Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 147(15): 1147-1161, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36856044

RESUMEN

BACKGROUND: The human heart primarily metabolizes fatty acids, and this decreases as alternative fuel use rises in heart failure with reduced ejection fraction (HFrEF). Patients with severe obesity and diabetes are thought to have increased myocardial fatty acid metabolism, but whether this is found in those who also have heart failure with preserved ejection fraction (HFpEF) is unknown. METHODS: Plasma and endomyocardial biopsies were obtained from HFpEF (n=38), HFrEF (n=30), and nonfailing donor controls (n=20). Quantitative targeted metabolomics measured organic acids, amino acids, and acylcarnitines in myocardium (72 metabolites) and plasma (69 metabolites). The results were integrated with reported RNA sequencing data. Metabolomics were analyzed using agnostic clustering tools, Kruskal-Wallis test with Dunn test, and machine learning. RESULTS: Agnostic clustering of myocardial but not plasma metabolites separated disease groups. Despite more obesity and diabetes in HFpEF versus HFrEF (body mass index, 39.8 kg/m2 versus 26.1 kg/m2; diabetes, 70% versus 30%; both P<0.0001), medium- and long-chain acylcarnitines (mostly metabolites of fatty acid oxidation) were markedly lower in myocardium from both heart failure groups versus control. In contrast, plasma levels were no different or higher than control. Gene expression linked to fatty acid metabolism was generally lower in HFpEF versus control. Myocardial pyruvate was higher in HFpEF whereas the tricarboxylic acid cycle intermediates succinate and fumarate were lower, as were several genes controlling glucose metabolism. Non-branched-chain and branched-chain amino acids (BCAA) were highest in HFpEF myocardium, yet downstream BCAA metabolites and genes controlling BCAA metabolism were lower. Ketone levels were higher in myocardium and plasma of patients with HFrEF but not HFpEF. HFpEF metabolomic-derived subgroups were differentiated by only a few differences in BCAA metabolites. CONCLUSIONS: Despite marked obesity and diabetes, HFpEF myocardium exhibited lower fatty acid metabolites compared with HFrEF. Ketones and metabolites of the tricarboxylic acid cycle and BCAA were also lower in HFpEF, suggesting insufficient use of alternative fuels. These differences were not detectable in plasma and challenge conventional views of myocardial fuel use in HFpEF with marked diabetes and obesity and suggest substantial fuel inflexibility in this syndrome.


Asunto(s)
Diabetes Mellitus , Insuficiencia Cardíaca , Humanos , Insuficiencia Cardíaca/metabolismo , Volumen Sistólico , Miocardio/metabolismo , Diabetes Mellitus/patología , Obesidad/patología , Ácidos Grasos
2.
Basic Res Cardiol ; 116(1): 25, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33847835

RESUMEN

We have previously shown that overexpression of SKI, an endogenous TGF-ß1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-ß signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI's effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI's capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.


Asunto(s)
Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Vía de Señalización Hippo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Remodelación Ventricular , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/patología , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fenotipo , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo
3.
Cell Tissue Res ; 385(3): 753-768, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34057573

RESUMEN

Fibroblast growth factor 2 (FGF2), produced as high (Hi-) and low (Lo-) molecular weight isoforms, is implicated in cardiac response to injury. The role of endogenous FGF2 isoforms during chronic stress is not well defined. We investigated the effects of endogenous Hi-FGF2 in a mouse model of simulated pressure-overload stress achieved by transverse aortic constriction (TAC) surgery. Hi-FGF2 knockout mice, expressing only Lo-FGF2, FGF2(Lo), and wild-type mice, FGF2(WT), expressing both Hi-FGF2 and Lo-FGF2, were used. By echocardiography, a decline in systolic function was observed in FGF2(WT) but not FGF2(Lo) mice compared to corresponding sham-operated animals at 4-8 weeks post-TAC surgery. TAC surgery increased markers of myocardial stress/damage including B-type natriuretic peptide (BNP) and the pro-cell death protein BCL2/adenovirus E1B 19 kDa protein-interacting protein-3 (Bnip3) in FGF2(WT) but not FGF2(Lo) mice. In FGF2(Lo) mice, cardiac levels of activated FGF receptor 1 (FGFR1), and downstream signals, including phosphorylated mTOR and p70S6 kinase, were elevated post-TAC. Finally, NR1D1 (nuclear receptor subfamily 1 group D member 1), implicated in cardioprotection from pressure-overload stress, was downregulated or upregulated in the presence or absence, respectively, of Hi-FGF2 expression, post-TAC surgery. In wild-type cardiomyocyte cultures, endothelin-1 (added to simulate pressure-overload signals) caused NR1D1 downregulation and BNP upregulation, similar to the effect of TAC surgery on the FGF2(WT) mice. The NR1D1 agonist SR9009 prevented BNP upregulation, simulating post-TAC findings in FGF2(Lo) mice. We propose that elimination of Hi-FGF2 is cardioprotective during pressure-overload by increasing FGFR1-associated signaling and NR1D1 expression.


Asunto(s)
Presión Sanguínea/genética , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Ratas , Transducción de Señal
4.
Am J Physiol Heart Circ Physiol ; 316(2): H279-H288, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30412444

RESUMEN

Cardiac fibroblast growth factor 2 (FGF2) exerts multiple paracrine activities related to cardiac response to injury. Endogenous FGF2 is composed of a mixture of 70% high- and 30% low-molecular-weight isoforms (Hi-FGF2 and Lo-FGF2, respectivley); although exogenously added Lo-FGF2 is cardioprotective, the roles of endogenous Hi-FGF2 or Lo-FGF2 have not been well defined. Therefore, we investigated the effect of elimination of Hi-FGF2 expression on susceptibility to acute cardiac damage in vivo caused by an injection of the genotoxic drug doxorubicin (Dox). Mice genetically depleted of endogenous Hi-FGF2 and expressing only Lo-FGF2 [FGF2(Lo) mice] were protected from the Dox-induced decline in ejection fraction displayed by their wild-type FGF2 [FGF2(WT)] mouse counterparts, regardless of sex, as assessed by echocardiography for up to 10 days post-Dox treatment. Because cardiac FGF2 is produced mainly by nonmyocytes, we next addressed potential contribution of fibroblast-produced FGF2 on myocyte vulnerability to Dox. In cocultures of neonatal rat cardiomyocytes (r-cardiomyocytes) with mouse fibroblasts from FGF2(WT) or FGF2(Lo) mice, only the FGF2(Lo)-fibroblast cocultures protected r-cardiomyocytes from Dox-induced mitochondrial and cellular damage. When r-cardiomyocytes were cocultured with or exposed to conditioned medium from human fibroblasts, neutralizing antibodies for human Hi-FGF-2, but not total FGF2, mitigated Dox-induced injury of cardiomyocytes. We conclude that endogenous Hi-FGF2 reduces cardioprotection by endogenous Lo-FGF2. Antibody-based neutralization of endogenous Hi-FGF2 may offer a prophylactic treatment against agents causing acute cardiac damage. NEW & NOTEWORTHY Cardiomyocytes, in vivo and in vitro, were protected from the deleterious effects of the anticancer drug doxorubicin by the genetic elimination or antibody-based neutralization of endogenous paracrine high-molecular-weight fibroblast growth factor 2 isoforms. These findings have a translational potential for mitigating doxorubicin-induced cardiac damage in patients with cancer by an antibody-based treatment.


Asunto(s)
Doxorrubicina/toxicidad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miofibroblastos/metabolismo , Animales , Gasto Cardíaco , Cardiotoxicidad , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Corazón/fisiología , Humanos , Masculino , Ratones , Ratas
5.
Cell Tissue Res ; 374(3): 607-617, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30159756

RESUMEN

Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. HO-1 upregulation was reported to require protein kinase CK2 activity. We show that a mutant non-mitogenic FGF2 (S117A-FGF2), which does not activate CK2, is cardioprotective against acute cardiac ischemic injury. We now investigate the potential of S117A-FGF2 to protect cardiomyocytes against acute Dox injury and decrease Dox-induced upregulation of oxidized phospholipids. The roles of CK2 and HO-1 in cardiomyocyte protection are also addressed.Rat neonatal cardiomyocyte cultures were used as an established in vitro model of acute Dox toxicity. Pretreatment with S117A-FGF2 protected against Dox-induced: oxidative stress; upregulation of fragmented and non-fragmented oxidized phosphatidylcholine species, measured by LC/MS/MS; and cardiomyocyte injury and cell death measured by LDH release and a live-dead assay. CK2 inhibitors (TBB and Ellagic acid), did not affect protection by S117A-FGF2 but prevented protection by mitogenic FGF2. Furthermore, protection by S117A-FGF2, unlike that of FGF2, was not prevented by HO-1 inhibitors and S117A-FGF2 did not upregulate HO-1. Protection by S117A-FGF2 required the activity of FGF receptor 1 and ERK.We conclude that mitogenic and non-mitogenic FGF2 protect from acute Dox toxicity by common (FGFR1) and distinct, CK2/HO-1- dependent or CK2/HO-1-independent (respectively), pathways. Non-mitogenic FGF2 merits further consideration as a preventative treatment against Dox cardiotoxicity.


Asunto(s)
Cardiotónicos/farmacología , Quinasa de la Caseína II/metabolismo , Citoprotección/efectos de los fármacos , Doxorrubicina/toxicidad , Factor 2 de Crecimiento de Fibroblastos/farmacología , Hemo-Oxigenasa 1/metabolismo , Miocitos Cardíacos/patología , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Fosfolípidos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
6.
bioRxiv ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38915649

RESUMEN

BACKGROUND: ATP-citrate lyase (ACLY) converts citrate into acetyl-CoA and oxaloacetate in the cytosol. It plays a prominent role in lipogenesis and fat accumulation coupled to excess glucose, and its inhibition is approved for treating hyperlipidemia. In RNAseq analysis of human failing myocardium, we found ACLY gene expression is reduced; however the impact this might have on cardiac function and/or metabolism has not been previously studied. As new ACLY inhibitors are in development for cancer and other disorders, such understanding has added importance. METHODS: Cardiomyocytes, ex-vivo beating hearts, and in vivo hearts with ACLY inhibited by selective pharmacologic (BMS303141, ACLYi) or genetic suppression, were studied. Regulation of ACLY gene/protein expression, and effects of ACLYi on function, cytotoxicity, tricarboxylic acid (TCA)-cycle metabolism, and redox and NAD+/NADH balance were assessed. Mice with cardiac ACLY knockdown induced by AAV9-acly-shRNA or cardiomyocyte tamoxifen-inducible Acly knockdown were studied. RESULTS: Acly gene expression was reduced more in obese patients with heart failure and preserved EF (HFpEF) than HF with reduced EF. In vivo pressure-overload and in vitro hormonal stress increased ACLY protein expression, whereas it declined upon fatty-acid exposure. Acute ACLYi (1-hr) dose-dependently induced cytotoxicity in adult and neonatal cardiomyocytes, and caused substantial reduction of systolic and diastolic function in myocytes and ex-vivo beating hearts. In the latter, ATP/ADP ratio also fell and lactate increased. U13C-glucose tracing revealed an ACLYdependent TCA-bypass circuit in myocytes, where citrate generated in mitochondria is transported to the cytosol, metabolized by ACLY and then converted to malate to re-enter mitochondria,bypassing several NADH-generating steps. ACLYi lowered NAD+/NADH ratio and restoring this balance ameliorated cardiomyocyte toxicity. Oxidative stress was undetected with ACLYi. Adult hearts following 8-weeks of reduced cardiac and/or cardiomyocyte ACLY downregulation exhibited ventricular dilation and reduced function that was prevented by NAD augmentation. Cardiac dysfunction from ACLY knockdown was worse in hearts subjected to sustained pressureoverload, supporting a role in stress responses. CONCLUSIONS: ACLY supports normal cardiac function through maintenance of the NAD+/NADH balance and is upregulated by hemodynamic and hormonal stress, but depressed by lipid excess. ACLY levels are most reduced in human HFpEF with obesity potentially worsening cardio-metabolic reserve.

7.
medRxiv ; 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37873321

RESUMEN

Heart failure with preserved ejection fraction (HFpEF) accounts for >50% of all heart failure world-wide and remains a major unmet medical need. The most effective recently approved treatments were first developed for diabetes, suggesting metabolic defects are paramount. Myocardial metabolomics in human HFpEF has identified reduced fatty acid and branched chain amino acid catabolism, but the status of glycolysis is unknown. Here we performed targeted metabolomics and protein analysis of glycolytic pathway enzymes in myocardial biopsies of patients with HFpEF versus HF with reduced ejection fraction (HFrEF0 or non-failing controls. Glucose was increased in HFpEF myocardium, but immediate downstream glycolytic metabolites (glucose-6 phosphate, fructose 1,6 diphosphate), were more reduced in HFpEF than the other groups, as were their associated synthetic enzymes hexokinase and phosphofructokinase. Pyruvate was also reduced in HFpEF versus controls. These changes were either not present or substantially less so in HFrEF. Suppression of proximal glycolysis was also coupled to lower metabolites and proteins in the pentose phosphate pathway but was independent of diabetes or obesity. These findings support marked metabolic inflexibility in HFpEF and identifies very proximal blockade in glucose metabolism. Efforts to improve metabolic use of carbohydrates in HFpEF will likely need to target these proximal glycolytic enzymes.

9.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37788104

RESUMEN

MTORC1 integrates signaling from the immune microenvironment to regulate T cell activation, differentiation, and function. TSC2 in the tuberous sclerosis complex tightly regulates mTORC1 activation. CD8+ T cells lacking TSC2 have constitutively enhanced mTORC1 activity and generate robust effector T cells; however, sustained mTORC1 activation prevents generation of long-lived memory CD8+ T cells. Here we show that manipulating TSC2 at Ser1365 potently regulated activated but not basal mTORC1 signaling in CD8+ T cells. Unlike nonstimulated TSC2-KO cells, CD8+ T cells expressing a phosphosilencing mutant TSC2-S1365A (TSC2-SA) retained normal basal mTORC1 activity. PKC and T cell receptor (TCR) stimulation induced TSC2 S1365 phosphorylation, and preventing this with the SA mutation markedly increased mTORC1 activation and T cell effector function. Consequently, SA CD8+ T cells displayed greater effector responses while retaining their capacity to become long-lived memory T cells. SA CD8+ T cells also displayed enhanced effector function under hypoxic and acidic conditions. In murine and human solid-tumor models, SA CD8+ T cells used as adoptive cell therapy displayed greater antitumor immunity than WT CD8+ T cells. These findings reveal an upstream mechanism to regulate mTORC1 activity in T cells. The TSC2-SA mutation enhanced both T cell effector function and long-term persistence/memory formation, supporting an approach to engineer better CAR-T cells for treating cancer.


Asunto(s)
Esclerosis Tuberosa , Ratones , Humanos , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina , Linfocitos T CD8-positivos , Mutación , Diferenciación Celular , Microambiente Tumoral
10.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34061779

RESUMEN

The emergence of the novel SARS coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has resulted in an unprecedented pandemic that has been accompanied by a global health crisis. Although the lungs are the main organs involved in COVID-19, systemic disease with a wide range of clinical manifestations also develops in patients infected with SARS-CoV-2. One of the major systems affected by this virus is the cardiovascular system. The presence of preexisting cardiovascular disease increases mortality in patients with COVID-19, and cardiovascular injuries, including myocarditis, cardiac rhythm abnormalities, endothelial cell injury, thrombotic events, and myocardial interstitial fibrosis, are observed in some patients with COVID-19. The underlying pathophysiology of COVID-19-associated cardiovascular complications is not fully understood, although direct viral infection of myocardium and cytokine storm have been suggested as possible mechanisms of myocarditis. In this Review, we summarize available data on SARS-CoV-2-related cardiac damage and discuss potential mechanisms of cardiovascular implications of this rapidly spreading virus.


Asunto(s)
COVID-19/complicaciones , Enfermedades Cardiovasculares/etiología , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiología , Enfermedades Cardiovasculares/diagnóstico , Fibrosis/diagnóstico , Fibrosis/etiología , Humanos , Miocarditis/diagnóstico , Miocarditis/etiología , Disautonomías Primarias/diagnóstico , Disautonomías Primarias/etiología , Trombosis/diagnóstico , Trombosis/etiología , Vasculitis/diagnóstico , Vasculitis/etiología , Tratamiento Farmacológico de COVID-19
11.
J Clin Invest ; 131(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34060484

RESUMEN

Iron plays an important role in mammalian physiological processes. It is a critical component for the function of many proteins, including enzymes that require heme and iron-sulfur clusters. However, excess iron is also detrimental because of its ability to catalyze the formation of reactive oxygen species. As a result, cellular and systemic iron levels are tightly regulated to prevent oxidative damage. Iron deficiency can lead to a number of pathological conditions, the most prominent being anemia. Iron deficiency should be corrected to improve adult patients' symptoms and to facilitate normal growth during fetal development and childhood. However, inappropriate use of intravenous iron in chronic conditions, such as cancer and heart failure, in the absence of clear iron deficiency can lead to unwanted side effects. Thus, this form of therapy should be reserved for certain patients who cannot tolerate oral iron and need rapid iron replenishment. Here, we will review cellular and systemic iron homeostasis and will discuss complications of iron deficiency.


Asunto(s)
Anemia Ferropénica , Insuficiencia Cardíaca , Hierro , Neoplasias , Adulto , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/metabolismo , Animales , Niño , Enfermedad Crónica , Desarrollo Fetal/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Hierro/metabolismo , Hierro/uso terapéutico , Deficiencias de Hierro , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Neurosciences (Riyadh) ; 15(3): 193-5, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20831029

RESUMEN

A 12-year-old girl was admitted to the Emergency Department with seizures and headache for 2 months. A CT scan and MRI showed a mass in the right frontal lobe with obvious mass effect. Surgery was carried out, and the resultant pathology was found to be aspergillosis. This study reports aspergillosis in an immunocompetent host following recurrent fungal otitis media. Although this condition is rare, it should be considered in patients with a history of fungal infections.


Asunto(s)
Aspergilosis/complicaciones , Aspergilosis/inmunología , Enfermedades del Sistema Nervioso Central , Inmunocompetencia , Aspergilosis/diagnóstico , Encéfalo/patología , Encéfalo/fisiopatología , Enfermedades del Sistema Nervioso Central/complicaciones , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/microbiología , Niño , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Tomografía Computarizada por Rayos X/métodos
13.
FEBS J ; 287(5): 1005-1034, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31545550

RESUMEN

Temozolomide (TMZ) is a chemotherapy agent used to treat Grade IV astrocytoma, also known as glioblastoma (GBM). TMZ treatment causes DNA damage that results in tumor cell apoptosis and increases the survival rate of GBM patients. However, chemoresistance as a result of TMZ-induced autophagy significantly reduces this anticancer effects over time. Statins are competitive inhibitors of HMG-CoA reductase, the rate-limiting enzyme of the mevalonate (MEV) cascade. Statins are best known for their cholesterol (CH)-lowering effect. Long-term consumption of statins, prior to and in parallel with other cancer therapeutic approaches, has been reported to increase the survival rate of patients with various forms of cancers. In this study, we investigated the potentiation of TMZ-induced apoptosis by simvastatin (Simva) in human GBM cell lines and patient GBM cells, using cell monolayers and three-dimensional cell culture systems. The incubation of cells with a combination of Simva and TMZ resulted in a significant increase in apoptotic cells compared to cells treated with TMZ alone. Incubation of cells with CH or MEV cascade intermediates failed to compensate the decrease in cell viability induced by the combined Simva and TMZ treatment. Simva treatment inhibited the autophagy flux induced by TMZ by blocking autophago-lysosome formation. Our results suggest that Simva sensitizes GBM cells to TMZ-induced cell death in a MEV cascade-independent manner and identifies the inhibition of autophagosome-lysosome fusion as a promising therapeutic strategy in the treatment of GBM.


Asunto(s)
Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Muerte Celular/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Simvastatina/farmacología , Temozolomida/farmacología , Animales , Línea Celular Tumoral , Femenino , Glioblastoma/metabolismo , Humanos , Macrólidos/farmacología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Int J Food Sci Nutr ; 60 Suppl 5: 19-27, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19115122

RESUMEN

Two thousand three hundred and thirty-one schoolchildren were selected by multi-stage random sampling. The thyroid size was estimated by inspection and palpation. The urinary iodine concentration and serum retinol (SR) were measured in 454 and 468 schoolchildren, respectively. Overall, 32.9% of 2,331 children had goiter. The median urinary iodine concentration was 195.5 microg/l. The mean+/-standard deviation of SR in goitrous and non-goitrous children was 34.84+/-7.28 microg/dl and 33.73+/-7.87 microg/dl, respectively (P=0.14). There was no child with vitamin A deficiency (SR <20 microg/dl) in both the goitrous and non-goitrous groups. The prevalence of subjects with low vitamin A status (SR <30 microg/dl) in the goitrous and non-goitrous groups was 27.7% and 33.3%, respectively (P=0.21). In conclusion, the present study reveals that vitamin A deficiency and low vitamin A status are not among the contributing factors of goiter persistence in the studied area, and the role of other responsible factors should be investigated.


Asunto(s)
Bocio Endémico/epidemiología , Yodo/administración & dosificación , Estado Nutricional , Deficiencia de Vitamina A/epidemiología , Vitamina A/sangre , Adolescente , Índice de Masa Corporal , Niño , Estudios Transversales , Femenino , Ferritinas/sangre , Bocio Endémico/sangre , Bocio Endémico/etiología , Humanos , Yodo/deficiencia , Yodo/orina , Irán/epidemiología , Hierro/sangre , Masculino , Política Nutricional , Tamaño de los Órganos , Prevalencia , Índice de Severidad de la Enfermedad , Factores Sexuales , Cloruro de Sodio Dietético/administración & dosificación , Glándula Tiroides/anatomía & histología , Deficiencia de Vitamina A/sangre , Deficiencia de Vitamina A/complicaciones
15.
Neurosciences (Riyadh) ; 14(3): 277-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21048630

RESUMEN

Until now, cerebral venous sinus thrombosis (CVST) was principally an autopsy diagnosis; however, with the introduction of MRI and angiography, as well as enhanced clinical attentiveness, it is now reliably diagnosed during life. Herein, we describe a case of CVST accompanied by oligodendroglioma and pregnancy. In our patient, the following factors contributed to the formation of CVST: First, the pregnancy state, which is a known risk factor for developing venous thrombosis; and secondly, the oligodendroglioma could have changed the architecture of adjacent sinus (right lateral sinus) and provocation of the development of clot in the sinus.

16.
Chem Biol Interact ; 303: 35-39, 2019 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-30707978

RESUMEN

Doxorubicin (Dox), a widely used chemotherapy drug, can also cause cardiotoxic effects leading to heart failure. The excessive oxidative stress caused by Dox results in the modification of a variety of cellular molecules, including phospholipids. In cardiomyocytes, Dox increases oxidation of a species of phospholipids, phosphatidylcholine, which has been associated with increased cell death. Oxidized phospholipids (Ox-PL) are involved in development and progression of various pathologies, including atherosclerosis, thrombosis, and tissue inflammation. Moreover, Ox-PL and excess iron are associated with ferroptosis, a form of regulated cell death. Neutralizing Ox-PL increases resistance to ischemia-reperfusion injuries which is linked to preservation of the mitochondrial membrane potential. This review aims to discuss the potential role of Ox-PL in Dox-induced pathology and supports the notion that a better understanding of the field could point to new strategies to prevent cardiotoxicity.


Asunto(s)
Cardiotoxicidad/etiología , Doxorrubicina/efectos adversos , Fosfolípidos/metabolismo , Animales , Humanos , Oxidación-Reducción , Fosfolípidos/fisiología
17.
Saudi Med J ; 29(8): 1130-4, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18690305

RESUMEN

OBJECTIVE: To evaluate the effect of metformin treatment on the risperidone-induced body weight gain in patients. METHODS: In a 12-weeks, double-blind, placebo controlled, randomized trial between October 2006 and October 2007 which was conducted in the Child and Adolescent Psychiatric Consultation Center of Isfahan University of Medical Sciences, 49 patients were entered the study with schizophrenia diagnosis. Then metformin (500 mg bid) or placebo was administrated with risperidone (6 mg) for the patients. Weight, height, and body mass index BMI were measured at the beginning, at 4 weeks, and at 12 weeks of the study. Changes in weight and BMI were evaluated by using repeated measures analysis of variance. RESULTS: Seventeen patients were excluded from the study. Repeated measure analysis of variances showed a significant difference between weight and BMI in both metformin (p<0.001, p<0.015) and placebo group (p<0.013, p<0.005). CONCLUSION: Metformin treatment did not show a significant effect to control the body weight of patients after 12 weeks.


Asunto(s)
Antipsicóticos/efectos adversos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Risperidona/efectos adversos , Aumento de Peso/efectos de los fármacos , Niño , Método Doble Ciego , Humanos , Esquizofrenia/tratamiento farmacológico
18.
J Investig Med ; 66(8): 1083-1087, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30368483

RESUMEN

Patients with glioblastoma multiforme (GBM) have an average life expectancy of approximately 15 months. Recently, statins have emerged as a potential adjuvant cancer therapy due to their ability to inhibit cell proliferation and induce apoptosis in many types of cancer. The exact mechanisms that mediate the inhibitory actions of statins in cancer cells are largely unknown. The purpose of this proceeding paper is to discuss some of the known anticancer effects of statins, while focusing on GBM therapy that includes adjunct therapy of statins with chemotherapeutic agents.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos , Glioblastoma/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Temozolomida/uso terapéutico , Humanos , Modelos Biológicos
19.
Oncotarget ; 8(28): 46663-46680, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28445146

RESUMEN

Doxorubicin (Dox) is a cytotoxic drug widely incorporated in various chemotherapy protocols. Severe side effects such as cardiotoxicity, however, limit Dox application. Mechanisms by which Dox promotes cardiac damage and cardiomyocyte cell death have been investigated extensively, but a definitive picture has yet to emerge. Autophagy, regarded generally as a protective mechanism that maintains cell viability by recycling unwanted and damaged cellular constituents, is nevertheless subject to dysregulation having detrimental effects for the cell. Autophagic cell death has been described, and has been proposed to contribute to Dox-cardiotoxicity. Additionally, mitophagy, autophagic removal of damaged mitochondria, is affected by Dox in a manner contributing to toxicity. Here we will review Dox-induced cardiotoxicity and cell death in the broad context of the autophagy and mitophagy processes.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Autofagia/efectos de los fármacos , Cardiotoxicidad/etiología , Doxorrubicina/efectos adversos , Cardiopatías/etiología , Mitofagia/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/uso terapéutico , Biomarcadores , Cardiotoxicidad/diagnóstico , Cardiotoxicidad/metabolismo , Doxorrubicina/uso terapéutico , Cardiopatías/diagnóstico , Cardiopatías/metabolismo , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Oncotarget ; 8(50): 87415-87430, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29152091

RESUMEN

BACKGROUND: Cardiotoxic side effects impose limits to the use of anti-tumour chemotherapeutic drugs such as doxorubicin (Dox). There is a need for cardioprotective strategies to prevent the multiple deleterious effects of Dox. Here, we examined the ability of administered fibroblast growth factor-2 (FGF-2), a cardioprotective protein that is synthesized as high and low molecular weight (Hi-, Lo-FGF-2) isoforms, to prevent Dox-induced: oxidative stress; cell death; lysosome dysregulation; and inactivation of potent endogenous protective pathways, such as the anti-oxidant/detoxification nuclear factor erythroid-2-related factor (Nrf-2), heme oxygenase-1 (HO-1) axis. METHODS AND RESULTS: Brief pre-incubation of neonatal rat cardiomyocyte cultures with either Hi- or Lo-FGF-2 reduced the Dox-induced: oxidative stress; apoptotic/necrotic cell death; lysosomal dysregulation; decrease in active mammalian target of Rapamycin (mTOR). FGF-2 isoforms prevented the Dox-induced downregulation of Nrf-2, and promoted robust increases in the Nrf-2-downstream targets including the cardioprotective protein HO-1, and p62/SQSTM1, a multifunctional scaffold protein involved in autophagy. Chloroquine, an autophagic flux inhibitor, caused a further increase in p62/SQSTM1, indicating intact autophagic flux in the FGF-2-treated groups. A selective inhibitor for HO-1, Tin-Protoporphyrin, prevented the FGF-2 protection against cell death. The mTOR inhibitor Rapamycin prevented FGF-2 protection, and blocked the FGF-2 effects on Nrf-2, HO-1 and p62/SQSTM1. CONCLUSIONS: In an acute setting Hi- or Lo-FGF-2 protect cardiomyocytes against multiple Dox-induced deleterious effects, by a mechanism dependent on preservation of mTOR activity, Nrf-2 levels, and the upregulation of HO-1. Preservation/activation of endogenous anti-oxidant/detoxification defences by FGF-2 is a desirable property in the setting of Dox-cardiotoxicity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA