Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biochem Funct ; 41(8): 1220-1229, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37707291

RESUMEN

Transforming growth factor ß1 (TGFß1) induces a cellular process known as epithelial-mesenchymal transition (EMT) associated with metabolic reprogramming, including enhanced glycolysis. Given the involvement of 6-phosphofructo-2-kinase/fructose 2,6-bisphosphatase (PFKFB) enzymes in glycolysis, we aimed to investigate whether TGFß1 regulates expressions of PFKFB genes and if PFKFBs are required for TGFß1-driven phenotypes. A549 and MCF-10A cell lines were used as TGFß1-driven EMT models. Messenger RNA expressions of PFKFB and EMT genes were determined by real-time quantitative polymerase chain reaction. A small interfering RNA approach was used to deplete PFKFB4 expression. A Matrigel invasion assay was conducted to assess the effect of PFKFB4 silencing on the TGFß1-enhanced invasion of A549 cells. F2,6BP levels were analyzed using an enzyme-coupled assay. Glucose and lactate concentrations were determined using colorimetric assays. TGFß1 robustly induced expression of the fourth isoform of PFKFBs, PFKFB4, in both cell lines. PFKFB4 depletion partially inhibits mesenchymal transdifferentiation caused by TGFß1 in A549 cells, as assessed by microscopy. Inductions of Snail in MCF-10A cells and Fibronectin in A549 cells and repressions of E-cadherin in both cell lines by TGFß1 are attenuated by PFKFB4 silencing. PFKFB4 silencing reduces F2,6BP and glycolytic activity, although TGFß1 alone does not affect these parameters. Finally, PFKFB4 depletion suppresses the TGFß1-driven invasion of A549 cells through Matrigel. Presented data suggest that TGFß1 induces the expression of PFKFB4 in A549 and MCF-10 cells, and PFKFB4 may be required for TGFß1-driven phenotypes such as EMT and invasion in these models.


Asunto(s)
Fosfofructoquinasa-2 , Factor de Crecimiento Transformador beta1 , Humanos , Células A549 , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Fructosa , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo
2.
Turk J Med Sci ; 53(3): 640-646, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37476896

RESUMEN

BACKGROUND: The study aims to profile the dual-specificity phosphatases (DUSP) expression in response to Transforming growth factor ß1 (TGFß1)-induced epithelial- mesenchymal transition (EMT) in ovarian adenocarcinoma cells. METHODS: The ovarian adenocarcinoma cell line SKOV3 was used as a TGFß1-induced EMT model. Cells were incubated with 5 ng/mL TGFß1 to induce EMT. EMT was confirmed with real-time qPCR, western blot, and immunofluorescence analyses of various EMT markers. Western blot was used to analyze phospho- and total MAPK protein levels. Typical and atypical DUSPs mRNA expression profile was determined by real-time qPCR. RESULTS: The epithelial marker E-cadherin expressions were decreased and mesenchymal EMT markers Snail and Slug expression levelswere increased after TGFß1 induction. Phosphorylation of ERK1/2 and p38 MAPK were enhanced in response to TGFß1 treatment. The expression of DUSP2, DUSP6, DUSP8, DUSP10, and DUSP13 were decreased while DUSP7, DUSP16, DUSP18, DUSP21, and DUSP27 were increased by TGFß1. DISCUSSION: TGFß1 induced EMT which was accompanied by increased activity of MAPKs, and led to marked changes in expressions of several DUSPs in SKOV3 cells.


Asunto(s)
Adenocarcinoma , Transición Epitelial-Mesenquimal , Humanos , Transición Epitelial-Mesenquimal/genética , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Línea Celular , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Adenocarcinoma/metabolismo , Células Epiteliales/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo
3.
Mol Neurobiol ; 60(11): 6248-6263, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37439958

RESUMEN

Heat shock response (HSR) which is regulated by heat shock factor 1 (HSF1) is the most important mechanism and the major regulator that prevents protein aggregation in neurodegenerative diseases. Excitotoxicity, which is the accumulation of excess glutamate in synaptic cleft, is observed in age-dependent neurodegenerative diseases and also in stroke, epilepsy, and brain trauma. Only a few studies in the literature show the link between excitotoxicity and HSR. In this study, we aimed to show the molecular mechanism underlying this link. We applied heat shock (HS) treatment and induced excitotoxicity with kainic acid (KA) in neuroblastoma (SHSY-5Y) and glia (immortalized human astrocytes (IHA)) cells. We observed that, only in SHSY-5Y cells, heat shock preconditioning increases cell survival after KA treatment. GLT-1 mRNA expression is increased as a result of KA treatment and HS due to the elevation of HSF1 binding to GLT-1 promoter which was induced by HSF1 phosphorylation and sumolation in SHSY-5Y cells. Additionally, glutamine synthetase and glutaminase expressions are increased after HS preconditioning in SHSY-5Y cells indicating that HS activates glutamate metabolism modulators and accelerates the glutamate cycle. In glia cells, we did not observe the effect of HS preconditioning. In summary, heat shock preconditioning might be protective against excitotoxicity-related cell death and degeneration.


Asunto(s)
Enfermedades Neurodegenerativas , Factores de Transcripción , Humanos , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Ácido Kaínico/toxicidad , Respuesta al Choque Térmico/genética , Ácido Glutámico/toxicidad
4.
Placenta ; 139: 200-211, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37463546

RESUMEN

INTRODUCTION: Placental inflammation is implicated in the pathophysiology of many pregnancy complications, including fetal growth restriction, preeclampsia, gestational diabetes, and choriocarcinoma. Mitochondrial dysfunction, one of the outcomes of placental inflammation, is characterized by loss of membrane potential, accumulation of oxygen radicals, mitochondrial protein folding defects, and disturbances in mitochondrial dynamics. Protein kinase R (PKR) is stimulated by double-stranded RNA and bacterial endotoxins in the presence of pathogens and is a critical immune response enzyme. PKR is also correlated with the cell death response during endoplasmic reticulum stress. In this study, we aim to investigate the effects of PKR activity stimulated by lipopolysaccharide (LPS), and double-stranded RNA analog (Poly I:C) on mitochondrial unfolded protein response (mtUPR), mitochondrial membrane potential, apoptosis, and oxidative stress in placental trophoblasts. METHODS: We applied LPS and Poly I:C to BeWo cells to induce PKR activation. In addition, cells were treated with 2-aminopurine (2-AP) to inhibit the kinase activity of PKR. Protein levels of ATP-dependent Clp protease proteolytic subunit (CLPP) and heat shock protein 60 (HSP60) were determined after treatments. Apoptotic markers were detected by real-time PCR and flow cytometry. PKR-induced reactive oxygen radicals (ROS) accumulation and mitochondrial membrane potential change were assessed by flow cytometry. RESULTS: It was determined that PKR activation-induced apoptosis in BeWo cells by reducing the levels of mtUPR proteins (CLPP and HSP60) and caused a decrease in mitochondrial membrane potential. PKR inhibition was sufficient for decreases in apoptotic markers and caused a reduction in the ratio of depolarized and ROS (+) cells. DISCUSSION: Our results showed that LPS and Poly I:C administration stimulated PKR in BeWo cells in vitro. Furthermore, PKR activation is correlated with the levels of proteins involved in mitochondrial homeostasis and apoptosis. Our findings will contribute to understanding the role of PKR activation in placental inflammation and related diseases.


Asunto(s)
Apoptosis , Inflamación , Placenta , Respuesta de Proteína Desplegada , eIF-2 Quinasa , Femenino , Humanos , Embarazo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Inflamación/metabolismo , Lipopolisacáridos , Placenta/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , ARN Bicatenario/metabolismo , Poli I-C/metabolismo
5.
Cytotechnology ; 75(1): 27-38, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36713065

RESUMEN

The unlimited proliferation capacity of embryonic stem cells (ESCs) coupled with their capability to differentiate into several cell types makes them an attractive candidate for studying the molecular mechanisms regulating self-renewal and transition from pluripotent state. Although the roles of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase family (PFKFB1-4) in cell survival, proliferation, and differentiation in tumor cells have been studied, their role in mouse ESC (mESC) biology is currently unkown. In the current study, Pfkfb isoenzyme expressions were analyzed in R1 and J1 mESCs that were cultured in the presence and absence of leukemia inhibitory factor (LIF). We report that expression of the Pfkfb3 isoenzyme was markedly increased when mESCs were promoted to differentiate upon LIF removal. We then demonstrated that Pfkfb3 silencing induced the differentiation marker Brachyury suggesting that Pfkfb3 may be required for the regulation of mesodermal differentiation of mESCs. Furthermore, we show that the increase in Pfkfb3 expression is required for the growth of early differentiated mESCs. Although these results provide important insights into the early differentiation of mESCs with regard to Pfkfb expressions, further mechanistic studies will be needed for understanding the pathways and mechanisms involved in regulation of proliferation and early differentiation of mESCs through Pfkfb3.

6.
Mol Biol Rep ; 50(1): 493-506, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36352179

RESUMEN

BACKGROUND: Gestational diabetes mellitus (GDM) is a metabolic complication that affects millions of pregnant women in the world. Placental tissue function is endangered by hyperglycemia during GDM, which is correlated to increased incidences of pregnancy complications. Recently we showed that due to a significant decrease in mitochondrial fusion, mitochondrial dynamics equilibrium is altered in placental tissues from GDM patients. Evidence for the role of reduced mitochondrial fusion in the disruption of mitochondrial function in placental cells is limited. METHODS AND RESULTS: Here we show that chemical inhibition of mitochondrial fission in cultured placental trophoblast cells leads to an increase in mitochondrial fusion and improves the physiological state of these cells and hence, their capacity to cope in a hyperglycemic environment. Specifically, mitochondrial fission inhibition led to a reduction in reactive oxygen species (ROS) generation, mitochondrial unfolded protein marker expressions, and mitochondrial depolarization. It supported the increase in mitochondrial antioxidant enzyme expressions as well. Mitochondrial fission inhibition also increases the placental cell insulin sensitivity during hyperglycemia. CONCLUSION: Our results suggest that mitochondrial fusion/fission equilibrium is critical for placental cell function and signify the therapeutic potential of small molecule inhibitors of fission during GDM.


Asunto(s)
Diabetes Gestacional , Hiperglucemia , Embarazo , Femenino , Humanos , Placenta/metabolismo , Trofoblastos/metabolismo , Dinámicas Mitocondriales , Insulina/metabolismo , Diabetes Gestacional/metabolismo , Hiperglucemia/metabolismo
7.
Turk J Med Sci ; 53(6): 1565-1573, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38813509

RESUMEN

Background/aim: Type 1 diabetes mellitus (T1DM) is caused by the autoimmune-mediated destruction of insulin-producing cells (IPCs) and still has no effective cure. Better understanding of the molecular mechanisms involved in the differentiation of embryonic stem cells (ESCs) into IPCs may help us improve the therapeutic strategies for treating T1DM. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (Pfkfb1-4) are key regulators of glucose metabolism. Although Pfkfb3 has been shown to be required for the growth of early differentiated mouse ESCs (mESCs), more studies are needed to further assess the roles of Pfkfb isoenzymes in embryonic development and differentiation, particularly into specific cell types. In this study, we aimed to elucidate the changes in the expression of Pfkfb isoenzymes on the differentiation of mESCs into IPCs. Materials and methods: A 3-step protocol was used to differentiate R1 and J1 mESCs into IPCs. The changes in the gene expression of MafA, MafB, Ins2, and Nkx6.1 (IPC specific markers) and Pfkfb1-4 were analyzed using real-time quantitative polymerase chain reaction (qPCR). Insulin expression and secretion were determined by immunofluorescence (IF) staining and the enzyme linked immunosorbent assay (ELISA), respectively. Results: Upon differentiation, the IPC specific markers in differentiated cells were upregulated. Continued differentiation was confirmed by the development of insulin-positive islet-like clusters that secreted insulin in response to glucose uptake. Expressions of the Pfkfb2 and Pfkfb3 isoenzymes were markedly increased in various stages of differentiation. Conclusion: These findings suggest that Pfkfb2 and Pfkfb3 may impact the differentiation of mESCs into IPCs and the regulation of the insulin response to glucose levels. This study also lays a foundation for researchers to further probe the roles of Pfkfb isoenzymes on the differentiation of mESCs into IPCs and may open new avenues for regenerative medicine.


Asunto(s)
Diferenciación Celular , Isoenzimas , Células Madre Embrionarias de Ratones , Fosfofructoquinasa-2 , Animales , Ratones , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/enzimología , Isoenzimas/metabolismo , Isoenzimas/genética , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Fosfofructoquinasa-2/metabolismo , Fosfofructoquinasa-2/genética
8.
Mol Biol Rep ; 49(11): 10195-10204, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36053282

RESUMEN

BACKGROUND: Transforming Growth Factor ß (TGFß) proteins are potent inducers of the epithelial-mesenchymal transition (EMT) in tumor cells. Although mitogen-activated protein kinase (MAPK) family has been shown to be involved in TGFß-induced EMT, role of Dual Specificity Phosphatases (DUSP), key regulators of MAPK activity, in TGFß-induced EMT is largely unkonwn. METHODS AND RESULTS: Real-time qPCR analyses were performed to determine the effect of TGFß1 on expression of EMT genes and DUSP proteins in the non-small cell lung cancer model A549 and pancreatic adenocarcinoma model PANC1 cells. Western blot analyses were conducted to study the changes in protein levels of EMT proteins and select DUSP proteins, as well as phosphorylations of MAPK proteins upon TGFß1 stimulation. Small interfering RNA (siRNA) was utilized to reduce expressions of DUSP genes. We observed that the EMT phenotype coincided with increases in phosphorylations of the MAPK proteins ERK1/2, p38MAPK, and JNK upon TGFß1 stimulation. Real-time qPCR analysis showed increases in DUSP15 and DUSP26 mRNA levels and Western blot analysis confirmed the increase in DUSP26 protein levels in both A549 and PANC1 cells treated with TGFß1 relative to control. Silencing of DUSP26 expression by siRNA markedly suppressed the effect of TGFß1 on E-cadherin and mesenchymal genes in the cells. CONCLUSIONS: Data provided suggest that TGFß1 modulates the expression of DUSP genes and that upregulation of DUSP26 may be required for TGFß1-promoted EMT in A549 and PANC1 cells. Further studies should be carried out to elucidate the requirement of individual DUSPs in TGFß1-associated EMT in tumor cells.


Asunto(s)
Adenocarcinoma , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias Pancreáticas , Humanos , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación hacia Arriba , ARN Interferente Pequeño/farmacología , Neoplasias Pulmonares/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/farmacología , Células A549 , Línea Celular Tumoral
9.
Biochem Biophys Res Commun ; 606: 35-41, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35338857

RESUMEN

Transforming growth factor-beta (TGFß) proteins induce an epithelial-mesenchymal transition (EMT) programme that is associated with increased invasive and drug-resistant phenotype of carcinoma cells. In addition to the canonical pathway involving SMAD proteins, the mitogen-activated kinase (MAPK) pathway via extracellular signal-regulated kinases ½ (ERK1/2) is also involved in promoting and maintaining a mesenchymal phenotype by tumor cells following TGFß signal activation. As dual-specificity phosphatases (DUSPs) regulate ERK1/2 activity by dephosphorylation, we aimed to examine DUSPs' expression upon TGFß stimulation and whether DUSPs play a role in the EMT and related phenotypes promoted by TGFß1 in A549 cells. We found that TGFß1 stimulation led to marked changes in several DUSP proteins, including significant decreases in DUSP4 and DUSP13 expressions. We then showed that the ectopic co-expression of DUSP4/13 suppresses TGFß1-induced ERK1/2 phosphorylation and protein levels of the EMT transcription factors Snail and Slug proteins. We then demonstrated that DUSP4/13 co-expression partially inhibited TGFß1-promoted migration, invasion, and chemoresistance in A549 cells. Collectively, this report provides data for the involvement of DUSP4/13 in malignant phenotypes regulated by TGFß1 in A549 cells.


Asunto(s)
Movimiento Celular , Resistencia a Antineoplásicos , Fosfatasas de Especificidad Dual , Transición Epitelial-Mesenquimal , Factor de Crecimiento Transformador beta1 , Células A549 , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos , Factor de Crecimiento Transformador beta1/farmacología
10.
Mol Biol Rep ; 49(2): 1077-1083, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34773180

RESUMEN

BACKGROUND: The accumulation of excess glutamate in the synapse leads to excitotoxicity, which is the underlying reason of neuronal death in intracranial tumors. METHODS AND RESULTS: We identified the expression levels of glutamate dehydrogenase, glutamine synthetase and sirtuin 4 in U87 cell line and various intracranial tumors. mRNA expressions of glutamate dehydrogenase (GDH), glutamine synthetase (GS) and sirtuin 4 (SIRT4) were analyzed in various intracranial tumors using qPCR. GDH, GS and SIRT4 protein expressions were analyzed in glioblastoma (U87) and glial (IHA-immortalized human astrocytes) cell lines via western blotting. The protein expressions of SIRT4 and GS were shown to be elevated and GDH protein expression was reduced in U87 cells in comparison to IHA cells. All types of intracranial tumors displayed lower GS mRNA expressions compared to controls. SIRT4 mRNA expressions were also shown to be lower in all the tumors and grades, although not significantly. GDH mRNA expression was found to be similar in all groups. CONCLUSION: The molecular mechanisms of glutamate metabolism and excitotoxicity should be discovered to develop therapies against intracranial tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Adolescente , Adulto , Anciano , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular , Niño , Preescolar , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/metabolismo , Glutamato Deshidrogenasa/genética , Glutamato-Amoníaco Ligasa/genética , Ácido Glutámico/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Neuroglía/metabolismo , Estudios Retrospectivos , Sirtuinas/genética
11.
Placenta ; 115: 106-114, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34600274

RESUMEN

INTRODUCTION: Gestational diabetes mellitus (GDM) poses a risk factor for fetal mortality and morbidity by directly affecting the placenta and fetus. Mitochondria are dynamic organelles that play a key role in energy production and conversion in placental tissue. Mitochondrial fusion and fission proteins are important in terms of providing mitochondrial dynamics, the adaptation of the cell to different conditions, and maintaining the metabolic stability of the cells. Although GDM shares many features with Type 2 diabetes mellitus (T2DM), different effects of these conditions on the mother and the child suggest that GDM may have specific pathological effects on placental cells. The aim of this study is to investigate the expression of mitochondrial dynamics, and mitochondrial protein folding markers in placentas from GDM patients and women with pre-existing diabetes mellitus. METHODS: Placentas were properly collected from women, who had pre-existing diabetes (Pre-DM), from women with gestational diabetes mellitus (GDM) and from healthy (non-diabetic) pregnant women. Levels of mitochondrial fusion markers were determined in these placentas by real time quantitative PCR and Western blot experiments. RESULTS: mRNA expressions and protein levels of mitochondrial fusion markers, mitofusin 1, mitofusin 2 (MFN1 and MFN2) and optical atrophy 1 (OPA1) proteins were found to be significantly lower in both Pre-DM placentas and those with GDM compared to healthy (non-diabetic) control group. Likewise, proteins involved in mitochondrial protein folding were also found to be significantly reduced compared to control group. DISCUSSION: Diabetes during pregnancy leads to processes that correlate with mitochondria dysfunction in placenta. Our results showed that mitochondrial fusion markers significantly decrease in placental tissue of women with GDM, compared to the healthy non-diabetic women. The decrease in mitochondrial fusion markers was more severe during GDM compared to the Pre-DM. Our results suggest that there may be differences in the pathophysiology of these conditions.


Asunto(s)
Diabetes Gestacional/metabolismo , Expresión Génica/fisiología , Dinámicas Mitocondriales/fisiología , Proteínas Mitocondriales/genética , Placenta/metabolismo , Adulto , Índice de Masa Corporal , Femenino , GTP Fosfohidrolasas/genética , Humanos , Proteínas de Transporte de Membrana Mitocondrial/genética , Obesidad/complicaciones , Obesidad/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , ARN Mensajero/análisis
13.
Biochem Biophys Res Commun ; 571: 118-124, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34325126

RESUMEN

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


Asunto(s)
Antineoplásicos/farmacología , Bencenoacetamidas/farmacología , Glutaminasa/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Fosfofructoquinasa-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Tiadiazoles/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales , Glutaminasa/genética , Glutaminasa/metabolismo , Humanos , Mutación , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
14.
Cell Metab ; 33(2): 319-333.e6, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33340456

RESUMEN

Endoplasmic reticulum stress (ERS) has a pathophysiological role in obesity-associated insulin resistance. Yet, the coordinated tissue response to ERS remains unclear. Increased connexin 43 (Cx43)-mediated intercellular communication has been implicated in tissue-adaptive and -maladaptive response to various chronic stresses. Here, we demonstrate that in hepatocytes, ERS results in increased Cx43 expression and cell-cell coupling. Co-culture of ER-stressed "donor" cells resulted in intercellular transmission of ERS and dysfunction to ERS-naive "recipient" cells ("bystander response"), which could be prevented by genetic or pharmacologic suppression of Cx43. Hepatocytes from obese mice were able to transmit ERS to hepatocytes from lean mice, and mice lacking liver Cx43 were protected from diet-induced ERS, insulin resistance, and hepatosteatosis. Taken together, our results indicate that in obesity, the increased Cx43-mediated cell-cell coupling allows intercellular propagation of ERS. This novel maladaptive response to over-nutrition exacerbates the tissue ERS burden, promoting hepatosteatosis and impairing whole-body glucose metabolism.


Asunto(s)
Hepatocitos/metabolismo , Obesidad/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Conexina 43/deficiencia , Conexina 43/metabolismo , Estrés del Retículo Endoplásmico , Femenino , Humanos , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
15.
J Neurosci Methods ; 348: 109014, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33309587

RESUMEN

BACKGROUND: Presence of multimodal brain graphs derived from different neuroimaging modalities is inarguably one of the most critical challenges in building unified classification models that can be trained and tested on any brain graph regardless of its size and the modality it was derived from. EXISTING METHODS: One solution is to learn a model for each modality independently, which is cumbersome and becomes more time-consuming as the number of modalities increases. Another traditional solution is to build a model inputting multimodal brain graphs for the target prediction task; however, this is only applicable to datasets where all samples have joint neuro-modalities. NEW METHOD: In this paper, we propose to build a unified brain graph classification model trained on unpaired multimodal brain graphs, which can classify any brain graph of any size. This is enabled by incorporating a graph alignment step where all multi-modal graphs of different sizes and heterogeneous distributions are mapped to a common template graph. Next, we design a graph alignment strategy to the target fixed-size template and further apply linear discriminant analysis (LDA) to the aligned graphs as a supervised dimensionality reduction technique for the target classification task. RESULTS: We tested our method on unpaired autistic and healthy brain connectomes derived from functional and morphological MRI datasets (two modalities). CONCLUSION: Our results showed that our unified model method not only has great promise in solving such a challenging problem but achieves comparable performance to models trained on each modality independently.


Asunto(s)
Trastorno Autístico , Conectoma , Encéfalo/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Neuroimagen
16.
J BUON ; 25(4): 2051-2058, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33099952

RESUMEN

PURPOSE: The purpose of our study was to investigate the mRNA expression profile of glutamate transporter 1 (GLT-1) in different types and grades of brain tumors, such as glioblastoma multiforme, astrocytomas (pilocytic, diffuse, anaplastic), oligodendrogliomas, ependydomas, medulloblastomas, and meningiomas using Real Time Quantitative PCR technique (qRT-PCR). METHODS: A total of 66 surgically removed primary brain tumors were collected retrospectively and the total RNA was isolated from each tumor sample. cDNA was generated and GLT-1 mRNA expression was evaluated with quantitative qRT-PCR. RESULTS: The mRNA expression of GLT-1 was significantly lower in primary brain tumors when compared to control brain tissues. GLT-1 expression was inversely correlated with the tumor grade, implicating its potential role in tumor progression. GLT-1 mRNA expression was lowest in grade 4 tumors, such as glioblastoma multiforme and medulloblastomas. The tumors with grade 3 and 4 combined displayed lower expression compared to tumors with grades 1 and 2. In grade 4 tumors, female patients displayed lower GLT-1 expression compared to male patients. In addition, glioblastoma multiforme patients older than 65 years of age showed lower GLT-1 expression when compared to the patients younger than 65. CONCLUSION: qRT-PCR was found to be a sensitive method in detecting GLT-1 expression in brain tumors. This study may lay the foundation for the future research about the excitotoxicity and brain tumors and GLT-1 might be a potential biomarker. Targeted therapies based on excitotoxic molecular pathways against gliomas should be designed to effectively combat these diseases.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Glioblastoma/metabolismo , Adulto , Factores de Edad , Anciano , Neoplasias Encefálicas/patología , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Estudios Retrospectivos , Factores Sexuales , Adulto Joven
17.
Mol Cell Biochem ; 470(1-2): 115-129, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32415418

RESUMEN

Tumor cells increase glucose metabolism through glycolysis and pentose phosphate pathways to meet the bioenergetic and biosynthetic demands of rapid cell proliferation. The family of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1-4) are key regulators of glucose metabolism via their synthesis of fructose-2,6-bisphosphate (F2,6BP), a potent activator of glycolysis. Previous studies have reported the co-expression of PFKFB isozymes, as well as the mRNA splice variants of particular PFKFB isozymes, suggesting non-redundant functions. Majority of the evidence demonstrating a requirement for PFKFB activity in increased glycolysis and oncogenic properties in tumor cells comes from studies on PFKFB3 and PFKFB4 isozymes. In this study, we show that the PFKFB2 isozyme is expressed in tumor cell lines of various origin, overexpressed and localizes to the nucleus in pancreatic adenocarcinoma, relative to normal pancreatic tissue. We then demonstrate the differential intracellular localization of two PFKFB2 mRNA splice variants and that, when ectopically expressed, cytoplasmically localized mRNA splice variant causes a greater increase in F2,6BP which coincides with an increased glucose uptake, as compared with the mRNA splice variant localizing to the nucleus. We then show that PFKFB2 expression is required for steady-state F2,6BP levels, glycolytic activity, and proliferation of pancreatic adenocarcinoma cells. In conclusion, this study may provide a rationale for detailed investigation of PFKFB2's requirement for the glycolytic and oncogenic phenotype of pancreatic adenocarcinoma cells.


Asunto(s)
Adenocarcinoma/enzimología , Glucólisis , Páncreas/enzimología , Neoplasias Pancreáticas/enzimología , Fosfofructoquinasa-2/fisiología , Adenocarcinoma/patología , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Citoplasma/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/fisiología , Neoplasias Pancreáticas/patología , Fenotipo , Fosfofructoquinasa-2/genética , Empalme del ARN , ARN Mensajero/metabolismo
18.
Turk J Biol ; 44(2): 93-102, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32256145

RESUMEN

Type 2 diabetes mellitus is characterized by insulin resistance and hypersecretion of insulin from the pancreas to compensate for decreased insulin sensitivity in the peripheral tissues. In later stages of the disease insulin-secreting beta cell degeneration commences and patients require insulin replacement therapy in order to accomplish proper regulation of their blood glucose. Endoplasmic reticulum (ER) stress in the beta cells is one of the factors contributing to this detrimental effect. Protein kinase R (PKR) is a cellular stress kinase activated by ER stress and contributing to degeneration of pancreatic islets. In order to determine whether inhibition of PKR activation by specific small molecule inhibitors of PKR ameliorates pancreatic insulin secretion capacity, we treated beta cells with two imidazole/oxindole-derived inhibitors of PKR kinase, imoxin (C16) and 2-aminopurine (2-AP), in the presence of ER stress. Our results demonstrate that PKR inhibition suppresses tunicamycin-mediated ER stress without altering the insulin production capacity of the cells. Palmitic acid-mediated suppression of insulin secretion, however, was subdued significantly by PKR inhibitor treatment through an ER stress-related mechanism. We suggest that PKR inhibitor treatment may be used to increase the insulin secretion capacity of the pancreas in later stages of diabetes.

19.
J BUON ; 24(2): 779-790, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31128036

RESUMEN

PURPOSE: Gastroenteropancreatic tumors (GEPNETs) is a heterogeneous disease with variable clinical course. While promising therapeutic options exist for other adult cancers, there are no new molecular-based treatments developed for GEPNETs. One of the main targets of cancer immunotherapy is the Programmed Cell Death Ligand-1 (PD-L1) pathway. Our purpose was to investigate the profile of PD-L1 expression in different organs of GEPNETs and compare the conventional immunohistochemistry (IHC) with the RNA expression analysis via real time polymerase chain reaction (RT-PCR) in order to determine which patients might be appropriate for immune check point-targeted therapy. METHODS: A total of 59 surgically or endoscopically resected GEPNET tissues were retrospectively collected. The expression of PD-L1 and mRNA was evaluated with IHC. RESULTS: The expression of PD-L1 was significantly associated with the high-grade classification (p=0.012). PD-L1 mRNA expression in tumor samples appeared to be higher compared to the corresponding normal tissues. In appendix, stomach and small intestine, the expression of PD-L1 mRNA was higher in the tumor tissues compared to the respective controls. In pancreas and colon, control tissues tend to have a higher PD-L1 mRNA expression compared to tumor tissues. PD-L1 mRNA expression was higher in GEP carcinomas (p=0.0031). CONCLUSION: RT-PCR was found to be more sensitive in detecting PD-L1 expression than conventional IHC. This study may provide an important starting point and useful background information for future research about immunotherapy for appendix, stomach and small intestine neuroendocrine carcinomas.


Asunto(s)
Antígeno B7-H1/genética , Carcinoma Neuroendocrino/genética , Inmunohistoquímica , Neoplasias Intestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Neoplasias Gástricas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/patología , Carcinoma Neuroendocrino/terapia , Colon/metabolismo , Femenino , Mucosa Gástrica/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Neoplasias Intestinales/terapia , Intestino Delgado/metabolismo , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/inmunología , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/terapia , Páncreas/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , ARN Mensajero/genética , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Adulto Joven
20.
Case Rep Obstet Gynecol ; 2018: 9719374, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30050708

RESUMEN

BACKGROUND: Niemann-Pick Disease Type B (NPD B) is a rare lysosomal storage disorder resulting from an inherited deficiency of acid sphingomyelinase activity. Here, we report the case of a splenectomized patient with NPD B who died because of severe postpartum hemorrhage (PPH). CASE PRESENTATION: A 23-year-old nulliparous woman was admitted to intensive care unit (ICU) after cardiopulmonary arrest during urgent hysterectomy because of severe postpartum bleeding. The patient concealed her disease from her family and obstetricians during her pregnancy, and her NPD B diagnosis was revealed during her stay in ICU while searching for the cause of the splenectomy and severe bleeding. Unfortunately, she had a detrimental course with hypoxic brain injury leading to brain death. CONCLUSIONS: In conclusion, physicians should keep in mind that patients with a history of splenectomy and/or uncontrollable hemorrhage must be carefully evaluated for rare diseases like lysosomal storage diseases and that NPD B can cause mortality because of postpartum bleeding. Adult intensivists should be familiar with adult presentations of rare metabolic or genetic diseases as more and more children with metabolic or genetic diseases will survive to adulthood and will be admitted to and unfortunately will even die in the adult ICU.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...