Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Clin. transl. oncol. (Print) ; 25(7): 2183-2191, jul. 2023. ilus, graf
Article En | IBECS | ID: ibc-222387

Purpose Cutaneous melanoma is an aggressive and deadly cancer resulting from malignant transformation of cells involved in skin pigmentation. Glycolysis is widely implicated in cancer progression, but its precise role in melanoma has not been extensively studied. Here, we investigated the role of the glycolysis regulator phosphofructokinase 1 platelet isoform (PFKP) in melanoma progression. Methods PFKP expression in human melanoma tissues was analyzed by immunohistochemistry. Knockdown of PFKP by siRNA and overexpression of PFKP were performed to evaluate its functions in vitro. CCK-8 assay was used to assess cell proliferation. Glycolytic activity was determined via measurement of extracellular acidification rate (ECAR), lactic acid level, and ATP content. A tumor xenograft model was used to test the function of PFKP in vivo. Results PFKP upregulation was observed in human melanoma tissues and correlated with poor patient survival. Knockdown of PFKP in human melanoma cells suppressed cell proliferation and reduced ECAR, ATP levels, and lactic acid levels, while overexpression of PFKP displayed the opposite effects. In vivo, knockdown of PFKP in melanoma cells markedly reduced tumorigenesis. Inhibitory effects on cell proliferation, glycolysis, and tumorigenesis due to PFKP knockdown were further augmented upon treatment with the glycolysis inhibitor 2-deoxy-D-glucose (2-DG). Conclusion Collectively, these results indicate that PFKP expression in melanoma cells increases proliferation and glycolytic activity in vitro and promotes tumorigenesis in vivo, suggesting that suppression of PKFP and inhibition of glycolysis may potently suppress melanoma progression (AU)


Humans , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Melanoma/genetics , Melanoma/metabolism , Adenosine Triphosphate/metabolism , Cell Proliferation , Carcinogenesis , Cell Line, Tumor , Glycolysis/genetics , Lactic Acid/metabolism
2.
Clin Transl Oncol ; 25(7): 2183-2191, 2023 Jul.
Article En | MEDLINE | ID: mdl-36792847

PURPOSE: Cutaneous melanoma is an aggressive and deadly cancer resulting from malignant transformation of cells involved in skin pigmentation. Glycolysis is widely implicated in cancer progression, but its precise role in melanoma has not been extensively studied. Here, we investigated the role of the glycolysis regulator phosphofructokinase 1 platelet isoform (PFKP) in melanoma progression. METHODS: PFKP expression in human melanoma tissues was analyzed by immunohistochemistry. Knockdown of PFKP by siRNA and overexpression of PFKP were performed to evaluate its functions in vitro. CCK-8 assay was used to assess cell proliferation. Glycolytic activity was determined via measurement of extracellular acidification rate (ECAR), lactic acid level, and ATP content. A tumor xenograft model was used to test the function of PFKP in vivo. RESULTS: PFKP upregulation was observed in human melanoma tissues and correlated with poor patient survival. Knockdown of PFKP in human melanoma cells suppressed cell proliferation and reduced ECAR, ATP levels, and lactic acid levels, while overexpression of PFKP displayed the opposite effects. In vivo, knockdown of PFKP in melanoma cells markedly reduced tumorigenesis. Inhibitory effects on cell proliferation, glycolysis, and tumorigenesis due to PFKP knockdown were further augmented upon treatment with the glycolysis inhibitor 2-deoxy-D-glucose (2-DG). CONCLUSION: Collectively, these results indicate that PFKP expression in melanoma cells increases proliferation and glycolytic activity in vitro and promotes tumorigenesis in vivo, suggesting that suppression of PKFP and inhibition of glycolysis may potently suppress melanoma progression.


Melanoma , Phosphofructokinase-1, Type C , Skin Neoplasms , Humans , Adenosine Triphosphate/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/genetics , Glycolysis/genetics , Lactic Acid/metabolism , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism
3.
Genes Genomics ; 44(12): 1509-1517, 2022 12.
Article En | MEDLINE | ID: mdl-35917090

BACKGROUND: Overexpression of PD-L1 is observed in many types of human cancer, including glioblastoma (GBM) and contributes to tumor immune evasion. In addition, GBM shows highly-activated aerobic glycolysis due to overexpression of phosphofructokinase 1 platelet isoform (PFKP), which the key enzyme in the glycolysis. However, it remains unclear whether the metabolic enzyme PFKP plays a role in the regulation of PD-L1 expression and GBM immune evasion. OBJECTIVE: We aimed to investigate the non-metabolic role of PFKP in PD-L1 expression-induced GBM immune evasion. METHODS: The mechanisms of PFKP-induced PD-L1 expression were studied by several experiments, including real-time PCR, immunoblot analysis, and ATP production. The coculture experiments using GBM cell and T cells were performed to evaluate the effect of PFKP on T cell activation. The clinical relationship between PFKP and PD-L1 was analyzed in The Cancer Genome Atlas (TCGA) database and in human GBM specimens. RESULTS: We showed that PFKP promotes EGFR activation-induced PD-L1 expression in human GBM cells. Importantly, we demonstrated that EGFR-phosphorylated PFKP Y64 plays an important role in AKT-mediated ß-catenin transactivation and subsequent PD-L1 transcriptional expression, thereby enhancing the GBM immune evasion. In addition, based on our findings, the levels of PFKP Y64 phosphorylation are positively correlated with PD-L1 expression in human GBM specimens, highlighting the clinical significance of PFKP Y64 phosphorylation in the GBM immune evasion. CONCLUSION: These findings provide new mechanistic insight into the regulation of PD-L1 expression by a non-metabolic function of PFKP on tumor cells.


Glioblastoma , Phosphofructokinase-1, Type C , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Immune Evasion , Phosphorylation , Protein Isoforms/metabolism , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism
4.
Oncol Rep ; 47(1)2022 Jan.
Article En | MEDLINE | ID: mdl-34751415

The platelet isoform of phosphofructokinase (PFKP) is one of the key enzymes in the glycolytic pathway. PFKP is highly expressed in several cancers, and it has been reported to be involved in the progression of cancer cells. However, its oncological role in breast cancer (BC) remains unclear. The present study aimed to evaluate the function of PFKP in BC cells and its expression level in patients with BC. Firstly, the mRNA and protein expression of PFKP was evaluated in BC and non­cancerous mammary cell lines. Polymerase chain reaction (PCR) array analysis was conducted to evaluate the correlation between PFKP and 84 cancer­related genes. Then, PFKP knockdown was conducted using small interfering RNA, and cell proliferation, invasiveness and migration were analyzed. Furthermore, the association between PFKP mRNA expression and clinicopathological factors was investigated in 167 patients with BC. PFKP was highly expressed in estrogen receptor­negative and human epidermal growth factor receptor 2­negative BC cell lines. PCR array analysis demonstrated that the expression level of PFKP was significantly correlated with that of transforming growth factor­ß1 and MYC proto­oncogene. PFKP knockdown significantly decreased the proliferation and invasiveness of MCF7, SK­BR­3, and MDA­MB­231 cells. Furthermore, cell migration was inhibited in SK­BR­3 and MDA­MB­231 cells. In the clinical specimens, patients with T2/T3/T4, lymph node metastasis, or stage II/III/IV exhibited higher expression of PFKP mRNA than patients with less severe disease. In conclusion, the present findings indicated that PFKP is involved in promoting tumor­progressive oncological roles in BC cells across different subtypes and is considered a possible novel therapeutic target for BC.


Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Phosphofructokinase-1, Type C/genetics , Phosphofructokinases/genetics , Adult , Aged , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged
5.
Nat Commun ; 12(1): 7035, 2021 12 02.
Article En | MEDLINE | ID: mdl-34857756

RNA editing is a feature of RNA maturation resulting in the formation of transcripts whose sequence differs from the genome template. Brain RNA editing may be altered in Alzheimer's disease (AD). Here, we analyzed data from 1,865 brain samples covering 9 brain regions from 1,074 unrelated subjects on a transcriptome-wide scale to identify inter-regional differences in RNA editing. We expand the list of known brain editing events by identifying 58,761 previously unreported events. We note that only a small proportion of these editing events are found at the protein level in our proteome-wide validation effort. We also identified the occurrence of editing events associated with AD dementia, neuropathological measures and longitudinal cognitive decline in: SYT11, MCUR1, SOD2, ORAI2, HSDL2, PFKP, and GPRC5B. Thus, we present an extended reference set of brain RNA editing events, identify a subset that are found to be expressed at the protein level, and extend the narrative of transcriptomic perturbation in AD to RNA editing.


Alzheimer Disease/genetics , ORAI2 Protein/genetics , RNA Editing , RNA/genetics , Synaptotagmins/genetics , Transcriptome , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Atlases as Topic , Brain/metabolism , Brain/pathology , Brain Chemistry , Gene Expression Profiling , Humans , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , ORAI2 Protein/metabolism , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism , RNA/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Synaptotagmins/metabolism
6.
J Clin Invest ; 131(16)2021 08 16.
Article En | MEDLINE | ID: mdl-34255748

PFKP (phosphofructokinase, platelet), the major isoform of PFK1 expressed in T cell acute lymphoblastic leukemia (T-ALL), is predominantly expressed in the cytoplasm to carry out its glycolytic function. Our study showed that PFKP is a nucleocytoplasmic shuttling protein with functional nuclear export and nuclear localization sequences (NLSs). Cyclin D3/CDK6 facilitated PFKP nuclear translocation by dimerization and by exposing the NLS of PFKP to induce the interaction between PFKP and importin 9. Nuclear PFKP stimulated the expression of C-X-C chemokine receptor type 4 (CXCR4), a chemokine receptor regulating leukemia homing/infiltration, to promote T-ALL cell invasion, which depended on the activity of c-Myc. In vivo experiments showed that nuclear PFKP promoted leukemia homing/infiltration into the bone marrow, spleen, and liver, which could be blocked with CXCR4 antagonists. Immunohistochemical staining of tissues from a clinically well-annotated cohort of T cell lymphoma/leukemia patients showed nuclear PFKP localization in invasive cancers, but not in nonmalignant T lymph node or reactive hyperplasia. The presence of nuclear PFKP in these specimens correlated with poor survival in patients with T cell malignancy, suggesting the potential utility of nuclear PFKP as a diagnostic marker.


Phosphofructokinase-1, Type C/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, CXCR4/metabolism , Active Transport, Cell Nucleus , Animals , Biomarkers, Tumor/metabolism , Cyclin-Dependent Kinase 6/metabolism , Female , Humans , Karyopherins/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Models, Molecular , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Phosphofructokinase-1, Type C/chemistry , Phosphofructokinase-1, Type C/genetics , Prognosis , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myc/metabolism , Tumor Cells, Cultured
7.
Toxicol Appl Pharmacol ; 425: 115606, 2021 08 15.
Article En | MEDLINE | ID: mdl-34087332

Triptolide (TP), a primary bioactive ingredient isolated from the traditional Chinese herbal medicine Tripterygium wilfordii Hook. F. (TWHF), has attracted great interest for its therapeutic biological activities in inflammation and autoimmune disease. However, its clinical use is limited by severe testicular toxicity, and the underlying mechanism has not been elucidated. Our preliminary evidence demonstrated that TP disrupted glucose metabolism and caused testicular toxicity. During spermatogenesis, Sertoli cells (SCs) provide lactate as an energy source to germ cells by glycolysis. The transcription factors GATA-binding protein 4 (GATA4) and specificity protein 1 (Sp1) can regulate glycolysis. Based on this evidence, we speculate that TP causes abnormal glycolysis in SCs by influencing the expression of the transcription factors GATA4 and Sp1. The mechanism of TP-induced testicular toxicity was investigated in vitro and in vivo. The data indicated that TP decreased glucose consumption, lactate production, and the mRNA levels of glycolysis-related transporters and enzymes. TP also downregulated the protein expression of the transcription factors GATA4 and Sp1, as well as the glycolytic enzyme phosphofructokinase platelet (PFKP). Phosphorylated GATA4 and nuclear GATA4 protein levels were reduced in a dose- and time-dependent manner after TP incubation. Similar effects were observed in shGata4-treated TM4 cells and BALB/c mice administered 0.4 mg/kg TP for 28 days, and glycolysis was also inhibited. Gata4 knockdown downregulated Sp1 and PFKP expression. Furthermore, the Sp1 inhibitor plicamycin inhibited PFKP protein levels in TM4 cells. In conclusion, TP inhibited GATA4-mediated glycolysis by suppressing Sp1-dependent PFKP expression in SCs and caused testicular toxicity.


Diterpenes/pharmacology , GATA4 Transcription Factor/metabolism , Glycolysis/drug effects , Phenanthrenes/pharmacology , Phosphofructokinase-1, Type C/metabolism , Sertoli Cells/drug effects , Sp1 Transcription Factor/metabolism , Animals , Cell Line , Cell Proliferation , Cell Survival/drug effects , Down-Regulation , Epoxy Compounds/pharmacology , GATA4 Transcription Factor/drug effects , GATA4 Transcription Factor/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred ICR , Phosphofructokinase-1, Type C/drug effects , Phosphofructokinase-1, Type C/genetics , Sertoli Cells/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/genetics
8.
Nucleic Acids Res ; 49(9): 4971-4988, 2021 05 21.
Article En | MEDLINE | ID: mdl-33849067

Castration-resistant prostate cancer (CRPC) is a terminal disease and the molecular underpinnings of CRPC development need to be better understood in order to improve its treatment. Here, we report that a transcription factor Yin Yang 1 (YY1) is significantly overexpressed during prostate cancer progression. Functional and cistrome studies of YY1 uncover its roles in promoting prostate oncogenesis in vitro and in vivo, as well as sustaining tumor metabolism including the Warburg effect and mitochondria respiration. Additionally, our integrated genomics and interactome profiling in prostate tumor show that YY1 and bromodomain-containing proteins (BRD2/4) co-occupy a majority of gene-regulatory elements, coactivating downstream targets. Via gene loss-of-function and rescue studies and mutagenesis of YY1-bound cis-elements, we unveil an oncogenic pathway in which YY1 directly binds and activates PFKP, a gene encoding the rate-limiting enzyme for glycolysis, significantly contributing to the YY1-enforced Warburg effect and malignant growth. Altogether, this study supports a master regulator role for YY1 in prostate tumorigenesis and reveals a YY1:BRD2/4-PFKP axis operating in advanced prostate cancer with implications for therapy.


Gene Expression Regulation, Neoplastic , Phosphofructokinase-1, Type C/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , YY1 Transcription Factor/metabolism , Animals , Carcinogenesis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Glycolysis , HEK293 Cells , Humans , Male , Mice, SCID , Phosphofructokinase-1, Type C/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Transcription Factors/metabolism , Transcriptional Activation , YY1 Transcription Factor/genetics , YY1 Transcription Factor/physiology
9.
Cell Commun Signal ; 19(1): 18, 2021 02 15.
Article En | MEDLINE | ID: mdl-33588886

BACKGROUND: Our previous studies have shown that the E3 ubiquitin ligase of HMG-CoA reductase degradation 1 (HRD1) functions as a tumor suppressor, as overexpression of HRD1 suppressed breast cancer proliferation and invasion. However, its role in breast cancer cell glucose metabolism was unclear. Here, our aim was to uncover the role and molecular mechanisms of HRD1 in regulating aerobic glycolysis in breast cancer. METHODS: The effect of HRD1 on robic glycolysis in breast cancer cells were assessed. Then the proliferation, colony formation ability, invasion and migration of breast cancer cells were evaluated. The relationship between HRD1 and PFKP was validated by Mass spectrometry analysis, immunofluorescence and co-immunoprecipitation. The level of PFKP ubiquitination was measured using ubiquitylation assay. Furthermore, the tumor growth and metastasis in mice xenografts were observed. RESULTS: We found that upregulation of HRD1 clearly decreased aerobic glycolysis, and subsequently inhibited breast cancer proliferation and invasion. Mass spectrometry analysis results revealed a large HRD1 interactome, which included PFKP (platelet isoform of phosphofructokinase), a critical enzyme involved in the Warburg Effect in breast cancer. Mechanistically, HRD1 interacted and colocalized with PFKP in the cytoplasm, targeted PFKP for ubiquitination and degradation, and ultimately reduced PFKP expression and activity in breast cancer cells. HRD1 inhibited breast cancer growth and metastasis in vivo through a PFKP-dependent way CONCLUSIONS: Our findings reveal a new regulatory role of HRD1 in Warburg effect and provide a key contributor in breast cancer metabolism. Video abstract.


Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Phosphofructokinase-1, Type C/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphate/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Movement , Cell Proliferation , Disease Progression , Female , Glucose/metabolism , Glycolysis , Humans , Lactic Acid/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Nude , Phosphofructokinase-1, Type C/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Up-Regulation
10.
Mol Cell ; 81(5): 922-939.e9, 2021 03 04.
Article En | MEDLINE | ID: mdl-33434505

R-2-hydroxyglutarate (R-2HG), a metabolite produced by mutant isocitrate dehydrogenases (IDHs), was recently reported to exhibit anti-tumor activity. However, its effect on cancer metabolism remains largely elusive. Here we show that R-2HG effectively attenuates aerobic glycolysis, a hallmark of cancer metabolism, in (R-2HG-sensitive) leukemia cells. Mechanistically, R-2HG abrogates fat-mass- and obesity-associated protein (FTO)/N6-methyladenosine (m6A)/YTH N6-methyladenosine RNA binding protein 2 (YTHDF2)-mediated post-transcriptional upregulation of phosphofructokinase platelet (PFKP) and lactate dehydrogenase B (LDHB) (two critical glycolytic genes) expression and thereby suppresses aerobic glycolysis. Knockdown of FTO, PFKP, or LDHB recapitulates R-2HG-induced glycolytic inhibition in (R-2HG-sensitive) leukemia cells, but not in normal CD34+ hematopoietic stem/progenitor cells, and inhibits leukemogenesis in vivo; conversely, their overexpression reverses R-2HG-induced effects. R-2HG also suppresses glycolysis and downregulates FTO/PFKP/LDHB expression in human primary IDH-wild-type acute myeloid leukemia (AML) cells, demonstrating the clinical relevance. Collectively, our study reveals previously unrecognized effects of R-2HG and RNA modification on aerobic glycolysis in leukemia, highlighting the therapeutic potential of targeting cancer epitranscriptomics and metabolism.


Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Antineoplastic Agents/pharmacology , Glutarates/pharmacology , Glycolysis/genetics , Lactate Dehydrogenases/genetics , Leukemia, Myeloid, Acute/drug therapy , Phosphofructokinase-1, Type C/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , HEK293 Cells , Humans , K562 Cells , Lactate Dehydrogenases/antagonists & inhibitors , Lactate Dehydrogenases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Phosphorylation/drug effects , Phosphofructokinase-1, Type C/antagonists & inhibitors , Phosphofructokinase-1, Type C/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
11.
Front Endocrinol (Lausanne) ; 12: 797025, 2021.
Article En | MEDLINE | ID: mdl-35095764

Background: Glycolysis dysfunction is an important pathogenesis of podocyte injury in diabetic kidney disease (DKD). Foot process fusion of podocytes and increased albuminuria are markers of early DKD. Moreover, cytoskeletal remodeling has been found to be involved in the foot process fusion of podocytes. However, the connections between cytoskeletal remodeling and alterations of glycolysis in podocytes in DKD have not been clarified. Methods: mRNA sequencing of glomeruli obtained from db/db and db/m mice with albuminuria was performed to analyze the expression profiling of genes in glucose metabolism. Expressions of phosphofructokinase platelet type (PFKP) in the glomeruli of DKD patients were detected. Clotrimazole (CTZ) was used to explore the renal effects of PFKP inhibition in diabetic mice. Using Pfkp siRNA or recombinant plasmid to manipulate PFKP expression, the effects of PFKP on high glucose (HG) induced podocyte damage were assessed in vitro. The levels of fructose-1,6-bisphosphate (FBP) were measured. Targeted metabolomics was performed to observe the alterations of the metabolites in glucose metabolism after HG stimulation. Furthermore, aldolase type b (Aldob) siRNA or recombinant plasmid were applied to evaluate the influence of FBP level alteration on podocytes. FBP was directly added to podocyte culture media. Db/db mice were treated with FBP to investigate its effects on their kidney. Results: mRNA sequencing showed that glycolysis enzyme genes were altered, characterized by upregulation of upstream genes (Hk1, and Pfkp) and down-regulation of downstream genes of glycolysis (Pkm, and Ldha). Moreover, the expression of PFKP was increased in glomeruli of DKD patients. The CTZ group presented more severe renal damage. In vitro, the Pfkp siRNA group and ALDOB overexpression group showed much more induced cytoskeletal remodeling in podocytes, while overexpression of PFKP and suppression of ALDOB in vitro rescued podocytes from cytoskeletal remodeling through regulation of FBP levels and inhibition of the RhoA/ROCK1 pathway. Furthermore, targeted metabolomics showed FBP level was significantly increased in HG group compared with the control group. Exogenous FBP addition reduced podocyte cytoskeletal remodeling and renal damage of db/db mice. Conclusions: These findings provide evidence that PFKP may be a potential target for podocyte injury in DN and provide a rationale for applying podocyte glycolysis enhancing agents in patients with DKD.


Cytoskeleton/metabolism , Diabetic Nephropathies/genetics , Fructosediphosphates/metabolism , Phosphofructokinase-1, Type C/genetics , Podocytes/metabolism , RNA, Messenger/metabolism , Adult , Albuminuria , Animals , Cytoskeleton/pathology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Female , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Mice , Middle Aged , Phosphofructokinase-1, Type C/metabolism , Podocytes/pathology , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
12.
Cardiovasc Res ; 117(6): 1532-1545, 2021 05 25.
Article En | MEDLINE | ID: mdl-32717063

AIMS: Pathological cardiac remodelling is characterized by cardiomyocyte (CM) hypertrophy and fibroblast activation, which can ultimately lead to maladaptive hypertrophy and heart failure (HF). Genome-wide expression analysis on heart tissue has been instrumental for the identification of molecular mechanisms at play. However, these data were based on signals derived from all cardiac cell types. Here, we aimed for a more detailed view on molecular changes driving maladaptive CM hypertrophy to aid in the development of therapies to reverse pathological remodelling. METHODS AND RESULTS: Utilizing CM-specific reporter mice exposed to pressure overload by transverse aortic banding and CM isolation by flow cytometry, we obtained gene expression profiles of hypertrophic CMs in the more immediate phase after stress, and CMs showing pathological hypertrophy. We identified subsets of genes differentially regulated and specific for either stage. Among the genes specifically up-regulated in the CMs during the maladaptive phase we found known stress markers, such as Nppb and Myh7, but additionally identified a set of genes with unknown roles in pathological hypertrophy, including the platelet isoform of phosphofructokinase (PFKP). Norepinephrine-angiotensin II treatment of cultured human CMs induced the secretion of N-terminal-pro-B-type natriuretic peptide (NT-pro-BNP) and recapitulated the up-regulation of these genes, indicating conservation of the up-regulation in failing CMs. Moreover, several genes induced during pathological hypertrophy were also found to be increased in human HF, with their expression positively correlating to the known stress markers NPPB and MYH7. Mechanistically, suppression of Pfkp in primary CMs attenuated stress-induced gene expression and hypertrophy, indicating that Pfkp is an important novel player in pathological remodelling of CMs. CONCLUSION: Using CM-specific transcriptomic analysis, we identified novel genes induced during pathological hypertrophy that are relevant for human HF, and we show that PFKP is a conserved failure-induced gene that can modulate the CM stress response.


Cardiomegaly/genetics , Gene Expression Profiling , Myocytes, Cardiac/metabolism , Transcriptome , Ventricular Remodeling/genetics , Animals , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism
13.
Int J Mol Sci ; 21(20)2020 Oct 13.
Article En | MEDLINE | ID: mdl-33066028

Neural tube closure is a critical early step in central nervous system development that requires precise control of metabolism to ensure proper cellular proliferation and differentiation. Dysregulation of glucose metabolism during pregnancy has been associated with neural tube closure defects (NTDs) in humans suggesting that the developing neuroepithelium is particularly sensitive to metabolic changes. However, it remains unclear how metabolic pathways are regulated during neurulation. Here, we used single-cell mRNA-sequencing to analyze expression of genes involved in metabolism of carbon, fats, vitamins, and antioxidants during neurulation in mice and identify a coupling of glycolysis and cellular proliferation to ensure proper neural tube closure. Using loss of miR-302 as a genetic model of cranial NTD, we identify misregulated metabolic pathways and find a significant upregulation of glycolysis genes in embryos with NTD. These findings were validated using mass spectrometry-based metabolite profiling, which identified increased glycolytic and decreased lipid metabolites, consistent with a rewiring of central carbon traffic following loss of miR-302. Predicted miR-302 targets Pfkp, Pfkfb3, and Hk1 are significantly upregulated upon NTD resulting in increased glycolytic flux, a shortened cell cycle, and increased proliferation. Our findings establish a critical role for miR-302 in coordinating the metabolic landscape of neural tube closure.


Cell Cycle , Glycolysis , MicroRNAs/metabolism , Neural Tube/metabolism , Neurulation , Animals , Cells, Cultured , Hexokinase/genetics , Hexokinase/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neural Tube/embryology , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1, Type C/metabolism , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism
14.
Mol Biol Rep ; 47(10): 8271-8272, 2020 Oct.
Article En | MEDLINE | ID: mdl-32915402

Rapid utilization of glucose is a functional marker of cancer cells, and has been exploited in the clinical diagnosis of malignancies using imaging technology. Biochemically, an increase in the rate of glycolysis, (i.e.) the process of conversion of glucose into pyruvate accelerates the net rate of glucose consumption. One of the critical determinants of glycolytic flux is the enzyme, phosphofructokinase (PFK) which converts fructose-6-phosphate into fructose 1,6, bisphosphate. PFK activity is allosterically inhibited or upregulated by cellular ATP or AMP, respectively. In a recent report of Cellular Oncology, Shen et al., have investigated one of the forms of PFK known as the platelet-type PFK (PFKP) in lung cancer. Using clinical samples as well as experimental models the authors unravel the cancer-related roles of PFKP and demonstrate that PFKP phenotype may predict the prognosis of lung cancer. In this letter, the findings are discussed in the light of recent research to expand the potential application and clinical impact of PFKP phenotype in lung cancer.


Biomarkers, Tumor/metabolism , Lung Neoplasms/enzymology , Phosphofructokinase-1, Type C/metabolism , Biomarkers, Tumor/genetics , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Phosphofructokinase-1, Type C/genetics , Prognosis
15.
Exp Cell Res ; 396(1): 112282, 2020 11 01.
Article En | MEDLINE | ID: mdl-32919954

In the present study, we have explored the prognostic value of the Phosphofructokinase Platelet-type (PFKP) expression and its therapeutic relevance in metastatic breast cancer. PFKP immunohistochemistry was performed on Invasive ductal carcinomas (IDCs; n = 87) of breast, and its association with clinicopathological parameters were evaluated. Using online meta-analysis tools, PFKP's prognostic value was investigated in overall breast cancer as well as in triple negative subtype (TNBCs). For in vitro analysis, MDA-MB-231 cells model was used in order to elucidate mechanisms behind PFKP regulated glycolysis and its impact on cancer cell physiology. Therapeutic relevance of PFKP was further evaluated using PFKP siRNA and Quercetin. PFKP protein expression was found to be positively associated with nodal invasion (p = 0.009), receptor (ER & PR) negative status (p = 0.005 & p = 0.028) and reduced overall survival in breast cancer patients (p = 0.014). In MDA-MB-231 cells, quercetin treatment impaired PFKP-LDHA signaling axis thereby inhibiting aerobic glycolysis mediated increased migration of cancer cells. Our present study demonstrates that elevated PFKP levels are associated with basal cells/TNBC subtypes and might serve as prognostic indicator for TNBC patients. Ability of quercetin to inhibit aerobic glycolysis, cell migration and clonogenic potential of malignant breast cancer cells advocates possibility of quercetin in aggressive breast cancer treatment.


Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Gene Expression Regulation, Neoplastic , L-Lactate Dehydrogenase/genetics , Phosphofructokinase-1, Type C/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Antineoplastic Agents, Phytogenic/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Glucose/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Middle Aged , Neoplasm Staging , Phosphofructokinase-1, Type C/antagonists & inhibitors , Phosphofructokinase-1, Type C/metabolism , Prognosis , Quercetin/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Survival Analysis , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
16.
Int J Mol Sci ; 20(5)2019 Mar 07.
Article En | MEDLINE | ID: mdl-30866497

Prostate cancer (PCa) is a clinically heterogeneous disease and currently, accurate diagnostic and prognostic molecular biomarkers are lacking. This study aimed to identify novel DNA hypermethylation markers for PCa with future potential for blood-based testing. Accordingly, to search for genes specifically hypermethylated in PCa tissue samples and not in blood cells or other cancer tissue types, we performed a systematic analysis of genome-wide DNA methylation data (Infinium 450K array) available in the Marmal-aid database for 4072 malignant/normal tissue samples of various types. We identified eight top candidate markers (cg12799885, DOCK2, FBXO30, GRASP, HIF3A, MOB3B, PFKP, and TPM4) that were specifically hypermethylated in PCa tissue samples and hypomethylated in other benign and malignant tissue types, including in peripheral blood cells. Potential as diagnostic and prognostic biomarkers was further assessed by the quantitative methylation specific PCR (qMSP) analysis of 37 nonmalignant and 197 PCa tissue samples from an independent population. Here, all eight hypermethylated candidates showed high sensitivity (75⁻94%) and specificity (84⁻100%) for PCa. Furthermore, DOCK2, GRASP, HIF3A and PKFP hypermethylation was significantly associated with biochemical recurrence (BCR) after radical prostatectomy (RP; 197 patients), independent of the routine clinicopathological variables. DOCK2 is the most promising single candidate marker (hazard ratio (HR) (95% confidence interval (CI)): 1.96 (1.24⁻3.10), adjusted p = 0.016; multivariate cox regression). Further validation studies are warranted and should investigate the potential value of these hypermethylation candidate markers for blood-based testing also.


Basic Helix-Loop-Helix Transcription Factors/genetics , Carrier Proteins/genetics , DNA Methylation , Guanine Nucleotide Exchange Factors/genetics , Membrane Proteins/genetics , Phosphofructokinase-1, Type C/genetics , Prostatic Neoplasms/diagnosis , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins , Biomarkers, Tumor/genetics , Epigenesis, Genetic , GTPase-Activating Proteins , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Repressor Proteins , Sensitivity and Specificity , Survival Analysis
17.
Nat Commun ; 10(1): 809, 2019 02 18.
Article En | MEDLINE | ID: mdl-30778058

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers. It thrives in a nutrient-poor environment; however, the mechanisms by which PDAC cells undergo metabolic reprogramming to adapt to metabolic stress are still poorly understood. Here, we show that microRNA-135 is significantly increased in PDAC patient samples compared to adjacent normal tissue. Mechanistically, miR-135 accumulates specifically in response to glutamine deprivation and requires ROS-dependent activation of mutant p53, which directly promotes miR-135 expression. Functionally, we found miR-135 targets phosphofructokinase-1 (PFK1) and inhibits aerobic glycolysis, thereby promoting the utilization of glucose to support the tricarboxylic acid (TCA) cycle. Consistently, miR-135 silencing sensitizes PDAC cells to glutamine deprivation and represses tumor growth in vivo. Together, these results identify a mechanism used by PDAC cells to survive the nutrient-poor tumor microenvironment, and also provide insight regarding the role of mutant p53 and miRNA in pancreatic cancer cell adaptation to metabolic stresses.


Carcinoma, Pancreatic Ductal/genetics , Glycolysis/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Phosphofructokinase-1, Type C/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Glutamine/genetics , Glutamine/metabolism , Humans , Male , Mice, Nude , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphofructokinase-1, Type C/metabolism , Stress, Physiological/genetics , Xenograft Model Antitumor Assays
18.
Exp Cell Res ; 370(1): 46-57, 2018 09 01.
Article En | MEDLINE | ID: mdl-29894707

The tumor starvation microenvironment plays a pivotal role in the malignant progression of cancer, which is closely related to autophagy, glycolysis, and epithelial mesenchymal transition (EMT). Nevertheless, the underlying mechanisms of the starvation-mediated malignant phenotype are still not well documented. In this study, we aimed to investigate the effect of starvation on glycolysis, autophagy, and EMT in OSCC and to further elucidate the key metabolic modulator. The results showed that starvation can induce autophagy, EMT, and enhanced glycolysis in OSCC cells. We determined that the expression of the key glycolytic enzyme phosphofructokinase-platelet (PFKP) obviously increased under starvation conditions and that PFKP knockdown inhibited starvation-mediated glycolysis, autophagy and EMT in OSCC cells. Moreover, we confirmed that PFKP knockdown inhibited OSCC xenograft growth in vivo. In addition, PFKP expression was significantly increased in OSCC patients and its upregulation was associated with the presence of tumor pathological differentiation and lymph node metastasis. Taken together, our findings demonstrate that PFKP is necessary for starvation-mediated autophagy, glycolysis, and EMT, thereby promoting the malignant progression of OSCC.


Autophagy/genetics , Carcinoma, Squamous Cell/genetics , Epithelial-Mesenchymal Transition/genetics , Glycolysis/genetics , Mouth Neoplasms/genetics , Phosphofructokinase-1, Type C/genetics , Starvation/genetics , Animals , Carcinoma, Squamous Cell/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/pathology , Starvation/pathology , Tumor Microenvironment/genetics , Up-Regulation/genetics
19.
Mol Cell ; 70(2): 197-210.e7, 2018 04 19.
Article En | MEDLINE | ID: mdl-29677490

EGFR activates phosphatidylinositide 3-kinase (PI3K), but the mechanism underlying this activation is not completely understood. We demonstrated here that EGFR activation resulted in lysine acetyltransferase 5 (KAT5)-mediated K395 acetylation of the platelet isoform of phosphofructokinase 1 (PFKP) and subsequent translocation of PFKP to the plasma membrane, where the PFKP was phosphorylated at Y64 by EGFR. Phosphorylated PFKP binds to the N-terminal SH2 domain of p85α, which is distinct from binding of Gab1 to the C-terminal SH2 domain of p85α, and recruited p85α to the plasma membrane resulting in PI3K activation. PI3K-dependent AKT activation results in enhanced phosphofructokinase 2 (PFK2) phosphorylation and production of fructose-2,6-bisphosphate, which in turn promotes PFK1 activation. PFKP Y64 phosphorylation-enhanced PI3K/AKT-dependent PFK1 activation and GLUT1 expression promoted the Warburg effect, tumor cell proliferation, and brain tumorigenesis. These findings underscore the instrumental role of PFKP in PI3K activation and enhanced glycolysis through PI3K/AKT-dependent positive-feedback regulation.


Brain Neoplasms/enzymology , Glioblastoma/enzymology , Glycolysis , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-1, Type C/metabolism , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Feedback, Physiological , Fructosediphosphates/metabolism , Glioblastoma/genetics , Glioblastoma/pathology , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , src Homology Domains
20.
Nat Commun ; 8: 14374, 2017 02 08.
Article En | MEDLINE | ID: mdl-28176759

Dynamic regulation of glucose flux between aerobic glycolysis and the pentose phosphate pathway (PPP) during epithelial-mesenchymal transition (EMT) is not well-understood. Here we show that Snail (SNAI1), a key transcriptional repressor of EMT, regulates glucose flux toward PPP, allowing cancer cell survival under metabolic stress. Mechanistically, Snail regulates glycolytic activity via repression of phosphofructokinase, platelet (PFKP), a major isoform of cancer-specific phosphofructokinase-1 (PFK-1), an enzyme involving the first rate-limiting step of glycolysis. The suppression of PFKP switches the glucose flux towards PPP, generating NADPH with increased metabolites of oxidative PPP. Functionally, dynamic regulation of PFKP significantly potentiates cancer cell survival under metabolic stress and increases metastatic capacities in vivo. Further, knockdown of PFKP rescues metabolic reprogramming and cell death induced by loss of Snail. Thus, the Snail-PFKP axis plays an important role in cancer cell survival via regulation of glucose flux between glycolysis and PPP.


Glucose/metabolism , Neoplasms/pathology , Oxidative Stress/genetics , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1/genetics , Snail Family Transcription Factors/metabolism , Cell Survival/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycolysis , Humans , NADP/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Pentose Phosphate Pathway/genetics , Phosphofructokinase-1/metabolism , Phosphofructokinase-1, Type C/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors/genetics
...