Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 525
Filtrar
1.
J Biochem Mol Toxicol ; 38(8): e23799, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39132768

RESUMEN

It is well established that pyruvate kinase M2 (PKM2) activity contributes to metabolic reprogramming in various cancers, including colorectal cancer (CRC). Estrogen or 17ß-estradiol (E2) signaling is also known to modulate glycolysis markers in cancer cells. However, whether the inhibition of PKM2 combined with E2 treatment could adversely affect glucose metabolism in CRC cells remains to be investigated. First, we confirmed the metabolic plasticity of CRC cells under varying environmental conditions. Next, we identified glycolysis markers that were upregulated in CRC patients and assessed in vitro mRNA levels following E2 treatment. We found that PKM2 expression, which is highly upregulated in CRC clinical samples, is not altered by E2 treatment in CRC cells. In this study, glucose uptake, generation of reactive oxygen species (ROS), lactate production, cell viability, and apoptosis were evaluated in CRC cells following E2 treatment, PKM2 silencing, or a combination of both. Compared to individual treatments, combination therapy resulted in a significant reduction in cell viability and enhanced apoptosis. Glucose uptake and ROS production were markedly reduced in PKM2-silenced E2-treated cells. The data presented here suggest that E2 signaling combined with PKM2 inhibition cumulatively targets glucose metabolism in a manner that negatively impacts CRC cell growth. These findings hold promise for novel therapeutic strategies targeting altered metabolic pathways in CRC.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Hormonas Tiroideas/metabolismo , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Estrógenos/farmacología , Proteínas de Unión a Hormona Tiroide , Estradiol/farmacología , Apoptosis/efectos de los fármacos , Glucosa/metabolismo , Proteínas Portadoras/metabolismo , Piruvato Quinasa/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Glucólisis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Femenino
2.
Int J Mol Sci ; 25(14)2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39063228

RESUMEN

Metabolic dysfunction-associated fatty liver disease (MAFLD) presents a significant global health challenge, characterized by the accumulation of liver fat and impacting a considerable portion of the worldwide population. Despite its widespread occurrence, effective treatments for MAFLD are limited. The liver-specific isoform of pyruvate kinase (PKL) has been identified as a promising target for developing MAFLD therapies. Urolithin C, an allosteric inhibitor of PKL, has shown potential in preliminary studies. Expanding upon this groundwork, our study delved into delineating the structure-activity relationship of urolithin C via the synthesis of sulfone-based urolithin analogs. Our results highlight that incorporating a sulfone moiety leads to substantial PKL inhibition, with additional catechol moieties further enhancing this effect. Despite modest improvements in liver cell lines, there was a significant increase in inhibition observed in HepG2 cell lysates. Specifically, compounds 15d, 9d, 15e, 18a, 12d, and 15a displayed promising IC50 values ranging from 4.3 µM to 18.7 µM. Notably, compound 15e not only demonstrated a decrease in PKL activity and triacylglycerol (TAG) content but also showed efficient cellular uptake. These findings position compound 15e as a promising candidate for pharmacological MAFLD treatment, warranting further research and studies.


Asunto(s)
Hígado , Piruvato Quinasa , Sulfonas , Humanos , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Sulfonas/química , Sulfonas/farmacología , Sulfonas/síntesis química , Células Hep G2 , Hígado/metabolismo , Relación Estructura-Actividad , Regulación Alostérica/efectos de los fármacos , Diseño de Fármacos , Cumarinas/química , Cumarinas/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química
3.
Chem Res Toxicol ; 37(8): 1394-1403, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39066737

RESUMEN

Aerobic glycolysis is a metabolic reprogramming of tumor cells that is essential for sustaining their phenotype of fast multiplication by continuously supplying energy and mass. Pyruvate kinase M2 (PKM2) has a vital role in this process, which has given it high interest as a target for anticancer drug development. With potent toxicity to many types of cancer cells, polyphyllin II (PP2), a steroidal saponin isolated from the herbaceous plant Rhizoma paridis, brought to our attention that it might interfere with the PKM2 activity. In this study, we discovered that PP2 was a novel agonist of PKM2. PP2 activated recombinant PKM2 and changed the protein's oligomeric state to activate intracellular PKM2. At the same time, PP2 suppressed its protein kinase function by decreasing the content of nuclear PKM2. The mRNA levels of its downstream genes, such as Glut1, LDHA, and MYC, were inhibited. In addition, PP2 induced oxidative stress by downregulating the expression and activity of antioxidant proteins such as NQO1, TrxR, and Trx in HT-1080 cells, which in turn led to mitochondrial dysfunction and ultimately induced apoptosis. Moreover, PP2 reduced the proliferation and migration of HT-1080 cells. Thus, targeting the glycolysis pathway offers an unprecedented mode of action for comprehending PP2's pharmacological impacts and advances PP2's further development in fibrosarcoma therapy.


Asunto(s)
Apoptosis , Piruvato Quinasa , Saponinas , Línea Celular Tumoral , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , Saponinas/farmacología , Saponinas/toxicidad , Activación Enzimática/efectos de los fármacos , Homeostasis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Apoptosis/efectos de los fármacos , Fibrosarcoma , Humanos
4.
Bioorg Med Chem Lett ; 110: 129865, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38950758

RESUMEN

Pyruvate kinase (PK) is an essential component of cellular metabolism, converting ADP and phosphoenolpyruvate (PEP) to pyruvate in the final step of glycolysis. Of the four unique isoforms of pyruvate kinase, R (PKR) is expressed exclusively in red blood cells and is a tetrameric enzyme that depends on fructose-1,6-bisphosphate (FBP) for activation. PKR deficiency leads to hemolysis of red blood cells resulting in anemia. Activation of PKR in both sickle cell disease and beta-thalassemia patients could lead to improved red blood cell fitness and survival. The discovery of a novel series of substituted urea PKR activators, via the serendipitous identification and diligent characterization of a minor impurity in an High Throughput Screening (HTS) hit will be discussed.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Piruvato Quinasa , Piruvato Quinasa/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Humanos , Descubrimiento de Drogas , Relación Estructura-Actividad , Urea/química , Urea/farmacología , Activadores de Enzimas/farmacología , Activadores de Enzimas/química , Activadores de Enzimas/síntesis química , Estructura Molecular , Animales
5.
Life Sci ; 351: 122837, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879156

RESUMEN

AIM: Chronic sympathetic stimulation has been identified as a primary factor in the pathogenesis of cardiac hypertrophy (CH). However, there is no appropriate treatment available for the management of CH. Recently, it has been revealed that pyruvate kinase M2 (PKM2) plays a significant role in cardiac remodeling, fibrosis, and hypertrophy. However, the therapeutic potential of selective PKM2 inhibitor has not yet been explored in cardiac hypertrophy. Thus, in the current study, we have studied the cardioprotective potential of Compound 3K, a selective PKM2 inhibitor in isoproterenol-induced CH model. METHODS: To induce cardiac hypertrophy, male Wistar rats were subcutaneously administered isoproterenol (ISO, 5 mg/kg/day) for 14 days. Compound 3K at dosages of 2 and 4 mg/kg orally was administered to ISO-treated rats for 14 days to explore its effects on various parameters like ECG, ventricular functions, hypertrophic markers, histology, inflammation, and protein expression were performed. RESULTS: Fourteen days administration of ISO resulted in the induction of CH, which was evidenced by alterations in ECG, ventricular dysfunctions, increase in hypertrophy markers, and fibrosis. The immunoblotting of hypertrophy heart revealed the significant rise in PKM2 and reduction in PKM1 protein expression. Treatment with Compound 3K led to downregulation of PKM2 and upregulation of PKM1 protein expression. Compound 3K showed cardioprotective effects by improving ECG, cardiac functions, hypertrophy markers, inflammation, and fibrosis. Further, it also reduced cardiac expression of PKM2-associated splicing protein, HIF-1α, and caspase-3. CONCLUSION: Our findings suggest that Compound 3K has a potential cardioprotective effect via PKM2 inhibition in isoproterenol-induced CH.


Asunto(s)
Cardiomegalia , Isoproterenol , Piruvato Quinasa , Animales , Masculino , Ratas , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/prevención & control , Cardiomegalia/metabolismo , Cardiotónicos/farmacología , Fibrosis , Isoproterenol/toxicidad , Piruvato Quinasa/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Ratas Wistar , Transducción de Señal/efectos de los fármacos
6.
Bioorg Chem ; 147: 107425, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38714117

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) comprises a broad range of liver disease including hepatocellular carcinoma (HCC) with is no FDA-approved drug. Liver pyruvate kinase (PKL) is a major regulator of metabolic flux and ATP generation in liver presenting a potential target for the treatment of NAFLD. Based on our recent finding of JNK-5A's effectiveness in inhibiting PKLR expression through a drug repositioning pipeline, this study aims to improve its efficacy further. We synthesized a series of JNK-5A analogues with targeted modifications, guided by molecular docking studies. These compounds were evaluated for their activities on PKL expression, cell viability, triacylglyceride (TAG) levels, and the expressions of steatosis-related proteins in the human HepG2 cell line. Subsequently, the efficacy of these compounds was assessed in reducing TAG level and toxicity. Compounds 40 (SET-151) and 41 (SET-152) proved to be the most efficient in reducing TAG levels (11.51 ± 0.90 % and 10.77 ± 0.67 %) and demonstrated lower toxicity (61.60 ± 5.00 % and 43.87 ± 1.42 %) in HepG2 cells. Additionally, all synthesized compounds were evaluated for their anti-cancer properties revealing that compound 74 (SET-171) exhibited the highest toxicity in cell viability with IC50 values of 8.82 µM and 2.97 µM in HepG2 and Huh7 cell lines, respectively. To summarize, compounds 40 (SET-151) and 41 (SET-152) show potential for treating NAFLD, while compound 74 (SET-171) holds potential for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular , Diseño de Fármacos , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Inhibidores de Proteínas Quinasas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Relación Estructura-Actividad , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Células Hep G2 , Estructura Molecular , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Simulación del Acoplamiento Molecular , Relación Dosis-Respuesta a Droga , Supervivencia Celular/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química
7.
Eur J Med Chem ; 272: 116426, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38718622

RESUMEN

Pyruvate kinase isoform 2 (PKM2) is closely related to the regulation of Th17/Treg balance, which is considered to be an effective strategy for UC therapy. Parthenolide (PTL), a natural product, only possesses moderate PKM2-activating activity. Thus, five series of PTL derivatives are designed and synthesized to improve PKM2-activated activities and anti-UC abilities. Through detailed structure optimization, B4 demonstrates potent T-cell anti-proliferation activity (IC50 = 0.43 µM) and excellent PKM2-activated ability (AC50 = 0.144 µM). Subsequently, through mass spectrometry analysis, B4 is identified to interact with Cys423 of PKM2 via covalent-bond. Molecular docking and molecular dynamic simulation results reveal that the trifluoromethoxy of B4 forms a stronger hydrophobic interaction with Ala401, Pro402, and Ile403. In addition, B4 has a significant effect only on Th17 cell differentiation, thereby regulating the Th17/Treg balance. The effect of B4 on Th17/Treg imbalance can be attributed to inhibition of PKM2 dimer translocation and suppression of glucose metabolism. Finally, B4 can notably ameliorate the symptoms of dextran sulfate sodium (DSS)-induced colitis in mouse model in vivo. Thus, B4 is confirmed as a potent PKM2 activator, and has the potential to develop as a novel anti-UC agent.


Asunto(s)
Colitis Ulcerosa , Diseño de Fármacos , Lactonas , Piruvato Quinasa , Sesquiterpenos , Sesquiterpenos/farmacología , Sesquiterpenos/química , Sesquiterpenos/síntesis química , Animales , Ratones , Piruvato Quinasa/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Lactonas/farmacología , Lactonas/química , Lactonas/síntesis química , Relación Estructura-Actividad , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Humanos , Estructura Molecular , Proliferación Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Relación Dosis-Respuesta a Droga , Masculino , Sulfato de Dextran , Simulación del Acoplamiento Molecular , Hormonas Tiroideas/metabolismo , Células Th17/efectos de los fármacos , Proteínas de Unión a Hormona Tiroide
8.
Toxicol Appl Pharmacol ; 485: 116905, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38521371

RESUMEN

Myocardial infarction (MI) or heart attack arises from acute or chronic prolonged ischemic conditions in the myocardium. Although several risk factors are associated with MI pathophysiology, one of the risk factors is an imbalance in the oxygen supply. The current available MI therapies are still inadequate due to the complexity of MI pathophysiology. Pyruvate kinase M2 (PKM2) has been implicated in numerous CVDs pathologies. However, the effect of specific pharmacological intervention targeting PKM2 has not been studied in MI. Therefore, in this study, we explored the effect of compound 3K, a PKM2-specific inhibitor, in isoproterenol-induced acute MI model. In this study, in order to induce MI in rats, isoproterenol (ISO) was administered at a dose of 100 mg/kg over two days at an interval of 24 h. Specific PKM2 inhibitor, compound 3K (2 and 4 mg/kg), was administered in MI rats to investigate its cardioprotective potential. After the last administration of compound 3K, ECG and hemodynamic parameters were recorded using a PV-loop system. Cardiac histology, western blotting, and plasmatic cardiac damage markers were evaluated to elucidate the underlying mechanisms. Treatment of compound 3K significantly reduced ISO-induced alterations in ECG, ventricular functions, cardiac damage, infarct size, and cardiac fibrosis. Compound 3K treatment produced significant increase in PKM1 expression and decrease in PKM2 expression. In addition, HIF-1α, caspase-3, c-Myc, and PTBP1 expression were also reduced after compound 3K treatment. This study demonstrates the cardioprotective potential of compound 3K in MI, and its mechanisms of cardioprotective action.


Asunto(s)
Cardiotónicos , Isoproterenol , Infarto del Miocardio , Piruvato Quinasa , Animales , Isoproterenol/toxicidad , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/prevención & control , Infarto del Miocardio/patología , Masculino , Ratas , Piruvato Quinasa/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Ratas Wistar , Miocardio/patología , Miocardio/metabolismo , Miocardio/enzimología , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Inhibidores de Proteínas Quinasas/farmacología , Hormonas Tiroideas
9.
Mol Metab ; 66: 101625, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36374631

RESUMEN

OBJECTIVE: The endocrine pancreatic ß-cells play a pivotal role in maintaining whole-body glucose homeostasis and its dysregulation is a consistent feature in all forms of diabetes. However, knowledge of intracellular regulators that modulate ß-cell function remains incomplete. We investigated the physiological role of ROCK1 in the regulation of insulin secretion and glucose homeostasis. METHODS: Mice lacking ROCK1 in pancreatic ß-cells (RIP-Cre; ROCK1loxP/loxP, ß-ROCK1-/-) were studied. Glucose and insulin tolerance tests as well as glucose-stimulated insulin secretion (GSIS) were measured. An insulin secretion response to a direct glucose or pyruvate or pyruvate kinase (PK) activator stimulation in isolated islets from ß-ROCK1-/- mice or ß-cell lines with knockdown of ROCK1 was also evaluated. A proximity ligation assay was performed to determine the physical interactions between PK and ROCK1. RESULTS: Mice with a deficiency of ROCK1 in pancreatic ß-cells exhibited significantly increased blood glucose levels and reduced serum insulin without changes in body weight. Interestingly, ß-ROCK1-/- mice displayed a progressive impairment of glucose tolerance while maintaining insulin sensitivity mostly due to impaired GSIS. Consistently, GSIS markedly decreased in ROCK1-deficient islets and ROCK1 knockdown INS-1 cells. Concurrently, ROCK1 blockade led to a significant decrease in intracellular calcium and ATP levels and oxygen consumption rates in isolated islets and INS-1 cells. Treatment of ROCK1-deficient islets or ROCK1 knockdown ß-cells either with pyruvate or a PK activator rescued the impaired GSIS. Mechanistically, we observed that glucose stimulation in ß-cells greatly enhanced ROCK1 binding to PK. CONCLUSIONS: Our findings demonstrate that ß-cell ROCK1 is essential for glucose-stimulated insulin secretion and for glucose homeostasis and that ROCK1 acts as an upstream regulator of glycolytic pyruvate kinase signaling.


Asunto(s)
Secreción de Insulina , Insulina , Piruvato Quinasa , Quinasas Asociadas a rho , Animales , Ratones , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina/fisiología , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Piruvatos
10.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119206, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35026348

RESUMEN

Pyruvate kinase isoform M2 (PKM2) is a rate-limiting glycolytic enzyme that is widely expressed in embryonic tissues. The expression of PKM2 declines in some tissues following embryogenesis, while other pyruvate kinase isozymes are upregulated. However, PKM2 is highly expressed in cancer cells and is believed to play a role in supporting anabolic processes during tumour formation. In this study, PKM2 was identified as an inositol 1,4,5-trisphosphate receptor (IP3R)-interacting protein by mass spectrometry. The PKM2:IP3R interaction was further characterized by pull-down and co-immunoprecipitation assays, which showed that PKM2 interacted with all three IP3R isoforms. Moreover, fluorescence microscopy indicated that both IP3R and PKM2 localized at the endoplasmic reticulum. PKM2 binds to IP3R at a highly conserved 21-amino acid site (corresponding to amino acids 2078-2098 in mouse type 1 IP3R isoform). Synthetic peptides (denoted 'TAT-D5SD' and 'D5SD'), based on the amino acid sequence at this site, disrupted the PKM2:IP3R interaction and potentiated IP3R-mediated Ca2+ release both in intact cells (TAT-D5SD peptide) and in a unidirectional 45Ca2+ flux assay on permeabilized cells (D5SD peptide). The TAT-D5SD peptide did not affect the enzymatic activity of PKM2. Reducing PKM2 protein expression using siRNA increased IP3R-mediated Ca2+ signalling in intact cells without altering the ER Ca2+ content. These data identify PKM2 as an IP3R-interacting protein that inhibits intracellular Ca2+ signalling. The elevated expression of PKM2 in cancer cells is therefore not solely connected to its canonical role in glycolytic metabolism, rather PKM2 also has a novel non-canonical role in regulating intracellular signalling.


Asunto(s)
Señalización del Calcio , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Piruvato Quinasa/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Línea Celular , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/química , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
11.
Exp Eye Res ; 213: 108823, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34752817

RESUMEN

Choroidal neovascularization (CNV), a feature of neovasular age-related macular degeneration (AMD), acts as a leading cause of vision loss in the elderly. Shikonin (SHI), a natural bioactive compound extracted from Chinese herb radix arnebiae, exerts anti-inflammatory and anti-angiogenic roles and also acts as a potential pyruvate kinase M2 (PKM2) inhibitor in macrophages. The major immune cells macrophages infiltrate the CNV lesions, where the production of pro-angiognic cytokines from macrophage facilitates the development of CNV. PKM2 contributes to the neovascular diseases. In this study, we found that SHI oral gavage alleviated the leakage, area and volume of mouse laser-induced CNV lesion and inhibited macrophage infiltration without ocular cytotoxicity. Moreover, SHI inhibited the secretion of pro-angiogenic cytokine, including basic fibroblast growth factor (FGF2), insulin-like growth factor-1 (IGF1), chemokine (C-C motif) ligand 2 (CCL2), placental growth factor and vascular endothelial growth factor (VEGF), from primary human macrophages by down-regulating PKM2/STAT3/CD163 pathway, indicating a novel potential therapy strategy for CNV.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Naftoquinonas/uso terapéutico , Piruvato Quinasa/antagonistas & inhibidores , Inductores de la Angiogénesis/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Western Blotting , Células Cultivadas , Neovascularización Coroidal/enzimología , Cromatografía Líquida de Alta Presión , Colorantes/administración & dosificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Humanos , Etiquetado Corte-Fin in Situ , Verde de Indocianina/administración & dosificación , Macrófagos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Piruvato Quinasa/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo
12.
Sci Rep ; 11(1): 21726, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34741111

RESUMEN

Cryptotanshinone (CT) is an extract from the traditional Chinese medicine Salvia miltiorrhiza, which inhibits the growth of methicillin-resistant Staphylococcus aureus (MRSA) in vitro. This study aims to determine the antibacterial mechanisms of CT by integrating bioinformatics analysis and microbiology assay. The microarray data of GSE13203 was retrieved from the Gene Expression Omnibus (GEO) database to screen the differentially expressed genes (DEGs) of S. aureus strains that were treated with CT treatment. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to identify the potential target of CT. Data mining on the microarray dataset indicated that pyruvate kinase (PK) might be involved in the antimicrobial activities of CT. The minimum inhibition concentrations (MICs) of CT or vancomycin against the MRSA strain ATCC43300 and seven other clinical strains were determined using the broth dilution method. The effects of CT on the activity of PK were further measured. In vitro tests verified that CT inhibited the growth of an MRSA reference strain and seven other clinical strains. CT hampered the activity of the PK of ATCC43300 and five clinical MRSA strains. CT might hinder bacterial energy metabolism by inhibiting the activity of PK.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Fenantrenos/farmacología , Piruvato Quinasa/antagonistas & inhibidores , Biología Computacional , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Staphylococcus aureus Resistente a Meticilina/enzimología , Fenantrenos/uso terapéutico , Fitoterapia , Infecciones Estafilocócicas/tratamiento farmacológico
13.
ACS Appl Mater Interfaces ; 13(48): 56972-56987, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34797638

RESUMEN

Triple-negative breast cancer (TNBC) is typically associated with poor prognosis due to its only partial response to chemotherapy and lack of clinically established targeted therapies coupled with an aggressive disease course. Aerobic glycolysis is a hallmark of reprogrammed metabolic activity in cancer cells, which can be repressed by small-interfering RNA (siRNA). However, the lack of effective carriers to deliver vulnerable siRNA restricts the clinical potentials of glycolysis-based gene therapy for TNBC. Herein, we develop a tumor-targeted, biomimetic manganese dioxide (MnO2)-shrouded metal-organic framework (MOF) based nanomedicine to deliver siRNA against pyruvate kinase muscle isozyme M2 (siPKM2), wherein PKM2 is a rate-limiting enzyme in glycolysis, to inhibit the reprogrammed glycolysis of TNBC. This MOF-based genetic nanomedicine shows excellent monodispersity and stability and protects siPKM2 against degradation by nucleases. The nanomedicine not only substantially blocks the glycolytic pathway but also improves intracellular hypoxia in TNBC cells, with a resultant O2-enhanced anticancer effect. In the mice orthotopic TNBC model, the nanomedicine shows a remarkable therapeutic effect. Meanwhile, the Mn2+ ions released from acid microenvironment-responsive MnO2 enable in vivo monitoring of the therapeutic process with magnetic resonance imaging (MRI). Our study shows great promise with this MRI-visible MOF-based nanomedicine for treating TNBC by inhibition of glycolysis via the RNA interference.


Asunto(s)
Antineoplásicos/farmacología , Materiales Biomiméticos/farmacología , Inhibidores Enzimáticos/farmacología , Estructuras Metalorgánicas/farmacología , Piruvato Quinasa/antagonistas & inhibidores , Nanomedicina Teranóstica , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Femenino , Silenciador del Gen/efectos de los fármacos , Humanos , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Ensayo de Materiales , Estructuras Metalorgánicas/síntesis química , Estructuras Metalorgánicas/química , Ratones , Ratones Desnudos , Imagen Óptica , Óxidos/química , Óxidos/farmacología , Tamaño de la Partícula , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/metabolismo
14.
Dis Markers ; 2021: 1602797, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804260

RESUMEN

Retinitis pigmentosa (RP) is a major cause of blindness that is difficult to diagnose and treat. PKM2, a subtype of pyruvate kinase, is strongly associated with oxidative stress and is expressed in photoreceptors. We investigated whether PKM2 reduces photoreceptor cell apoptosis and evaluated possible antiapoptotic mechanisms in RP. We established RP models by exposing 661W cells to blue light and modulated PKM2 activity using a PKM2 inhibitor. We measured the apoptosis rates using calcein-acetoxymethyl ester/propidium iodide double staining and Cell Counting Kit-8, the oxidative stress levels using a reactive oxygen species assay, and the changes in protein expression by western blotting. Photodamage increased PKM2 expression, cellular oxidative stress, and apoptosis of 661W cells. PKM2 inhibition significantly reduced the levels of apoptosis and oxidative stress induced by photodamage. Our data suggest that PKM2 is a potential disease marker and therapeutic target for RP.


Asunto(s)
Luz/efectos adversos , Neuroprotección , Estrés Oxidativo , Células Fotorreceptoras/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Células Fotorreceptoras Retinianas Conos/metabolismo , Retinitis Pigmentosa/prevención & control , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Células Fotorreceptoras/patología , Células Fotorreceptoras/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Retinitis Pigmentosa/etiología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
15.
J Cell Mol Med ; 25(22): 10711-10723, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34687136

RESUMEN

Hypertensive cardiac remodelling is a common cause of heart failure. However, the molecular mechanisms regulating cardiac remodelling remain unclear. Pyruvate kinase isozyme type M2 (PKM2) is a key regulator of the processes of glycolysis and oxidative phosphorylation, but the roles in cardiac remodelling remain unknown. In the present study, we found that PKM2 was enhanced in angiotensin II (Ang II)-treated cardiac fibroblasts and hypertensive mouse hearts. Suppression of PKM2 by shikonin alleviated cardiomyocyte hypertrophy and fibrosis in Ang-II-induced cardiac remodelling in vivo. Furthermore, inhibition of PKM2 markedly attenuated the function of cardiac fibroblasts including proliferation, migration and collagen synthesis in vitro. Mechanistically, suppression of PKM2 inhibited cardiac remodelling by suppressing TGF-ß/Smad2/3, Jak2/Stat3 signalling pathways and oxidative stress. Together, this study suggests that PKM2 is an aggravator in Ang-II-mediated cardiac remodelling. The negative modulation of PKM2 may provide a promising therapeutic approach for hypertensive cardiac remodelling.


Asunto(s)
Angiotensina II/metabolismo , Janus Quinasa 2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Piruvato Quinasa/genética , Factor de Transcripción STAT3/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Remodelación Ventricular/genética , Animales , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica , Hipertensión/complicaciones , Hipertensión/etiología , Hipertensión/metabolismo , Masculino , Ratones , Modelos Biológicos , Piruvato Quinasa/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo
16.
Curr Top Med Chem ; 21(25): 2258-2271, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34348626

RESUMEN

INTRODUCTION: Pyruvate kinase isozyme M2 (PKM2) was observed to be overexpressed and play a key role in cell growth and cancer cells' metabolism. During the past years, phytochemicals have been developed as new treatment options for chemoprevention and cancer therapy. Natural resources, like shikonin (naphthoquinone) and its derivatives, have emerged to be high potential therapeutics in cancer treatment. METHODS: Our study aimed to design novel anti-tumour agents (PKM2 inhibitors) focusing on the shikonin scaffold with a better activity using computational methods. We applied a three-dimensional quantitative structure-activity relationship (3D-QSAR) approach using Field-based QSAR. RESULTS: The Comparative Molecular Field Analysis (CoMFA) and Comparative Molecular Similarity Indices Analysis (CoMSIA) were performed on a series of forty shikonin derivatives, including shikonin, to develop robust models and rationalize the PKM2 inhibitory activity profile by building a correlation between structural features and activity. CONCLUSION: These predictive computational models will further help the design and synthesis of potent PKM2 inhibitors and their fast biological assessment at a low cost.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Naftoquinonas/química , Piruvato Quinasa/antagonistas & inhibidores , Relación Estructura-Actividad Cuantitativa , Isoenzimas/antagonistas & inhibidores
17.
Int J Biol Sci ; 17(8): 1895-1908, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34131394

RESUMEN

Ovarian cancer is a common cause of death among gynecological cancers. Although ovarian cancer initially responds to chemotherapy, frequent recurrence in patients remains a therapeutic challenge. Pyruvate kinase M2 (PKM2) plays a pivotal role in regulating cancer cell survival. However, its therapeutic role remains unclear. Here, we investigated the anticancer effects of compound 3K, a specific PKM2 inhibitor, on the regulation of autophagic and apoptotic pathways in SK-OV-3 (PKM2-overexpressing human ovarian adenocarcinoma cell line). The anticancer effect of compound 3K was examined using MTT and colony formation assays in SK-OV-3 cells. PKM2 expression was positively correlated with the severity of the tumor, and expression of pro-apoptotic proteins increased in SK-OV-3 cells following compound 3K treatment. Compound 3K induced AMPK activation, which was accompanied by mTOR inhibition. Additionally, this compound inhibited glycolysis, resulting in reduced proliferation of SK-OV-3 cells. Compound 3K treatment suppressed tumor progression in an in vivo xenograft model. Our findings suggest that the inhibition of PKM2 by compound 3K affected the Warburg effect and induced autophagic cell death. Therefore, use of specific PKM2 inhibitors to block the glycolytic pathway and target cancer cell metabolism represents a promising therapeutic approach for treating PKM2-overexpressing ovarian cancer.


Asunto(s)
Adenocarcinoma , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas , Piruvato Quinasa/antagonistas & inhibidores , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Antineoplásicos/farmacología , Muerte Celular Autofágica/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre
18.
J Med Chem ; 63(21): 12623-12641, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33103423

RESUMEN

Natural bisindole alkaloids such as Hyrtinadine A and Alocasin A, which are known to exhibit diverse bioactivities, provide promising chemical scaffolds for drug development. By optimizing the Masuda borylation-Suzuki coupling sequence, a library of various natural product-derived and non-natural (di)azine-bridged bisindoles was created. While unsubstituted bisindoles were devoid of antibacterial activity, 5,5'-chloro derivatives were highly active against methicillin-resistant Staphylococcus aureus (MRSA) and further Gram-positive pathogens at minimal inhibitory concentrations ranging from 0.20 to 0.78 µM. These compounds showed strong bactericidal killing effects but only moderate cytotoxicity against human cell lines. Furthermore, the two front-runner compounds 4j and 4n exhibited potent in vivo efficacy against MRSA in a mouse wound infection model. Although structurally related bisindoles were reported to specifically target pyruvate kinase in MRSA, antibacterial activity of 4j and 4n is independent of pyruvate kinase. Rather, these compounds lead to bacterial membrane permeabilization and cellular efflux of low-molecular-weight molecules.


Asunto(s)
Alcaloides/química , Antibacterianos/farmacología , Indoles/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Alcaloides/metabolismo , Alcaloides/farmacología , Alcaloides/uso terapéutico , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/uso terapéutico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Femenino , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Peritonitis/tratamiento farmacológico , Peritonitis/patología , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Conejos
19.
Elife ; 92020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32812866

RESUMEN

Natural Killer (NK) cells have an important role in immune responses to viruses and tumours. Integrating changes in signal transduction pathways and cellular metabolism is essential for effective NK cells responses. The glycolytic enzyme Pyruvate Kinase Muscle 2 (PKM2) has described roles in regulating glycolytic flux and signal transduction, particularly gene transcription. While PKM2 expression is robustly induced in activated NK cells, mice lacking PKM2 in NK cells showed no defect in NK cell metabolism, transcription or antiviral responses to MCMV infection. NK cell metabolism was maintained due to compensatory PKM1 expression in PKM2-null NK cells. To further investigate the role of PKM2, we used TEPP-46, which increases PKM2 catalytic activity while inhibiting any PKM2 signalling functions. NK cells activated with TEPP-46 had reduced effector function due to TEPP-46-induced increases in oxidative stress. Overall, PKM2-regulated glycolytic metabolism and redox status, not transcriptional control, facilitate optimal NK cells responses.


Asunto(s)
Regulación de la Expresión Génica , Glucólisis , Células Asesinas Naturales/metabolismo , Piruvato Quinasa , Animales , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glucólisis/efectos de los fármacos , Glucólisis/genética , Ratones , Estrés Oxidativo , Piridazinas/farmacología , Pirroles/farmacología , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , Transducción de Señal
20.
JCI Insight ; 5(17)2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32750036

RESUMEN

One of the most significant adverse postburn responses is abnormal scar formation, such as keloids. Despite its prolificacy, the underlying pathophysiology of keloid development is unknown. We recently demonstrated that NLRP3 inflammasome, the master regulator of inflammatory and metabolic responses (e.g., aerobic glycolysis), is essential for physiological wound healing. Therefore, burn patients who develop keloids may exhibit altered immunometabolic responses at the site of injury, which interferes with normal healing and portends keloid development. Here, we confirmed keloid NLRP3 activation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.05], IL-18 [P < 0.01]) and upregulation in Glut1 (P < 0.001) and glycolytic enzymes. Burn skin similarly displayed enhanced glycolysis and Glut1 expression (P < 0.01). However, Glut1 was significantly higher in keloid compared with nonkeloid burn patients (>2 SD above mean). Targeting aberrant glucose metabolism with shikonin, a pyruvate kinase M2 inhibitor, dampened NLRP3-mediated inflammation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.01]) and improved healing in vivo. In summary, burn skin exhibited evidence of Warburg-like metabolism, similar to keloids. Targeting this altered metabolism could change the trajectory toward normal scarring, indicating the clinical possibility of shikonin for abnormal scar prevention.


Asunto(s)
Quemaduras/complicaciones , Glucólisis , Inflamación/prevención & control , Queloide/prevención & control , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Piel/efectos de los fármacos , Cicatrización de Heridas , Adulto , Animales , Antiinflamatorios no Esteroideos/farmacología , Estudios de Casos y Controles , Femenino , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Inflamasomas , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación , Queloide/etiología , Queloide/metabolismo , Queloide/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Naftoquinonas/farmacología , Piruvato Quinasa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA