have shown a relationship between cellular metabolism and the initiation of AMoL. Here, we use the open-access database TCGA to analyze the expression of enzymes in the metabolic cycle and find that PFKFB4 is highly expressed in AMoL. Subsequently, knocking down PFKFB4 in THP-1 and U937 cells significantly inhibits cell growth and increases the sensitivity of cells to chemical drug-induced apoptosis. In line with the gene-editing alterations, treatment with a PFKFB4 inhibitor exhibits similar effects on THP-1 and U937 proliferation and apoptosis. In addition, we find that PFKFB4 functions as a reliable target of the epigenetic regulator MLL, which is a well-known modulator in AMoL. Mechanistically, MLL promotes PFKFB4 expression at the transcriptional level through the putative E2F6 binding site in the promoter of the pfkfb4 gene. Taken together, our results suggest PFKFB4 serves as a downstream target of MLL and functions as a potent therapeutic target in AMoL.


Subject(s)
Leukemia, Monocytic, Acute/pathology , Phosphofructokinase-2/metabolism , Apoptosis/drug effects , Base Sequence , Cell Survival/drug effects , Gene Expression Regulation, Leukemic/drug effects , Gene Silencing/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Humans , Leukemia, Monocytic, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Phosphofructokinase-2/antagonists & inhibitors , Prognosis , Protein Kinase Inhibitors/pharmacology , THP-1 Cells , Transcription, Genetic/drug effects , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL