Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Commun Signal ; 22(1): 51, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233839

RESUMEN

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.


Asunto(s)
Citidina/análogos & derivados , Osteosarcoma , Fosfofructoquinasas , Humanos , Lactato Deshidrogenasa 5/metabolismo , Fosfofructoquinasas/metabolismo , Acetilación , ARN/metabolismo , Glucólisis/genética , Osteosarcoma/patología , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Factores de Empalme de ARN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Acetiltransferasas N-Terminal/metabolismo
2.
Cell Death Dis ; 13(10): 876, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36253358

RESUMEN

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.


Asunto(s)
Exosomas , Enfermedades Renales , Células Madre Mesenquimatosas , MicroARNs , Fosfofructoquinasa-1 Tipo Muscular , Humanos , Exosomas/genética , Exosomas/metabolismo , Fibrosis , Glucólisis/genética , Enfermedades Renales/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Músculos/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Isoformas de Proteínas/metabolismo
3.
Genes (Basel) ; 13(3)2022 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-35328104

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Edad de Inicio , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras , Femenino , Glucólisis/genética , Hexoquinasa , Humanos , Proteínas de la Membrana , Metástasis de la Neoplasia , Pakistán , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Hormonas Tiroideas , Proteínas de Unión a Hormona Tiroide
4.
Cell Death Dis ; 12(4): 408, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859186

RESUMEN

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.


Asunto(s)
Carcinoma Epitelial de Ovario/genética , Glucólisis/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Animales , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones
5.
Mol Cell ; 81(9): 1905-1919.e12, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33852893

RESUMEN

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.


Asunto(s)
Glucólisis , Fosforilación Oxidativa , Precursores del ARN/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Ácido Glutámico/metabolismo , Glucógeno/metabolismo , Glucólisis/genética , Células HEK293 , Células HeLa , Humanos , Células K562 , Mitocondrias/genética , Mitocondrias/metabolismo , Oxidación-Reducción , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Precursores del ARN/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética
6.
Genomics ; 113(1 Pt 1): 135-141, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33279650

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Neoplasias de Cabeza y Cuello/genética , MicroARNs/genética , Anciano , Biomarcadores de Tumor/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Femenino , Filaminas/genética , Filaminas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Profilinas/genética , Profilinas/metabolismo
7.
Biochem Biophys Res Commun ; 530(1): 67-74, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32828317

RESUMEN

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.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proteína Forkhead Box O3/metabolismo , Neoplasias Renales/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Transducción de Señal , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteína Forkhead Box O3/análisis , Humanos , Neoplasias Renales/diagnóstico , Neoplasias Renales/patología , Fosfofructoquinasa-1 Tipo Muscular/análisis , Pronóstico
8.
Proc Natl Acad Sci U S A ; 117(12): 6726-6732, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32156725

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Receptores LHRH/genética , Sermorelina/análogos & derivados , Empalme Alternativo , Animales , Apoptosis , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Femenino , Glucólisis , Humanos , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Receptores LHRH/antagonistas & inhibidores , Sermorelina/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Phys Biol ; 16(6): 066007, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31469100

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Fructosadifosfatos/metabolismo , Glucólisis , Proteínas de la Membrana/genética , Fosfofructoquinasa-1 Tipo Muscular/genética , Hormonas Tiroideas/genética , Proteínas Portadoras/metabolismo , Reprogramación Celular , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
10.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 602-607, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29563071

RESUMEN

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.


Asunto(s)
Calicreínas/metabolismo , Fragmentos de Péptidos/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Procesamiento Proteico-Postraduccional , Electroforesis en Gel de Poliacrilamida , Transferencia Resonante de Energía de Fluorescencia , Humanos , Calicreínas/genética , Cinética , Fragmentos de Péptidos/genética , Fosfofructoquinasa-1 Tipo Muscular/genética , Proteolisis , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Especificidad por Sustrato
11.
Biosci Biotechnol Biochem ; 81(8): 1478-1483, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28508704

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Glucosa/metabolismo , Músculo Esquelético/metabolismo , Sus scrofa/genética , Porcinos/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Transporte Biológico , Domesticación , Perfilación de la Expresión Génica , Gluconeogénesis/genética , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Glucólisis/genética , Hexoquinasa/genética , Hexoquinasa/metabolismo , Lactato Deshidrogenasas/genética , Lactato Deshidrogenasas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Especificidad de Órganos , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Especificidad de la Especie , Sus scrofa/metabolismo , Porcinos/metabolismo
12.
Am J Physiol Cell Physiol ; 304(2): C180-93, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23114964

RESUMEN

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.


Asunto(s)
Glucólisis/fisiología , Músculo Esquelético/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/análisis , Calcio/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Simulación por Computador , Concentración de Iones de Hidrógeno , Isquemia/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Masculino , Modelos Biológicos , Contracción Muscular/fisiología , Fosfocreatina/análisis , Fosfocreatina/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/química , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Condicionamiento Físico Animal/fisiología , Ratas , Ratas Wistar
13.
FASEB J ; 26(11): 4710-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22767230

RESUMEN

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.


Asunto(s)
Glucólisis/fisiología , MicroARNs/metabolismo , Estrés Oxidativo/fisiología , Adenocarcinoma/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Clonación Molecular , ADN Complementario/genética , Regulación de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , MicroARNs/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Reacción en Cadena de la Polimerasa , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
J Biol Chem ; 287(21): 17546-17553, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22474333

RESUMEN

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).


Asunto(s)
Adenosina Difosfato/química , Adenosina Trifosfato/química , Fosfofructoquinasa-1 Tipo Muscular/química , Adenosina Difosfato/genética , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Regulación Alostérica/fisiología , Sitios de Unión , Evolución Molecular , Humanos , Mutación , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo
15.
J Cell Biochem ; 113(5): 1692-703, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22213537

RESUMEN

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.


Asunto(s)
Neoplasias/enzimología , Neoplasias/genética , Fosfofructoquinasa-1/metabolismo , Animales , Secuencia de Bases , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Línea Celular Tumoral , ADN Complementario/genética , Activación Enzimática , Femenino , Células HeLa , Humanos , Cinética , Hígado/enzimología , Neoplasias Hepáticas Experimentales/enzimología , Neoplasias Hepáticas Experimentales/genética , Miocardio/enzimología , Fosfofructoquinasa-1/genética , Fosfofructoquinasa-1 Tipo Hepático/genética , Fosfofructoquinasa-1 Tipo Hepático/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Fosfofructoquinasa-1 Tipo C/genética , Fosfofructoquinasa-1 Tipo C/metabolismo , Polimorfismo Genético , Ratas , Ratas Wistar , Especificidad por Sustrato , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/genética
16.
PLoS One ; 6(5): e19645, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21573193

RESUMEN

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.


Asunto(s)
Neoplasias/enzimología , Neoplasias/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Western Blotting , Línea Celular Tumoral , ADN Complementario/genética , Endopeptidasa K/metabolismo , Escherichia coli/citología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Fructosadifosfatos/farmacología , Glucosa/metabolismo , Células HEK293 , Humanos , Ácido Láctico/biosíntesis , Ratones , Metástasis de la Neoplasia , Neoplasias/patología , Fragmentos de Péptidos/metabolismo , Fosfofructoquinasa-1 Tipo Muscular/antagonistas & inhibidores , Fosfofructoquinasa-1 Tipo Muscular/química , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Conejos , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína , Transfección , Transformación Genética/efectos de los fármacos
17.
Meat Sci ; 89(2): 217-20, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21592677

RESUMEN

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.


Asunto(s)
Nucleótidos de Adenina/análisis , Glucógeno Fosforilasa/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Carne , Músculo Esquelético/enzimología , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Conductividad Eléctrica , Genotipo , Glucógeno Fosforilasa/análisis , L-Lactato Deshidrogenasa/análisis , Mutación , Fosfofructoquinasa-1 Tipo Muscular/análisis , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Porcinos/clasificación , Porcinos/genética , Porcinos/fisiología
18.
Appl Physiol Nutr Metab ; 35(5): 657-70, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20962922

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica/fisiología , Metabolismo Energético/fisiología , Hockey/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Adulto , ATPasas Transportadoras de Calcio/metabolismo , Citrato (si)-Sintasa/metabolismo , Ejercicio Físico/fisiología , Humanos , Masculino , Fibras Musculares Esqueléticas/enzimología , Proteínas Musculares/metabolismo , Consumo de Oxígeno/fisiología , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Succinato Deshidrogenasa/metabolismo
19.
Free Radic Biol Med ; 48(7): 953-60, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20080177

RESUMEN

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.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Células Cultivadas , Desoxiglucosa/metabolismo , Estimulación Eléctrica , Glucosa/genética , Transportador de Glucosa de Tipo 4/genética , Glucosafosfato Deshidrogenasa/metabolismo , Glucólisis/efectos de los fármacos , Hexoquinasa/genética , Hexoquinasa/metabolismo , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/patología , Fosfofructoquinasa-1 Tipo Muscular/genética , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Ratas
20.
J Appl Physiol (1985) ; 107(6): 1771-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19797693

RESUMEN

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.


Asunto(s)
Músculo Esquelético/metabolismo , Consumo de Oxígeno/fisiología , Resistencia Física/fisiología , Esfuerzo Físico/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adulto , Análisis de Varianza , Rendimiento Atlético/fisiología , Western Blotting , Citrato (si)-Sintasa/metabolismo , Creatina Quinasa/metabolismo , Pruebas de Enzimas , Humanos , Ácido Láctico/sangre , Masculino , Músculo Esquelético/fisiología , Fosfofructoquinasa-1 Tipo Muscular/metabolismo , Potasio/sangre , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA