Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 22(1): 51, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233839

RESUMO

The dynamic changes of RNA N6-methyladenosine (m6A) during cancer progression participate in various cellular processes. However, less is known about a possible direct connection between upstream regulator and m6A modification, and therefore affects oncogenic progression. Here, we have identified that a key enzyme in N4-acetylcytidine (ac4C) acetylation NAT10 is highly expressed in human osteosarcoma tissues, and its knockdown enhanced m6A contents and significantly suppressed osteosarcoma cell growth, migration and invasion. Further results revealed that NAT10 silence inhibits mRNA stability and translation of m6A reader protein YTHDC1, and displayed an increase in glucose uptake, a decrease in lactate production and pyruvate content. YTHDC1 recognizes differential m6A sites on key enzymes of glycolysis phosphofructokinase (PFKM) and lactate dehydrogenase A (LDHA) mRNAs, which suppress glycolysis pathway by increasing mRNA stability of them in an m6A methylation-dependent manner. YTHDC1 partially abrogated the inhibitory effect caused by NAT10 knockdown in tumor models in vivo, lentiviral overexpression of YTHDC1 partially restored the reduced stability of YTHDC1 caused by lentiviral depleting NAT10 at the cellular level. Altogether, we found ac4C driven RNA m6A modification can positively regulate the glycolysis of cancer cells and reveals a previously unrecognized signaling axis of NAT10/ac4C-YTHDC1/m6A-LDHA/PFKM in osteosarcoma. Video Abstract.


Assuntos
Citidina/análogos & derivados , Osteossarcoma , Fosfofrutoquinases , Humanos , Lactato Desidrogenase 5/metabolismo , Fosfofrutoquinases/metabolismo , Acetilação , RNA/metabolismo , Glicólise/genética , Osteossarcoma/patologia , Fosfofrutoquinase-1 Muscular/metabolismo , Fatores de Processamento de RNA/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Acetiltransferases N-Terminal/metabolismo
2.
Cell Death Dis ; 13(10): 876, 2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36253358

RESUMO

Renal fibrosis is a common pathological feature and outcome of almost all chronic kidney diseases, and it is characterized by metabolic reprogramming toward aerobic glycolysis. Mesenchymal stem cell-derived exosomes (MSC-Exos) have been proposed as a promising therapeutic approach for renal fibrosis. In this study, we investigated the effect of MSC-Exos on glycolysis and the underlying mechanisms. We demonstrated that MSC-Exos significantly ameliorated unilateral ureter obstruction (UUO)-induced renal fibrosis by inhibiting glycolysis in tubular epithelial cells (TECs). miRNA sequencing showed that miR-21a-5p was highly enriched in MSC-Exos. Mechanistically, miR-21a-5p repressed the expression of phosphofructokinase muscle isoform (PFKM), a rate-limiting enzyme of glycolysis, thereby attenuating glycolysis in TECs. Additionally, knockdown of miR-21a-5p abolished the renoprotective effect of MSC-Exos. These findings revealed a novel role for MSC-Exos in the suppression of glycolysis, providing a new insight into the treatment of renal fibrosis.


Assuntos
Exossomos , Nefropatias , Células-Tronco Mesenquimais , MicroRNAs , Fosfofrutoquinase-1 Muscular , Humanos , Exossomos/genética , Exossomos/metabolismo , Fibrose , Glicólise/genética , Nefropatias/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Músculos/metabolismo , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Isoformas de Proteínas/metabolismo
3.
Genes (Basel) ; 13(3)2022 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-35328104

RESUMO

The reprogramming of energy metabolism is one of the hallmarks of cancer and is crucial for tumor progression. Altered aerobic glycolysis is a well-known characteristic of cancer cell metabolism. In the present study, the expression profiles of key metabolic genes (HK2, PFKM, and PKM2) were assessed in the breast cancer cohort of Pakistan using quantitative polymerase chain reaction (qPCR) and IHC. Expression patterns were correlated with molecular subtypes and clinical parameters in the patients. A significant upregulation of key glycolytic genes was observed in tumor samples in comparison to their adjacent controls (p < 0.0001). The expression of the studied glycolytic genes was significantly increased in late clinical stages, positive nodal involvement, and distant metastasis (p < 0.05). HK2 and PKM2 were found to be upregulated in luminal B, whereas PFKM was overexpressed in the luminal A subtype of breast cancer. The genes were positively correlated with the proliferation marker Ki67 (p < 0.001). Moreover, moderate positive linear correlations between HK2 and PKM2 (r = 0.476), HK2 and PFKM (r = 0.473), and PKM2 and PFKM (r = 0.501) were also observed (p < 0.01). These findings validate that the key regulatory genes in glycolysis can serve as potential biomarkers and/or molecular targets for breast cancer management. However, the clinical significance of these molecules needs to be further validated through in vitro and in vivo experiments.


Assuntos
Neoplasias da Mama , Idade de Início , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte , Feminino , Glicólise/genética , Hexoquinase , Humanos , Proteínas de Membrana , Metástase Neoplásica , Paquistão , Fosfofrutoquinase-1 Muscular/metabolismo , Hormônios Tireóideos , Proteínas de Ligação a Hormônio da Tireoide
4.
Cell Death Dis ; 12(4): 408, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859186

RESUMO

One of the malignant transformation hallmarks is metabolism reprogramming, which plays a critical role in the biosynthetic needs of unchecked proliferation, abrogating cell death programs, and immunologic escape. However, the mechanism of the metabolic switch is not fully understood. Here, we found that the S-nitrosoproteomic profile of endogenous nitrogen oxide in ovarian cancer cells targeted multiple components in metabolism processes. Phosphofructokinase (PFKM), one of the most important regulatory enzymes of glycolysis, was S-nitrosylated by nitric oxide synthase NOS1 at Cys351. S-nitrosylation at Cys351 stabilized the tetramer of PFKM, leading to resist negative feedback of downstream metabolic intermediates. The PFKM-C351S mutation decreased the proliferation rate of cultured cancer cells, and reduced tumor growth and metastasis in the mouse xenograft model. These findings indicated that S-nitrosylation at Cys351 of PFKM by NOS1 contributes to the metabolic reprogramming of ovarian cancer cells, highlighting a critical role of endogenous nitrogen oxide on metabolism regulations in tumor progression.


Assuntos
Carcinoma Epitelial do Ovário/genética , Glicólise/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Animais , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
5.
Mol Cell ; 81(9): 1905-1919.e12, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33852893

RESUMO

Oxidative phosphorylation (OXPHOS) and glycolysis are the two major pathways for ATP production. The reliance on each varies across tissues and cell states, and can influence susceptibility to disease. At present, the full set of molecular mechanisms governing the relative expression and balance of these two pathways is unknown. Here, we focus on genes whose loss leads to an increase in OXPHOS activity. Unexpectedly, this class of genes is enriched for components of the pre-mRNA splicing machinery, in particular for subunits of the U1 snRNP. Among them, we show that LUC7L2 represses OXPHOS and promotes glycolysis by multiple mechanisms, including (1) splicing of the glycolytic enzyme PFKM to suppress glycogen synthesis, (2) splicing of the cystine/glutamate antiporter SLC7A11 (xCT) to suppress glutamate oxidation, and (3) secondary repression of mitochondrial respiratory supercomplex formation. Our results connect LUC7L2 expression and, more generally, the U1 snRNP to cellular energy metabolism.


Assuntos
Glicólise , Fosforilação Oxidativa , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Ácido Glutâmico/metabolismo , Glicogênio/metabolismo , Glicólise/genética , Células HEK293 , Células HeLa , Humanos , Células K562 , Mitocôndrias/genética , Mitocôndrias/metabolismo , Oxirredução , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Precursores de RNA/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Ribonucleoproteína Nuclear Pequena U1/genética
6.
Genomics ; 113(1 Pt 1): 135-141, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33279650

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is a malignant tumor of the upper aerodigestive tract. The loss and gain of miRNA function promote cancer development through various mechanisms. RNA sequencing (RNA-seq) and miRNAs sequencing data from the Cancer Genome Atlas (TCGA) was used to show the dysfunctional miRNAs microenvironment and to provide useful biomarkers for miRNAs therapy. Seven miRNAs were found to be independent prognostic factors of HNSCC patients in the training cohort. A total of 60 target genes for these miRNAs were predicted. Nine target genes (CDCA4, CXCL14, FLNC, KLF7, NBEAL2, P4HA1, PFKM, PFN2 and SEPPINE1) were correlated with patient's overall survival (OS) outcomes. We identified novel miRNAs markers for the prognosis of head and neck squamous cell carcinoma.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Neoplasias de Cabeça e Pescoço/genética , MicroRNAs/genética , Idoso , Biomarcadores Tumorais/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Feminino , Filaminas/genética , Filaminas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Profilinas/genética , Profilinas/metabolismo
7.
Biochem Biophys Res Commun ; 530(1): 67-74, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32828317

RESUMO

Phosphofructokinase-M (PFKM) is a key enzyme in glycolysis. The expression and activity of PFKM is closely related to the occurrence and development of malignant tumors, but its role in the regulation of renal cell carcinoma (RCC) is still unknown. We found that the expression of PFKM was lower in RCC tumor tissue than in adjacent normal tissues, and that low expression of PFKM was related to the poor overall survival of RCC patients. In addition, our results showed that FOXO3 mediated PFKM inhibited the growth, migration and invasion of RCC cells, suggesting that PFKM is a protective factor for RCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Proteína Forkhead Box O3/metabolismo , Neoplasias Renais/metabolismo , Fosfofrutoquinase-1 Muscular/metabolismo , Transdução de Sinais , Carcinoma de Células Renais/diagnóstico , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteína Forkhead Box O3/análise , Humanos , Neoplasias Renais/diagnóstico , Neoplasias Renais/patologia , Fosfofrutoquinase-1 Muscular/análise , Prognóstico
8.
Proc Natl Acad Sci U S A ; 117(12): 6726-6732, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32156725

RESUMO

The extrahypothalamic growth hormone-releasing hormone (GHRH) and its cognate receptors (GHRH-Rs) and splice variants are expressed in a variety of cancers. It has been shown that the pituitary type of GHRH-R (pGHRH-R) mediates the inhibition of tumor growth induced by GHRH-R antagonists. However, GHRH-R antagonists can also suppress some cancers that do not express pGHRH-R, yet the underlying mechanisms have not been determined. Here, using human esophageal squamous cell carcinoma (ESCC) as a model, we were able to reveal that SV1, a known splice variant of GHRH-R, is responsible for the inhibition induced by GHRH-R antagonist MIA-602. We demonstrated that GHRH-R splice variant 1 (SV1) is a hypoxia-driven promoter of tumor progression. Hypoxia-elevated SV1 activates a key glycolytic enzyme, muscle-type phosphofructokinase (PFKM), through the nuclear factor kappa B (NF-κB) pathway, which enhances glycolytic metabolism and promotes progression of ESCC. The malignant actions induced by the SV1-NF-κB-PFKM pathway could be reversed by MIA-602. Altogether, our studies demonstrate a mechanism by which GHRH-R antagonists target SV1. Our findings suggest that SV1 is a hypoxia-induced oncogenic promoter which can be an alternative target of GHRH-R antagonists.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação Neoplásica da Expressão Gênica , Receptores LHRH/genética , Sermorelina/análogos & derivados , Processamento Alternativo , Animais , Apoptose , Proliferação de Células , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Glicólise , Humanos , Camundongos , Camundongos Nus , NF-kappa B/genética , NF-kappa B/metabolismo , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Receptores LHRH/antagonistas & inibidores , Sermorelina/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Phys Biol ; 16(6): 066007, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31469100

RESUMO

The glycolytic enzyme pyruvate kinase M2 (PKM2) exists in both catalytically inactive dimeric and active tetrameric forms. In cancer cells, PKM2 dimer predominance contributes to tumor growth by triggering glycolytic reprogramming. However, the mechanism that promotes PKM2 dimer predominance over tetramer in cancer cells remains elusive. Here, we show that pulsatile phosphofructokinase (PFK-1) activity results in PKM2 dimer predominance. Mathematical simulations predict that pulsatile PFK-1 activity prevents the formation of PKM2 tetramer even under high levels of fructose-1,6-bisphosphate (FBP), a PKM2 tetramer-promoting metabolite produced by PFK-1. We experimentally confirm these predictions at the single-molecule level by providing evidence for pulsatile PFK-1 activity-induced synchronized dissociation of PKM2 tetramers and the subsequent accumulation of PKM2 dimers under high levels of FBP in HeLa cells. Moreover, we show that pulsatile PFK-1 activity-induced PKM2 dimer predominance also controls cell proliferation. Thus, our study reveals the significance of pulsatile PFK-1 activity in cancer cell metabolism.


Assuntos
Proteínas de Transporte/genética , Frutosedifosfatos/metabolismo , Glicólise , Proteínas de Membrana/genética , Fosfofrutoquinase-1 Muscular/genética , Hormônios Tireóideos/genética , Proteínas de Transporte/metabolismo , Reprogramação Celular , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Fosfofrutoquinase-1 Muscular/metabolismo , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
10.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 602-607, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29563071

RESUMO

PURPOSE: Cancer cells consume more glucose than normal human cells and convert most glucose into lactate. It has been proposed that deregulated glycolysis is triggered by the posttranslational modification of 85 kDa muscle-type 6-phosphofructo-1-kinase (PFK-M) which is cleaved by a specific protease to form shorter, highly active, feedback-inhibition-resistant PFK-M fragments. PRINCIPAL RESULTS: To find the protease involved in PFK-M modification, analyses of the protease target sites on the human PFK-M enzyme yielding 45-47 kDa fragments were performed in silico. The results suggested that an enzyme in the kallikrein (KLK) family may be involved. Kallikreins can be self-activated in the cytosol and are often overexpressed in cancer cells. After incubating the internally quenched FRET peptide with a sequence characteristic of the target site, along with the active KLK6, the cleavage of the peptide was observed. The ability of KLK6 to cleave native PFK-M and form highly active citrate-resistant 45 kDa fragments was further confirmed by enzymatic tests and SDS-PAGE. A role of KLK6 in the posttranslational modification of native PFK-M was ultimately confirmed in vivo. A yeast strain that encoded native human PFK-M as the only PFK1 enzyme was additionally transformed with proKLK6 or KLK6 genes under the control of an inducible promoter. The transformants growth rate was found to increase after the induction of proKLK6 gene expression as compared to the strain with the native PFK-M enzyme. CONCLUSION: KLK6 may be the key protease involved in the modification of PFK-M and trigger deregulated glycolytic flux in cancer cells.


Assuntos
Calicreínas/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosfofrutoquinase-1 Muscular/metabolismo , Processamento de Proteína Pós-Traducional , Eletroforese em Gel de Poliacrilamida , Transferência Ressonante de Energia de Fluorescência , Humanos , Calicreínas/genética , Cinética , Fragmentos de Peptídeos/genética , Fosfofrutoquinase-1 Muscular/genética , Proteólise , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Especificidade por Substrato
11.
Biosci Biotechnol Biochem ; 81(8): 1478-1483, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28508704

RESUMO

Glucose metabolism is a basic biological process that shows substantial variation within and between species. Using pig as a model organism, we investigated differences in glucose metabolic genes in seven tissues from domesticated pigs (Rongchang pig and Tibetan pig, meanwhile, the Tibetan pig just as a special case of the domesticated pig under plateau condition) and wild boar. We found large differences in the expression of genes involved in multiple aspects of glucose metabolism, including genes associated with glucose transport, gluconeogenesis, and glycolysis. In addition, we identified microRNAs (miRNAs) that may be involved in the divergence of glucose metabolism in pig. A combined analysis of mRNA and miRNA expression indicated that some miRNA:mRNA pairs showed ab facto function in it. Our results provide a valuable resource for further determination of miRNA regulatory roles in pig glucose metabolism and reveal the divergence of glucose metabolism in pigs under domestication.


Assuntos
Regulação da Expressão Gênica , Glucose/metabolismo , Músculo Esquelético/metabolismo , Sus scrofa/genética , Suínos/genética , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Transporte Biológico , Domesticação , Perfilação da Expressão Gênica , Gluconeogênese/genética , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Glicólise/genética , Hexoquinase/genética , Hexoquinase/metabolismo , Lactato Desidrogenases/genética , Lactato Desidrogenases/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Especificidade de Órgãos , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Especificidade da Espécie , Sus scrofa/metabolismo , Suínos/metabolismo
12.
Am J Physiol Cell Physiol ; 304(2): C180-93, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23114964

RESUMO

The hypothesis was tested that the variation of in vivo glycolytic flux with contraction frequency in skeletal muscle can be qualitatively and quantitatively explained by calcium-calmodulin activation of phosphofructokinase (PFK-1). Ischemic rat tibialis anterior muscle was electrically stimulated at frequencies between 0 and 80 Hz to covary the ATP turnover rate and calcium concentration in the tissue. Estimates of in vivo glycolytic rates and cellular free energetic states were derived from dynamic changes in intramuscular pH and phosphocreatine content, respectively, determined by phosphorus magnetic resonance spectroscopy ((31)P-MRS). Computational modeling was applied to relate these empirical observations to understanding of the biochemistry of muscle glycolysis. Hereto, the kinetic model of PFK activity in a previously reported mathematical model of the glycolytic pathway (Vinnakota KC, Rusk J, Palmer L, Shankland E, Kushmerick MJ. J Physiol 588: 1961-1983, 2010) was adapted to contain a calcium-calmodulin binding sensitivity. The two main results were introduction of regulation of PFK-1 activity by binding of a calcium-calmodulin complex in combination with activation by increased concentrations of AMP and ADP was essential to qualitatively and quantitatively explain the experimental observations. Secondly, the model predicted that shutdown of glycolytic ATP production flux in muscle postexercise may lag behind deactivation of PFK-1 (timescales: 5-10 s vs. 100-200 ms, respectively) as a result of accumulation of glycolytic intermediates downstream of PFK during contractions.


Assuntos
Glicólise/fisiologia , Músculo Esquelético/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/análise , Cálcio/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Simulação por Computador , Concentração de Íons de Hidrogênio , Isquemia/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Masculino , Modelos Biológicos , Contração Muscular/fisiologia , Fosfocreatina/análise , Fosfocreatina/metabolismo , Fosfofrutoquinase-1 Muscular/química , Fosfofrutoquinase-1 Muscular/metabolismo , Condicionamento Físico Animal/fisiologia , Ratos , Ratos Wistar
13.
FASEB J ; 26(11): 4710-21, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22767230

RESUMO

Glycolysis is the initial step of glucose catabolism and is up-regulated in cancer cells (the Warburg Effect). Such shifts toward a glycolytic phenotype have not been explored widely in other biological systems, and the molecular mechanisms underlying the shifts remain unknown. With proteomics, we observed increased glycolysis in disused human diaphragm muscle. In disused muscle, lung cancer, and H(2)O(2)-treated myotubes, we show up-regulation of the rate-limiting glycolytic enzyme muscle-type phosphofructokinase (PFKm, >2 fold, P<0.05) and accumulation of lactate (>150%, P<0.05). Using microRNA profiling, we identify miR-320a as a regulator of PFKm expression. Reduced miR-320a levels (to ∼50% of control, P<0.05) are associated with the increased PFKm in each of these diverse systems. Manipulation of miR-320a levels both in vitro and in vivo alters PFKm and lactate levels in the expected directions. Further, miR-320a appears to regulate oxidative stress-induced PFKm expression, and reduced miR-320a allows greater induction of glycolysis in response to H(2)O(2) treatment. We show that this microRNA-mediated regulation occurs through PFKm's 3' untranslated region and that Ets proteins are involved in the regulation of PFKm via miR-320a. These findings suggest that oxidative stress-responsive microRNA-320a may regulate glycolysis broadly within nature.


Assuntos
Glicólise/fisiologia , MicroRNAs/metabolismo , Estresse Oxidativo/fisiologia , Adenocarcinoma/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Clonagem Molecular , DNA Complementar/genética , Regulação da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , MicroRNAs/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Reação em Cadeia da Polimerase , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
J Biol Chem ; 287(21): 17546-17553, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22474333

RESUMO

6-Phosphofructokinases (Pfk) are homo- and heterooligomeric, allosteric enzymes that catalyze one of the rate-limiting steps of the glycolysis: the phosphorylation of fructose 6-phosphate at position 1. Pfk activity is modulated by a number of regulators including adenine nucleotides. Recent crystal structures from eukaryotic Pfk revealed several adenine nucleotide binding sites. Herein, we determined the functional relevance of two adenine nucleotide binding sites through site-directed mutagenesis and enzyme kinetic studies. Subsequent characterization of Pfk mutants allowed the identification of the activating (AMP, ADP) and inhibitory (ATP, ADP) allosteric binding sites. Mutation of one binding site reciprocally influenced the allosteric regulation through nucleotides interacting with the other binding site. Such reciprocal linkage between the activating and inhibitory binding sites is in agreement with current models of allosteric enzyme regulation. Because the allosteric nucleotide binding sites in eukaryotic Pfk did not evolve from prokaryotic ancestors, reciprocal linkage of functionally opposed allosteric binding sites must have developed independently in prokaryotic and eukaryotic Pfk (convergent evolution).


Assuntos
Difosfato de Adenosina/química , Trifosfato de Adenosina/química , Fosfofrutoquinase-1 Muscular/química , Difosfato de Adenosina/genética , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/fisiologia , Sítios de Ligação , Evolução Molecular , Humanos , Mutação , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo
15.
J Cell Biochem ; 113(5): 1692-703, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22213537

RESUMO

Kinetic analysis of PFK-1 from rodent AS-30D, and human HeLa and MCF-7 carcinomas revealed sigmoidal [fructose 6-phosphate, Fru6P]-rate curves with different V(m) values when varying the allosteric activator fructose 2,6 bisphosphate (Fru2,6BP), AMP, Pi, NH(4)(+), or K(+). The rate equation that accurately predicted this behavior was the exclusive ligand binding concerted transition model together with non-essential hyperbolic activation. PFK-1 from rat liver and heart also exhibited the mixed cooperative-hyperbolic kinetic behavior regarding activators. Lowering pH induced decreased affinity for Fru6P, Fru2,6BP, citrate, and ATP (as inhibitor); as well as decreased V(m) and increased content of inactive (T) enzyme forms. High K(+) prompted increased (Fru6P) or decreased (activators) affinities; increased V(m); and increased content of active (R) enzyme forms. mRNA expression analysis and nucleotide sequencing showed that the three PFK-1 isoforms L, M, and C are transcribed in the three carcinomas. However, proteomic analysis indicated the predominant expression of L in liver, of M in heart and MCF-7 cells, of L>M in AS-30D cells, and of C in HeLa cells. PFK-1M showed the highest affinities for F6P and citrate and the lowest for ATP (substrate) and F2,6BP; PFK-1L showed the lowest affinity for F6P and the highest for F2,6BP; and PFK-1C exhibited the highest affinity for ATP (substrate) and the lowest for citrate. Thus, the present work documents the kinetic signature of each PFK-1 isoform, and facilitates the understanding of why this enzyme exerts significant or negligible glycolysis flux-control in normal or cancer cells, respectively, and how it regulates the onset of the Pasteur effect.


Assuntos
Neoplasias/enzimologia , Neoplasias/genética , Fosfofrutoquinase-1/metabolismo , Animais , Sequência de Bases , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , DNA Complementar/genética , Ativação Enzimática , Feminino , Células HeLa , Humanos , Cinética , Fígado/enzimologia , Neoplasias Hepáticas Experimentais/enzimologia , Neoplasias Hepáticas Experimentais/genética , Miocárdio/enzimologia , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-1 Hepática/genética , Fosfofrutoquinase-1 Hepática/metabolismo , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Fosfofrutoquinase-1 Tipo C/genética , Fosfofrutoquinase-1 Tipo C/metabolismo , Polimorfismo Genético , Ratos , Ratos Wistar , Especificidade por Substrato , Neoplasias do Colo do Útero/enzimologia , Neoplasias do Colo do Útero/genética
16.
PLoS One ; 6(5): e19645, 2011 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-21573193

RESUMO

BACKGROUND: Human cancers consume larger amounts of glucose compared to normal tissues with most being converted and excreted as lactate despite abundant oxygen availability (Warburg effect). The underlying higher rate of glycolysis is therefore at the root of tumor formation and growth. Normal control of glycolytic allosteric enzymes appears impaired in tumors; however, the phenomenon has not been fully resolved. METHODOLOGY/PRINCIPAL FINDINGS: In the present paper, we show evidence that the native 85-kDa 6-phosphofructo-1-kinase (PFK1), a key regulatory enzyme of glycolysis that is normally under the control of feedback inhibition, undergoes posttranslational modification. After proteolytic cleavage of the C-terminal portion of the enzyme, an active, shorter 47-kDa fragment was formed that was insensitive to citrate and ATP inhibition. In tumorigenic cell lines, only the short fragments but not the native 85-kDa PFK1 were detected by immunoblotting. Similar fragments were detected also in a tumor tissue that developed in mice after the subcutaneous infection with tumorigenic B16-F10 cells. Based on limited proteolytic digestion of the rabbit muscle PFK-M, an active citrate inhibition-resistant shorter form was obtained, indicating that a single posttranslational modification step was possible. The exact molecular masses of the active shorter PFK1 fragments were determined by inserting the truncated genes constructed from human muscle PFK1 cDNA into a pfk null E. coli strain. Two E. coli transformants encoding for the modified PFK1s of 45,551 Da and 47,835 Da grew in glucose medium. The insertion of modified truncated human pfkM genes also stimulated glucose consumption and lactate excretion in stable transfectants of non-tumorigenic human HEK cell, suggesting the important role of shorter PFK1 fragments in enhancing glycolytic flux. CONCLUSIONS/SIGNIFICANCE: Posttranslational modification of PFK1 enzyme might be the pivotal factor of deregulated glycolytic flux in tumors that in combination with altered signaling mechanisms essentially supports fast proliferation of cancer cells.


Assuntos
Neoplasias/enzimologia , Neoplasias/metabolismo , Fosfofrutoquinase-1 Muscular/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Western Blotting , Linhagem Celular Tumoral , DNA Complementar/genética , Endopeptidase K/metabolismo , Escherichia coli/citologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Frutosedifosfatos/farmacologia , Glucose/metabolismo , Células HEK293 , Humanos , Ácido Láctico/biossíntese , Camundongos , Metástase Neoplásica , Neoplasias/patologia , Fragmentos de Peptídeos/metabolismo , Fosfofrutoquinase-1 Muscular/antagonistas & inibidores , Fosfofrutoquinase-1 Muscular/química , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Coelhos , Proteínas Recombinantes/metabolismo , Análise de Sequência de Proteína , Transfecção , Transformação Genética/efeitos dos fármacos
17.
Meat Sci ; 89(2): 217-20, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21592677

RESUMO

Longissimus muscle samples from the pig genotypes Duroc (Du), Pietrain (MHS homozygote negative (PiNN), positive (PiPP)) and a Duroc-Pietrain crossbreed (DuPi) were analyzed. The PiPP samples showed a faster pH drop and higher electrical conductivity, drip loss and lightness values. Before slaughter the concentrations of the adenine nucleotides were comparable between the genotypes, but 40 min after slaughter (p.m.) the ATP concentrations decreased and IMP increased, to a higher extent in the PiPP pigs. The nucleotide values of the 12 h p.m. samples were again comparable. Activities of glycogen phosporylase (GP), phosphofructokinase (PFK) and lactate dehydrogenase (LDH) were nearly similar before slaughter. Forty minutes after slaughter the LDH activities increased in all pigs and the PFK activities in all genotypes but not in the PiPP. GP results were rather inconsistent indicating an earlier activation of this enzyme. The study showed that the reduced meat quality in the PiPP pigs is accompanied with rapid ATP degradation and accelerated enzyme activation.


Assuntos
Nucleotídeos de Adenina/análise , Glicogênio Fosforilase/metabolismo , L-Lactato Desidrogenase/metabolismo , Carne , Músculo Esquelético/enzimologia , Fosfofrutoquinase-1 Muscular/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Condutividade Elétrica , Genótipo , Glicogênio Fosforilase/análise , L-Lactato Desidrogenase/análise , Mutação , Fosfofrutoquinase-1 Muscular/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Suínos/classificação , Suínos/genética , Suínos/fisiologia
18.
Appl Physiol Nutr Metab ; 35(5): 657-70, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20962922

RESUMO

We hypothesized that a season of ice hockey would result in extensive remodeling of muscle. Tissue sampled from the vastus lateralis of 15 players (age = 20.6 ± 0.4 years; mean ± SE) prior to (PRE) and following (POST) a season was used to characterize specific adaptations. Measurement of representative metabolic pathway enzymes indicated higher maximal activities in POST than in PRE (p < 0.05) for succinic dehydrogenase (3.26 ± 0.31 vs. 3.91 ± 0.11 mol mg protein(-1) min(-1)), citrate synthase (7.26 ± 0.70 vs. 8.70 ± 0.55 mol mg protein(-1) min(-1)), and phosphofructokinase (12.8 ± 1.3 vs. 14.4 ± 0.96 mol mg protein(-1) min(-1)) only. The season resulted in an increase in Na+-K+-ATPase concentration (253 ± 6.3 vs. 265 ± 6.0 pmol g(-1) wet weight), a decrease (p < 0.05) in maximal activity of the sarcoplasmic reticulum Ca2+-ATPase (107 ± 4.2 micromol g protein(-1) min(-1) vs. 92.0 ± 4.6 micromol g protein(-1) min(-1)), and no change in the distribution (%) of fibre types. A smaller (p < 0.05) cross-sectional area (CSA) for both type I (-11.7%) and type IIA (-18.2%) fibres and a higher (p < 0.05) capillary count/CSA for type I (+17.9%) and type IIA (+17.2%) were also found over the season. No changes were found in peak oxygen consumption (51.4 ± 1.2 mL kg(-1) min(-1) vs. 52.3 ± 1.3 mL kg(-1) min(-1)). The results suggest, based on the alterations in oxidative and perfusion potentials and muscle mass, that the dominant adaptations are in support of oxidative metabolism, which occurs at the expense of fibre CSA and possibly force-generating potential.


Assuntos
Adaptação Fisiológica/fisiologia , Metabolismo Energético/fisiologia , Hóquei/fisiologia , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Adulto , ATPases Transportadoras de Cálcio/metabolismo , Citrato (si)-Sintase/metabolismo , Exercício Físico/fisiologia , Humanos , Masculino , Fibras Musculares Esqueléticas/enzimologia , Proteínas Musculares/metabolismo , Consumo de Oxigênio/fisiologia , Fosfofrutoquinase-1 Muscular/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Succinato Desidrogenase/metabolismo
19.
Free Radic Biol Med ; 48(7): 953-60, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20080177

RESUMO

Contractile activity induces a marked increase in glycolytic activity and gene expression of enzymes and transporters involved in glucose metabolism in skeletal muscle. Muscle contraction also increases the production of reactive oxygen species (ROS). In this study, the effects of treatment with N-acetylcysteine (NAC), a potent antioxidant compound, on contraction-stimulated glycolysis were investigated in electrically stimulated primary rat skeletal muscle cells. The following parameters were measured: 2-[(3)H]deoxyglucose (2-DG) uptake; activities of hexokinase, phosphofructokinase (PFK), and glucose-6-phosphate dehydrogenase (G6PDH); lactate production; and expression of the glucose transporter 4 (GLUT4), hexokinase II (HKII), and PFK genes after one bout of electrical stimulation in primary rat myotubes. NAC treatment decreased ROS signal by 49% in resting muscle cells and abolished the muscle contraction-induced increase in ROS levels. In resting cells, NAC decreased mRNA and protein contents of GLUT4, mRNA content and activity of PFK, and lactate production. NAC treatment suppressed the contraction-mediated increase in 2-DG uptake; lactate production; hexokinase, PFK, and G6PDH activities; and gene expression of GLUT4, HKII, and PFK. Similar to muscle contraction, exogenous H(2)O(2) (500 nM) administration increased 2-DG uptake; lactate production; hexokinase, PFK, and G6PDH activities; and gene expression of GLUT4, HKII, and PFK. These findings support the proposition that ROS endogenously produced play an important role in the changes in glycolytic activity and gene expression of GLUT4, HKII, and PFK induced by contraction in skeletal muscle cells.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Glucose/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Células Cultivadas , Desoxiglucose/metabolismo , Estimulação Elétrica , Glucose/genética , Transportador de Glucose Tipo 4/genética , Glucosefosfato Desidrogenase/metabolismo , Glicólise/efeitos dos fármacos , Hexoquinase/genética , Hexoquinase/metabolismo , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/patologia , Fosfofrutoquinase-1 Muscular/genética , Fosfofrutoquinase-1 Muscular/metabolismo , Ratos
20.
J Appl Physiol (1985) ; 107(6): 1771-80, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19797693

RESUMO

The present study examined muscle adaptations and alterations in work capacity in endurance-trained runners as a result of a reduced amount of training combined with speed endurance training. For a 6- to 9-wk period, 17 runners were assigned to either a speed endurance group with a 25% reduction in the amount of training but including speed endurance training consisting of six to twelve 30-s sprint runs 3-4 times/wk (SET group n = 12) or a control group (n = 5), which continued the endurance training ( approximately 55 km/wk). For the SET group, the expression of the muscle Na(+)-K(+) pump alpha(2)-subunit was 68% higher (P < 0.05) and the plasma K(+) level was reduced (P < 0.05) during repeated intense running after 9 wk. Performance in a 30-s sprint test and the first of the supramaximal exhaustive runs was improved (P < 0.05) by 7% and 36%, respectively, after the speed endurance training period. In the SET group, maximal O(2) uptake was unaltered, but the 3-km (3,000-m) time was reduced (P < 0.05) from 10.4 +/- 0.1 to 10.1 +/- 0.1 min and the 10-km (10,000-m) time was improved from 37.3 +/- 0.4 to 36.3 +/- 0.4 min (means +/- SE). Muscle protein expression and performance remained unaltered in the control group. The present data suggest that both short- and long-term exercise performances can be improved with a reduction in training volume if speed endurance training is performed and that the Na(+)-K(+) pump plays a role in the control of K(+) homeostasis and in the development of fatigue during repeated high-intensity exercise.


Assuntos
Músculo Esquelético/metabolismo , Consumo de Oxigênio/fisiologia , Resistência Física/fisiologia , Esforço Físico/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Adulto , Análise de Variância , Desempenho Atlético/fisiologia , Western Blotting , Citrato (si)-Sintase/metabolismo , Creatina Quinase/metabolismo , Ensaios Enzimáticos , Humanos , Ácido Láctico/sangue , Masculino , Músculo Esquelético/fisiologia , Fosfofrutoquinase-1 Muscular/metabolismo , Potássio/sangue , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA