Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Research (Wash D C) ; 7: 0365, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38654733

RESUMEN

Neutrophils are primed for neutrophil extracellular trap (NET) formation during diabetes, and excessive NET formation from primed neutrophils compromises wound healing in patients with diabetes. Here, we demonstrate that trained immunity mediates diabetes-induced NET priming in neutrophils. Under diabetic conditions, neutrophils exhibit robust metabolic reprogramming comprising enhanced glycolysis via the pentose phosphate pathway and fatty acid oxidation, which result in the accumulation of acetyl-coenzyme A. Adenosine 5'-triphosphate-citrate lyase-mediated accumulation of acetyl-coenzyme A and histone acetyltransferases further induce the acetylation of lysine residues on histone 3 (AcH3K9, AcH3K14, and AcH3K27) and histone 4 (AcH4K8). The pharmacological inhibition of adenosine 5'-triphosphate-citrate lyase and histone acetyltransferases completely inhibited high-glucose-induced NET priming. The trained immunity of neutrophils was further confirmed in neutrophils isolated from patients with diabetes. Our findings suggest that trained immunity mediates functional changes in neutrophils in diabetic environments, and targeting neutrophil-trained immunity may be a potential therapeutic target for controlling inflammatory complications of diabetes.

2.
Exp Mol Med ; 56(5): 1123-1136, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38689086

RESUMEN

Tumor-associated macrophages (TAMs) are vital contributors to the growth, metastasis, and therapeutic resistance of various cancers, including hepatocellular carcinoma (HCC). However, the exact phenotype of TAMs and the mechanisms underlying their modulation for therapeutic purposes have not been determined. Here, we present compelling evidence that glutamine-derived aspartate in TAMs stimulates spermidine production through the polyamine synthesis pathway, thereby increasing the translation efficiency of HIF-1α via eIF5A hypusination. Consequently, augmented translation of HIF-1α drives TAMs to undergo an increase glycolysis and acquire a metabolic phenotype distinct from that of M2 macrophages. Finally, eIF5A levels in tumor stromal lesions were greater than those in nontumor stromal lesions. Additionally, a higher degree of tumor stromal eIF5A hypusination was significantly associated with a more advanced tumor stage. Taken together, these data highlight the potential of inhibiting hypusinated eIF5A by targeting glutamine metabolism in TAMs, thereby opening a promising avenue for the development of novel therapeutic approaches for HCC.


Asunto(s)
Ácido Aspártico , Carcinoma Hepatocelular , Factor 5A Eucariótico de Iniciación de Traducción , Glutamina , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias Hepáticas , Factores de Iniciación de Péptidos , Proteínas de Unión al ARN , Macrófagos Asociados a Tumores , Factores de Iniciación de Péptidos/metabolismo , Factores de Iniciación de Péptidos/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Humanos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Glutamina/metabolismo , Ácido Aspártico/metabolismo , Ácido Aspártico/análogos & derivados , Biosíntesis de Proteínas , Animales , Línea Celular Tumoral , Ratones , Glucólisis , Lisina/análogos & derivados
3.
Biomedicines ; 12(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38540172

RESUMEN

Cell death is crucial for maintaining tissue balance and responding to diseases. However, under pathological conditions, the surge in dying cells results in an overwhelming presence of cell debris and the release of danger signals. In the liver, this gives rise to hepatic inflammation and hepatocellular cell death, which are key factors in various liver diseases caused by viruses, toxins, metabolic issues, or autoimmune factors. Both clinical and in vivo studies strongly affirm that hepatocyte death serves as a catalyst in the progression of liver disease. This advancement is characterized by successive stages of inflammation, fibrosis, and cirrhosis, culminating in a higher risk of tumor development. In this review, we explore pivotal forms of cell death, including apoptosis, pyroptosis, and necroptosis, examining their roles in both acute and chronic liver conditions, including liver cancer. Furthermore, we discuss the significance of cell death in liver surgery and ischemia-reperfusion injury. Our objective is to illuminate the molecular mechanisms governing cell death in liver diseases, as this understanding is crucial for identifying therapeutic opportunities aimed at modulating cell death pathways.

4.
Inflamm Res ; 72(10-11): 1981-1997, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37770568

RESUMEN

BACKGROUND: Classically activated M1 macrophages, characterized by aberrant glycolysis and secretion of inflammatory cytokines, play pivotal roles in inflammatory diseases, including inflammatory bowel disease (IBD). Recently, sodium-glucose co-transporter 2 (SGLT2) inhibitors were shown to suppress Na+/H+ exchanger 1 (NHE1) and Na+/Ca2+ exchanger 1 (NCX1) activity, regulating downstream intracellular Ca2+ concentrations in cardiomyocytes. However, whether SGLT2 inhibitors regulate M1 macrophage polarization by downregulating NHE1 and NCX1 remains unknown. METHODS: We analyzed cellular responses to SGLT2 inhibitors using mouse bone marrow-derived macrophages and peritoneal macrophages treated with lipopolysaccharide (LPS). To induce IBD, we used a dextran sulfate sodium salt-induced colitis mouse model. RESULTS: We observed that NHE1 and NCX1 were overexpressed in LPS-treated macrophages, leading to M1 macrophage polarization. Mechanistically, NHE1 and NCX1-mediated Ca2+ accumulation in the macrophage resulted in enhanced glycolysis by promoting PI3K/AKT/mTORC1 signaling. SGLT2 inhibitors suppressed both the expression levels and activities of NHE1 and NCX1, and consequently downregulated PI3K/AKT/mTORC1 signaling and glycolysis in LPS-treated macrophages. We observed inhibition of LPS-stimulated M1 polarization and cytokine production by SGLT2 inhibitors in vitro, ex vivo, and in an IBD mouse model. CONCLUSIONS: NHE1 promotes M1 macrophage polarization and SGLT2 inhibitors are a novel strategy to treat M1 macrophage-mediated inflammatory diseases, including IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Animales , Ratones , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Macrófagos/metabolismo , Modelos Animales de Enfermedad , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
5.
Exp Mol Med ; 55(4): 706-715, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37009798

RESUMEN

Proliferating cancer cells rely largely on glutamine for survival and proliferation. Glutamine serves as a carbon source for the synthesis of lipids and metabolites via the TCA cycle, as well as a source of nitrogen for amino acid and nucleotide synthesis. To date, many studies have explored the role of glutamine metabolism in cancer, thereby providing a scientific rationale for targeting glutamine metabolism for cancer treatment. In this review, we summarize the mechanism(s) involved at each step of glutamine metabolism, from glutamine transporters to redox homeostasis, and highlight areas that can be exploited for clinical cancer treatment. Furthermore, we discuss the mechanisms underlying cancer cell resistance to agents that target glutamine metabolism, as well as strategies for overcoming these mechanisms. Finally, we discuss the effects of glutamine blockade on the tumor microenvironment and explore strategies to maximize the utility of glutamine blockers as a cancer treatment.


Asunto(s)
Glutamina , Neoplasias , Humanos , Glutamina/metabolismo , Neoplasias/metabolismo , Aminoácidos/metabolismo , Ciclo del Ácido Cítrico , Oxidación-Reducción , Microambiente Tumoral
6.
Diabetes Metab J ; 47(3): 426-436, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36872065

RESUMEN

BACKGROUND: The Chinese visceral adiposity index (CVAI) and new visceral adiposity index (NVAI) are novel indices of visceral adiposity used to predict metabolic and cardiovascular diseases in Asian populations. However, the relationships of CVAI and NVAI with chronic kidney disease (CKD) have not been investigated. We aimed to characterize the relationships of CVAI and NVAI with the prevalence of CKD in Korean adults. METHODS: A total of 14,068 participants in the 7th Korea National Health and Nutrition Examination Survey (6,182 men and 7,886 women) were included. Receiver operating characteristic (ROC) analyses were employed to compare the associations between indices of adiposity and CKD, and a logistic regression model was used to characterize the relationships of CVAI and NVAI with CKD prevalence. RESULTS: The areas under the ROC curves for CVAI and NVAI were significantly larger than for the other indices, including the visceral adiposity index and lipid accumulation product, in both men and women (all P<0.001). In addition, high CVAI or NVAI was significantly associated with a high CKD prevalence in both men (odds ratio [OR], 2.14; 95% confidence interval [CI], 1.31 to 3.48 in CVAI and OR, 6.47; 95% CI, 2.91 to 14.38 in NVAI, P<0.05) and women (OR, 4.87; 95% CI, 1.85 to 12.79 in CVAI and OR, 3.03; 95% CI, 1.35 to 6.82 in NVAI, P<0.05); this association remained significant after adjustment for multiple confounding factors in men and women. CONCLUSION: CVAI and NVAI are positively associated with CKD prevalence in a Korean population. CVAI and NVAI may be useful for the identification of CKD in Asian populations, including in Korea.


Asunto(s)
Adiposidad , Insuficiencia Renal Crónica , Masculino , Humanos , Adulto , Femenino , Encuestas Nutricionales , Pueblo Asiatico , Obesidad Abdominal/complicaciones , Obesidad Abdominal/epidemiología , Obesidad Abdominal/diagnóstico , Insuficiencia Renal Crónica/epidemiología , Insuficiencia Renal Crónica/complicaciones , República de Corea/epidemiología
7.
Cell Biosci ; 13(1): 69, 2023 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-36998073

RESUMEN

BACKGROUND: N-methyl-D-aspartate receptors (NMDARs) are considered to be involved in several physiological and pathophysiological processes in addition to the progression of neurological disorders. However, how NMDARs are involved in the glycolytic phenotype of M1 macrophage polarization and the possibility of using them as a bio-imaging probe for macrophage-mediated inflammation remain unclear. METHODS: We analyzed cellular responses to NMDAR antagonism and small interfering RNAs using mouse bone marrow-derived macrophages (BMDMs) treated with lipopolysaccharide (LPS). An NMDAR targeting imaging probe, N-TIP, was produced via the introduction of NMDAR antibody and the infrared fluorescent dye FSD Fluor™ 647. N-TIP binding efficiency was tested in intact and LPS-stimulated BMDMs. N-TIP was intravenously administered to mice with carrageenan (CG)- and LPS-induced paw edema, and in vivo fluorescence imaging was conducted. The anti-inflammatory effects of dexamethasone were evaluated using the N-TIP-mediated macrophage imaging technique. RESULTS: NMDARs were overexpressed in LPS-treated macrophages, subsequently inducing M1 macrophage polarization. Mechanistically, NMDAR-mediated Ca2+ accumulation resulted in LPS-stimulated glycolysis via upregulation of PI3K/AKT/mTORC1 signaling. In vivo fluorescence imaging with N-TIP showed LPS- and CG-induced inflamed lesions at 5 h post-inflammation, and the inflamed lesions could be detected until 24 h. Furthermore, our N-TIP-mediated macrophage imaging technique helped successfully visualize the anti-inflammatory effects of dexamethasone in mice with inflammation. CONCLUSION: This study demonstrates that NMDAR-mediated glycolysis plays a critical role in M1 macrophage-related inflammation. Moreover, our results suggest that NMDAR targeting imaging probe may be useful in research on inflammatory response in vivo.

8.
Endocrinol Metab (Seoul) ; 37(5): 800-809, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36168774

RESUMEN

BACKGRUOUND: Excessive proliferation and migration of vascular smooth muscle cells (VSMCs), which contributes to the development of occlusive vascular diseases, requires elevated mitochondrial oxidative phosphorylation to meet the increased requirements for energy and anabolic precursors. Therefore, therapeutic strategies based on blockade of mitochondrial oxidative phosphorylation are considered promising for treatment of occlusive vascular diseases. Here, we investigated whether DN200434, an orally available estrogen receptor-related gamma inverse agonist, inhibits proliferation and migration of VSMCs and neointima formation by suppressing mitochondrial oxidative phosphorylation. METHODS: VSMCs were isolated from the thoracic aortas of 4-week-old Sprague-Dawley rats. Oxidative phosphorylation and the cell cycle were analyzed in fetal bovine serum (FBS)- or platelet-derived growth factor (PDGF)-stimulated VSMCs using a Seahorse XF-24 analyzer and flow cytometry, respectively. A model of neointimal hyperplasia was generated by ligating the left common carotid artery in male C57BL/6J mice. RESULTS: DN200434 inhibited mitochondrial respiration and mammalian target of rapamycin complex 1 activity and consequently suppressed FBS- or PDGF-stimulated proliferation and migration of VSMCs and cell cycle progression. Furthermore, DN200434 reduced carotid artery ligation-induced neointima formation in mice. CONCLUSION: Our data suggest that DN200434 is a therapeutic option to prevent the progression of atherosclerosis.


Asunto(s)
Aterosclerosis , Neointima , Ratas , Ratones , Masculino , Animales , Neointima/prevención & control , Neointima/tratamiento farmacológico , Neointima/metabolismo , Músculo Liso Vascular/metabolismo , Ratones Endogámicos C57BL , Proliferación Celular , Ratas Sprague-Dawley , Células Cultivadas , Arteria Carótida Común/metabolismo , Arterias Carótidas/cirugía , Arterias Carótidas/metabolismo , Mamíferos
9.
BMB Rep ; 55(11): 547-552, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36016501

RESUMEN

Sorafenib, originally identified as an inhibitor of multiple oncogenic kinases, induces ferroptosis in hepatocellular carcinoma (HCC) cells. Several pathways that mitigate sorafenib-induced ferroptosis confer drug resistance; thus strategies that enhance ferroptosis increase sorafenib efficacy. Orphan nuclear receptor estrogen-related receptor γ (ERRγ) is upregulated in human HCC tissues and plays a role in cancer cell proliferation. The aim of this study was to determine whether inhibition of ERRγ with DN200434, an orally available inverse agonist, can overcome resistance to sorafenib through induction of ferroptosis. Sorafenib-resistant HCC cells were less sensitive to sorafenibinduced ferroptosis and showed significantly higher ERRγ levels than sorafenib-sensitive HCC cells. DN200434 induced lipid peroxidation and ferroptosis in sorafenib-resistant HCC cells. Mechanistically, DN200434 increased mitochondrial ROS generation by reducing glutathione/glutathione disulfide levels, which subsequently reduced mTOR activity and GPX4 levels. DN200434 induced amplification of the antitumor effects of sorafenib was confirmed in a tumor xenograft model. The present results indicate that DN200434 may be a novel therapeutic strategy to re-sensitize HCC cells to sorafenib. [BMB Reports 2022; 55(11): 547-552].


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Sorafenib/farmacología , Sorafenib/uso terapéutico , Neoplasias Hepáticas/metabolismo , Estrógenos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos
10.
BMB Rep ; 55(9): 459-464, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35651333

RESUMEN

Various mechanisms have been suggested to explain the chemopreventive and tumor-inhibitory effects of melatonin. Despite the growing evidence supporting melatonin-induced mitochondrial dysfunction, it remains largely unknown how this phenomenon modulates metabolic reprogramming in cancer cells. The aim of our study was to identify the mechanism underlying the anti-proliferative and apoptotic effects of melatonin, which is known to inhibit glycolysis. We analyzed the time-dependent effects of melatonin on mitochondrial respiration and glycolysis in liver cancer cells. The results showed that from a cell bioenergetic point of view, melatonin caused an acute reduction in mitochondrial respiration, however, increased reactive oxygen species production, thereby inhibiting mTORC1 activity from an early stage post-treatment without affecting glycolysis. Nevertheless, administration of melatonin for a longer time reduced expression of c-Myc protein, thereby suppressing glycolysis via downregulation of HK2 and LDHA. The data presented herein suggest that melatonin suppresses mitochondrial respiration and glycolysis simultaneously in HCC cells, leading to anti-cancer effects. Thus, melatonin can be used as an adjuvant agent for therapy of liver cancer. [BMB Reports 2022; 55(9): 459-464].


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Melatonina , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Glucólisis , Humanos , Neoplasias Hepáticas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Melatonina/metabolismo , Melatonina/farmacología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Respiración
11.
J Exp Clin Cancer Res ; 41(1): 98, 2022 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-35287706

RESUMEN

BACKGROUND: Macropinocytosis, an important nutrient-scavenging pathway in certain cancer cells, allows cells to compensate for intracellular amino acid deficiency under nutrient-poor conditions. Ferroptosis caused by cysteine depletion plays a pivotal role in sorafenib responses during hepatocellular carcinoma (HCC) therapy. However, it is not known whether macropinocytosis functions as an alternative pathway to acquire cysteine in sorafenib-treated HCC, and whether it subsequently mitigates sorafenib-induced ferroptosis. This study aimed to investigate whether sorafenib drives macropinocytosis induction, and how macropinocytosis confers ferroptosis resistance on HCC cells. METHODS: Macropinocytosis, both in HCC cells and HCC tissues, was evaluated by measuring TMR-dextran uptake or lysosomal degradation of DQ-BSA, and ferroptosis was evaluated via C11-BODIPY fluorescence and 4-HNE staining. Sorafenib-induced ferroptosis and macropinocytosis were validated in tumor tissues taken from HCC patients who underwent ultrasound-guided needle biopsy. RESULTS: Sorafenib increased macropinocytosis in human HCC specimens and xenografted HCC tissues. Sorafenib-induced mitochondrial dysfunction was responsible for activation of PI3K-RAC1-PAK1 signaling, and amplified macropinocytosis in HCC. Importantly, macropinocytosis prevented sorafenib-induced ferroptosis by replenishing intracellular cysteine that was depleted by sorafenib treatment; this rendered HCC cells resistant to sorafenib. Finally, inhibition of macropinocytosis by amiloride markedly enhanced the anti-tumor effect of sorafenib, and sensitized resistant tumors to sorafenib. CONCLUSION: In summary, sorafenib induced macropinocytosis, which conferred drug resistance by mitigating sorafenib-induced ferroptosis. Thus, targeting macropinocytosis is a promising therapeutic strategy to facilitate ferroptosis-based therapy for HCC.


Asunto(s)
Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/tratamiento farmacológico , Cisteína/uso terapéutico , Ferroptosis/efectos de los fármacos , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/tratamiento farmacológico , Pinocitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib/uso terapéutico , Animales , Carcinoma Hepatocelular/patología , Cisteína/farmacología , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Sorafenib/farmacología
12.
Int J Mol Sci ; 23(3)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35163619

RESUMEN

Classically activated M1 macrophages reprogram their metabolism towards enhanced glycolysis to obtain energy and produce pro-inflammatory cytokines after activation by mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor (HIF)-1α. Thus, a strategy that constrains M1 polarization of macrophages via downregulation of glycolysis is essential for treating chronic inflammatory diseases. Cassiae semen has pharmacological activity against various inflammatory diseases. However, it is unclear whether specific compounds within Cassia seeds affect M1 polarization of macrophages. Here, we investigated whether Cassiaside C napthopyrone from Cassiae semen inhibits M1 polarization by downregulating glycolysis. We found that Cassiaside C reduced expression of inducible nitric oxide synthase and cyclooxygenase-2 and the phosphorylation of nuclear factor kappa B, all of which are upregulated in lipopolysaccharide (LPS)/interferon (IFN)-γ-treated Raw264.7 cells and peritoneal macrophages. Moreover, Cassiaside C-treated macrophages showed marked suppression of LPS/IFN-γ-induced HIF-1α, pyruvate dehydrogenase kinase 1, and lactate dehydrogenase A expression, along with downregulation of the phosphoinositide 3-kinases (PI3K)/AKT/mTORC1 signaling pathway. Consequently, Cassiaside C attenuated enhanced glycolysis and lactate production, but rescued diminished oxidative phosphorylation, in M1 polarized macrophages. Thus, Cassiaside C dampens M1 polarization of macrophages by downregulating glycolysis, which could be exploited as a therapeutic strategy for chronic inflammatory conditions.


Asunto(s)
Polaridad Celular , Glucólisis , Glicósidos , Activación de Macrófagos , Macrófagos , Animales , Ratones , Regulación de la Expresión Génica , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Transducción de Señal , Polaridad Celular/efectos de los fármacos , Glicósidos/farmacología
13.
Sci Rep ; 11(1): 22451, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789863

RESUMEN

The present study aimed to investigate changes in glucose metabolism and incretin hormone response following longer intestinal bypass reconstruction after distal gastrectomy (DG) in low BMI patients with gastric cancer and type 2 diabetes. A total of 20 patients were prospectively recruited and underwent either conventional Billroth I (BI), Billroth II with long-biliopancreatic limb (BII), or Roux-en-Y anastomosis with long-Roux limb (RY) after DG. A 75g-oral glucose tolerance test (OGTT) was given preoperatively; and at 5 days, 3 months, and 6 months postoperatively. Serum glucose, insulin, glucagon, glucagon-like peptide-1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) were serially measured. At 6 months after surgery, complete diabetes remission was achieved in 57.1% of the BII group but in no patients in the other two groups (p = 0.018). BII group showed a significant reduction in glucose concentration during OGTT at 6 months in contrast to the other 2 groups. In the BII group, a significant increase in GLP-1 secretion was observed after surgery but not maintained at 6 months, while postoperative hyperglucagonemia was alleviated along with a reduction in GIP. BII gastrojejunostomy with long biliopancreatic limb achieved better diabetes control with favorable incretin response after DG compared to BI or RY reconstruction.


Asunto(s)
Anastomosis en-Y de Roux/métodos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/complicaciones , Gastrectomía/métodos , Derivación Gástrica/métodos , Incretinas/sangre , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/cirugía , Anciano , Glucemia/metabolismo , Índice de Masa Corporal , Femenino , Estudios de Seguimiento , Polipéptido Inhibidor Gástrico/sangre , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Estudios Prospectivos , Resultado del Tratamiento
14.
Endocrinol Metab (Seoul) ; 36(5): 1142-1146, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34674507

RESUMEN

It has been suggested that the coronavirus disease 2019 (COVID-19) pandemic has had a negative impact on glycemic control in patients with type 2 diabetes mellitus (T2DM). However, no study has examined yearly trends in glycated hemoglobin (HbA1c) levels after the start of the COVID-19 outbreak. Here, we performed a retrospective analysis of HbA1c concentrations during the early period of the COVID-19 outbreak (COVID-19 cohort) and then compared the yearly trend in the mean HbA1c level, along with fluctuations in HbA1c levels, with those during previous years (non-COVID-19 cohorts). We observed that the mean HbA1c level in patients with T2DM increased during the first 6 months of the COVID-19 outbreak. After 6 months, HbA1c levels in the COVID-19 cohort returned to levels seen in the non-COVID-19 cohorts. The data suggest that vulnerable patients with T2DM should be monitored closely during the early period of a pandemic to ensure they receive appropriate care.


Asunto(s)
COVID-19/epidemiología , Diabetes Mellitus Tipo 2/sangre , Hemoglobina Glucada/análisis , Control Glucémico/tendencias , Adulto , Glucemia/análisis , COVID-19/diagnóstico , COVID-19/virología , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/epidemiología , Brotes de Enfermedades/estadística & datos numéricos , Femenino , Humanos , Masculino , Estudios Retrospectivos , SARS-CoV-2/genética , Factores de Tiempo
15.
Korean J Intern Med ; 36(4): 942-948, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34092049

RESUMEN

BACKGROUND/AIMS: Coronavirus disease 2019 (COVID-19) is a global pandemic that had affected more than 13,000 people in South Korea by July 2020. To prevent spread of COVID-19, tele-prescription was permitted temporarily. This study investigated the impact of tele-prescription on glycemic control in patients with type 2 diabetes. METHODS: Glycated hemoglobin (HbA1c) concentrations were retrospectively analyzed in patients with type 2 diabetes who were treated with tele-prescription because of COVID-19 and those who were treated by face-to-face care (non-tele-prescription group) enrolled at the same period of time. Mean HbA1c concentrations and mean change in HbA1c concentration (ΔHbA1c) were compared in these two groups. RESULTS: The mean HbA1c levels of patients were significantly higher after than before the tele-prescription period (7.46% ± 1.24% vs. 7.27% ± 1.13%, p < 0.05). Mean ΔHbA1c was significantly higher in the tele-prescription than in the non-tele-prescription group (0.19% ± 0.68% vs. 0.04% ± 0.95%, p < 0.05). HbA1c was significantly greater in patients taking fewer oral hypoglycemic agents, no insulin, fewer comorbidities (e.g., coronary artery disease, cerebrovascular accident, and diabetic neuropathy), and higher baseline HbA1c. CONCLUSION: Tele-prescription may worsen glycemic control in patients with type 2 diabetes during public health crises.


Asunto(s)
COVID-19 , Diabetes Mellitus Tipo 2 , Glucemia , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Hemoglobina Glucada/análisis , Control Glucémico , Humanos , Hipoglucemiantes/uso terapéutico , Prescripciones , República de Corea/epidemiología , Estudios Retrospectivos , SARS-CoV-2
16.
Int J Mol Sci ; 22(11)2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-34070527

RESUMEN

Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) contribute to the development of atherosclerosis and restenosis. Glycolysis and glutaminolysis are increased in rapidly proliferating VSMCs to support their increased energy requirements and biomass production. Thus, it is essential to develop new pharmacological tools that regulate metabolic reprogramming in VSMCs for treatment of atherosclerosis. The effects of 6-diazo-5-oxo-L-norleucine (DON), a glutamine antagonist, have been broadly investigated in highly proliferative cells; however, it is unclear whether DON inhibits proliferation of VSMCs and neointima formation. Here, we investigated the effects of DON on neointima formation in vivo as well as proliferation and migration of VSMCs in vitro. DON simultaneously inhibited FBS- or PDGF-stimulated glycolysis and glutaminolysis as well as mammalian target of rapamycin complex I activity in growth factor-stimulated VSMCs, and thereby suppressed their proliferation and migration. Furthermore, a DON-derived prodrug, named JHU-083, significantly attenuated carotid artery ligation-induced neointima formation in mice. Our results suggest that treatment with a glutamine antagonist is a promising approach to prevent progression of atherosclerosis and restenosis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diazooxonorleucina/farmacología , Glutamina/antagonistas & inhibidores , Glucólisis/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Neointima/tratamiento farmacológico , Fosforilación Oxidativa/efectos de los fármacos , Animales , Antimetabolitos Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Diazooxonorleucina/análogos & derivados , Glutamina/metabolismo , Inmunohistoquímica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Ratas Sprague-Dawley , Albúmina Sérica Bovina/farmacología
17.
Biochem Biophys Res Commun ; 560: 45-51, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-33965788

RESUMEN

Rapidly proliferating cells such as vascular smooth muscle cells (VSMCs) require metabolic programs to support increased energy and biomass production. Thus, targeting glutamine metabolism by inhibiting glutamine transport could be a promising strategy for vascular disorders such as atherosclerosis, stenosis, and restenosis. V-9302, a competitive antagonist targeting the glutamine transporter, has been investigated in the context of cancer; however, its role in VSMCs is unclear. Here, we examined the effects of blocking glutamine transport in fetal bovine serum (FBS)- or platelet-derived growth factor (PDGF)-stimulated VSMCs using V-9302. We found that V-9302 inhibited mTORC1 activity and mitochondrial respiration, thereby suppressing FBS- or PDGF-stimulated proliferation and migration of VSMCs. Moreover, V-9302 attenuated carotid artery ligation-induced neointima in mice. Collectively, the data suggest that targeting glutamine transport using V-9302 is a promising therapeutic strategy to ameliorate occlusive vascular disease.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Neointima/tratamiento farmacológico , Sistema de Transporte de Aminoácidos A/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos A/metabolismo , Animales , Arterias Carótidas/cirugía , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Ligadura , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/etiología , Neointima/patología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas Sprague-Dawley , Albúmina Sérica Bovina/farmacología
18.
Diabetes Metab J ; 45(1): 109-114, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264833

RESUMEN

This study investigated the impact of social distancing due to coronavirus disease 2019 (COVID-19) on glycemic control in people with type 2 diabetes mellitus (T2DM). We retrospectively analyzed the change in glycosylated hemoglobin level (ΔHbA1c) in people with T2DM who undertook social distancing because of COVID-19. We compared the ΔHbA1c between COVID-19 and non-COVID-19 cohorts that were enrolled at the same time of year. The ΔHbA1c of the COVID-19 cohort was significantly higher than that of two non-COVID-19 cohorts. Subgroup analysis according to age and baseline HbA1c level showed that social distancing significantly increased the mean HbA1c level of participants of <50 years. The ΔHbA1c of participants of <50 years and with HbA1c <7.0% in the COVID-19 cohort showed larger changes than other subgroups. In adjusted model, adjusted ΔHbA1c levels in the COVID-19 cohort remained significantly higher than those in the two other cohorts. Social distancing negatively impacts blood glucose control in people with T2DM, especially those who are younger and have good blood glucose control.


Asunto(s)
COVID-19/prevención & control , Diabetes Mellitus Tipo 2/sangre , Hemoglobina Glucada/análisis , Distanciamiento Físico , Anciano , COVID-19/diagnóstico , COVID-19/epidemiología , COVID-19/virología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , República de Corea/epidemiología , Estudios Retrospectivos , SARS-CoV-2/genética
19.
Diabetes Metab J ; 45(2): 260-269, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32662257

RESUMEN

Background: Umbilical cord-mesenchymal stem cell-conditioned medium (UC-MSC-CM) has emerged as a promising cell-free therapy. The aim of this study was to explore the therapeutic effects of UC-MSC-CM on insulin resistance in C2C12 cell. Methods: Insulin resistance was induced by palmitate. Effects of UC-MSC-CM on insulin resistance were evaluated using glucose uptake, glucose transporter type 4 (GLUT4) translocation, the insulin-signaling pathway, and mitochondrial contents and functions in C2C12 cell. Results: Glucose uptake was improved by UC-MSC-CM. UC-MSC-CM treatment increased only in membranous GLUT4 expression, not in cytosolic GLUT4 expression. It restored the insulin-signaling pathway in insulin receptor substrate 1 and protein kinase B. Mitochondrial contents evaluated by mitochondrial transcription factor A, mitochondrial DNA copy number, and peroxisome proliferator-activated receptor gamma coactivator 1-alpha were increased by UC-MSC-CM. In addition, UC-MSC-CM significantly decreased mitochondrial reactive oxygen species and increased fatty acid oxidation and mitochondrial membrane potential. There was no improvement in adenosine triphosphate (ATP) contents, but ATP synthesis was improved by UC-MSC-CM. Cytokine and active factor analysis of UC-MSC-CM showed that it contained many regulators inhibiting insulin resistance. Conclusion: UC-MSC-CM improves insulin resistance with multiple mechanisms in C2C12 cell.


Asunto(s)
Resistencia a la Insulina , Células Madre Mesenquimatosas , Medios de Cultivo Condicionados/farmacología , Humanos , Insulina , Cordón Umbilical
20.
Mol Cell ; 80(4): 592-606.e8, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33159855

RESUMEN

Despite its outstanding clinical success, immune checkpoint blockade remains ineffective in many patients. Accordingly, combination therapy capable of achieving greater antitumor immunity is urgently required. Here, we report that limiting glutamine metabolism in cancer cells bolsters the effectiveness of anti-programmed death ligand-1 (PD-L1) antibody. Inhibition of glutamine utilization increased PD-L1 levels in cancer cells, thereby inactivating co-cultured T cells. Under glutamine-limited conditions, reduced cellular GSH levels caused an upregulation of PD-L1 expression by impairing SERCA activity, which activates the calcium/NF-κB signaling cascade. Consequently, in tumors grown in immunocompetent mice, inhibition of glutamine metabolism decreased the antitumor activity of T cells. In combination with anti-PD-L1, however, glutamine depletion strongly promoted the antitumor efficacy of T cells in vitro and in vivo due to simultaneous increases in Fas/CD95 levels. Our results demonstrate the relevance of cancer glutamine metabolism to antitumor immunity and suggest that co-targeting of glutamine metabolism and PD-L1 represents a promising therapeutic approach.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/metabolismo , Glutamina/metabolismo , Glutatión/metabolismo , Neoplasias/inmunología , Neoplasias/prevención & control , Linfocitos T/inmunología , Anciano , Animales , Apoptosis , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Proliferación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...