Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.591
Filtrar
1.
PLoS One ; 19(5): e0303528, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753618

RESUMEN

Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,ß; TRα,ß) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,ß, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRß and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.


Asunto(s)
Arsénico , Hipotálamo , Mitocondrias , PPAR gamma , ARN Mensajero , Animales , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , PPAR gamma/metabolismo , PPAR gamma/genética , Arsénico/toxicidad , Receptores de Hormona Tiroidea/metabolismo , Receptores de Hormona Tiroidea/genética , Masculino , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Respiración de la Célula/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos
2.
Biomolecules ; 14(4)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38672489

RESUMEN

Colorectal cancer (CRC) is a leading cause of death worldwide. Conventional therapies are available with varying effectiveness. Acetate, a short-chain fatty acid produced by human intestinal bacteria, triggers mitochondria-mediated apoptosis preferentially in CRC but not in normal colonocytes, which has spurred an interest in its use for CRC prevention/therapy. We previously uncovered that acetate-induced mitochondrial-mediated apoptosis in CRC cells is significantly enhanced by the inhibition of the lysosomal protease cathepsin D (CatD), which indicates both mitochondria and the lysosome are involved in the regulation of acetate-induced apoptosis. Herein, we sought to determine whether mitochondrial function affects CatD apoptotic function. We found that enhancement of acetate-induced apoptosis by CatD inhibition depends on oligomycin A-sensitive respiration. Mechanistically, the potentiating effect is associated with an increase in cellular and mitochondrial superoxide anion accumulation and mitochondrial mass. Our results provide novel clues into the regulation of CatD function and the effect of tumor heterogeneity in the outcome of combined treatment using acetate and CatD inhibitors.


Asunto(s)
Apoptosis , Catepsina D , Neoplasias Colorrectales , Mitocondrias , Oligomicinas , Humanos , Acetatos/farmacología , Apoptosis/efectos de los fármacos , Catepsina D/metabolismo , Catepsina D/antagonistas & inhibidores , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Oligomicinas/farmacología
3.
Nutrients ; 15(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37432297

RESUMEN

Diosgenin (DIO) is a dietary steroid sapogenin possessing multiple biological functions, such as the amelioration of diabetes. However, the remission effect of DIO on diabetic nephropathy (DN) underlying oxidative stress and cell apoptosis remains unclear. Here, the effect of DIO on ROS generation and its induced cell apoptosis was studied in vitro and in vivo. Renal proximal tubular epithelial (HK-2) cells were treated with DIO (1, 2, 4 µM) under high glucose (HG, 30 mM) conditions. DN rats were induced by a high-fat diet combined with streptozotocin, followed by administration of DIO for 8 weeks. Our data suggested that DIO relieved the decline of HK-2 cell viability and renal pathological damage in DN rats. DIO also relieved ROS (O2- and H2O2) production. Mechanistically, DIO inhibited the expression of NOX4 and restored mitochondrial respiratory chain (MRC) complex I-V expressions. Further, DIO inhibited mitochondrial apoptosis by ameliorating mitochondrial membrane potential (MtMP) and down-regulating the expressions of CytC, Apaf-1, caspase 3, and caspase 9, while up-regulating Bcl2 expression. Moreover, the ER stress and its associated cell apoptosis were inhibited through decreasing PERK, p-PERK, ATF4, IRE1, p-CHOP, and caspase 12 expressions. Collectively, DIO inhibited ROS production by modulating NOX4 and MRC complexes, which then suppressed apoptosis regulated by mitochondria and ER stress, thereby attenuating DN.


Asunto(s)
Apoptosis , Neuropatías Diabéticas , Humanos , Línea Celular , Apoptosis/efectos de los fármacos , Diosgenina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Respiración de la Célula/efectos de los fármacos , Neuropatías Diabéticas/metabolismo , Animales , Ratas , Estrés del Retículo Endoplásmico , Ratas Sprague-Dawley , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
4.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35298717

RESUMEN

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Asunto(s)
Envejecimiento/metabolismo , Axones/patología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Metabolismo Energético , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Axones/efectos de los fármacos , Axones/metabolismo , Secuencia de Bases , Proteína beta Potenciadora de Unión a CCAAT/genética , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Metabolismo Energético/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucólisis/efectos de los fármacos , Células HEK293 , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Hígado/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Factores de Transcripción NFATC/metabolismo , Proyección Neuronal/efectos de los fármacos , Polímeros/metabolismo , Regiones Promotoras Genéticas/genética , Transporte de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley , Células Receptoras Sensoriales/patología , Transducción de Señal/efectos de los fármacos
5.
Biochem Biophys Res Commun ; 588: 140-146, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34954521

RESUMEN

Smoking cessation increases body weight. The underlying mechanisms, however, have not been fully understood. We here report an establishment of a mouse model that exhibits an augmented body weight gain after nicotine withdrawal. High fat diet-fed mice were infused with nicotine for two weeks, and then with vehicle for another two weeks using osmotic minipumps. Body weight increased immediately after nicotine cessation and was significantly higher than that of mice continued on nicotine. Mice switched to vehicle consumed more food than nicotine-continued mice during the first week of cessation, while oxygen consumption was comparable. Elevated expression of orexigenic agouti-related peptide was observed in the hypothalamic appetite center. Pair-feeding experiment revealed that the accelerated weight gain after nicotine withdrawal is explained by enhanced energy intake. As a showcase of an efficacy of pharmacologic intervention, exendin-4 was administered and showed a potent suppression of energy intake and weight gain in mice withdrawn from nicotine. Our current model provides a unique platform for the investigation of the changes of energy regulation after smoking cessation.


Asunto(s)
Nicotina/efectos adversos , Síndrome de Abstinencia a Sustancias/patología , Aumento de Peso , Proteína Relacionada con Agouti/metabolismo , Animales , Calorimetría , Respiración de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Ingestión de Energía/efectos de los fármacos , Exenatida/farmacología , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Síndrome de Abstinencia a Sustancias/genética , Aumento de Peso/efectos de los fármacos , Aumento de Peso/genética
6.
PLoS Genet ; 17(12): e1009971, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34965247

RESUMEN

Rothmund-Thomson syndrome (RTS) is an autosomal recessive genetic disorder characterized by poikiloderma, small stature, skeletal anomalies, sparse brows/lashes, cataracts, and predisposition to cancer. Type 2 RTS patients with biallelic RECQL4 pathogenic variants have multiple skeletal anomalies and a significantly increased incidence of osteosarcoma. Here, we generated RTS patient-derived induced pluripotent stem cells (iPSCs) to dissect the pathological signaling leading to RTS patient-associated osteosarcoma. RTS iPSC-derived osteoblasts showed defective osteogenic differentiation and gain of in vitro tumorigenic ability. Transcriptome analysis of RTS osteoblasts validated decreased bone morphogenesis while revealing aberrantly upregulated mitochondrial respiratory complex I gene expression. RTS osteoblast metabolic assays demonstrated elevated mitochondrial respiratory complex I function, increased oxidative phosphorylation (OXPHOS), and increased ATP production. Inhibition of mitochondrial respiratory complex I activity by IACS-010759 selectively suppressed cellular respiration and cell proliferation of RTS osteoblasts. Furthermore, systems analysis of IACS-010759-induced changes in RTS osteoblasts revealed that chemical inhibition of mitochondrial respiratory complex I impaired cell proliferation, induced senescence, and decreased MAPK signaling and cell cycle associated genes, but increased H19 and ribosomal protein genes. In summary, our study suggests that mitochondrial respiratory complex I is a potential therapeutic target for RTS-associated osteosarcoma and provides future insights for clinical treatment strategies.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Osteosarcoma/genética , ARN Largo no Codificante/genética , RecQ Helicasas/genética , Síndrome Rothmund-Thomson/genética , Adenosina Trifosfato/biosíntesis , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Senescencia Celular/genética , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación/genética , Osteoblastos/efectos de los fármacos , Osteogénesis/genética , Osteosarcoma/complicaciones , Osteosarcoma/patología , Oxadiazoles/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Piperidinas/farmacología , Síndrome Rothmund-Thomson/complicaciones , Síndrome Rothmund-Thomson/patología
7.
Cells ; 10(11)2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34831040

RESUMEN

Preeclampsia (PE) is characterized by new onset hypertension in association with placental ischemia, reduced fetal weight, elevated soluble fms-like tyrosine kinase-1 (sFlt-1), and placental mitochondrial (mt) dysfunction and oxidative stress (ROS). Progesterone induced blocking factor (PIBF) is a product of progesterone signaling that blocks inflammatory processes and we have previously shown PIBF to lower mean arterial blood pressure (MAP) and sFlt-1 in a rat model of PE. Infusion of sFlt-1 causes hypertension and many characteristics of PE in pregnant rodents, however, its role in causing mt dysfunction is unknown. Therefore, we hypothesize that PIBF will improve mt function and MAP in response to elevated sFlt-1 during pregnancy. We tested our hypothesis by infusing sFlt-1 via miniosmotic pumps in normal pregnant (NP) Sprague-Dawley rats (3.7 µg·kg-1·day-1) on gestation days (GD) 13-19 in the presence or absence of PIBF (2.0 µg/mL) injected intraperitoneally on GD 15 and examined mean arterial blood pressure (MAP) and placental mt ROS on GD 19. sFlt-1 increased MAP to 112 + 2 (n = 11) compared to NP rats (98 + 2 mmHg, n = 15, p < 0.05), which was lowered in the presence of sFlt-1 (100 + 1 mmHg, n = 5, p < 0.05). Placental mtATP was reduced in sFlt-1 infused rats versus NP controls, but was improved with PIBF. Placental mtROS was elevated with sFlt-1 compared to NP controls, but was reduced with PIBF. Sera from NP + sFlt-1 increased endothelial cell mtROS, which was attenuated with PIBF. These data demonstrate sFlt-1 induced HTN during pregnancy reduces placental mt function. Importantly, PIBF improved placental mt function and HTN, indicating the efficacy of improved progesterone signaling as potential therapeutics for PE.


Asunto(s)
Antígenos de Neoplasias/farmacología , Hipertensión/patología , Mitocondrias/patología , Placenta/metabolismo , Progesterona/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Presión Sanguínea , Peso Corporal/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Femenino , Feto/metabolismo , Hipertensión/sangre , Hipertensión/fisiopatología , Mitocondrias/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Solubilidad
8.
Cells ; 10(11)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34831251

RESUMEN

Cardiorenal syndrome (CRS) is a pathological link between the kidneys and heart, in which an insult in a kidney or heart leads the other organ to incur damage. CRS is classified into five subtypes, and type 3 (CRS3) is characterized by acute kidney injury as a precursor to subsequent cardiovascular changes. Mitochondrial dysfunction and oxidative and nitrosative stress have been reported in the pathophysiology of CRS3. It is known that vitamin C, an antioxidant, has proven protective capacity for cardiac, renal, and vascular endothelial tissues. Therefore, the present study aimed to assess whether vitamin C provides protection to heart and the kidneys in an in vivo CRS3 model. The unilateral renal ischemia and reperfusion (IR) protocol was performed for 60 min in the left kidney of adult mice, with and without vitamin C treatment, immediately after IR or 15 days after IR. Kidneys and hearts were subsequently collected, and the following analyses were conducted: renal morphometric evaluation, serum urea and creatinine levels, high-resolution respirometry, amperometry technique for NO measurement, gene expression of mitochondrial dynamic markers, and NOS. The analyses showed that the left kidney weight was reduced, urea and creatinine levels were increased, mitochondrial oxygen consumption was reduced, NO levels were elevated, and Mfn2 expression was reduced after 15 days of IR compared to the sham group. Oxygen consumption and NO levels in the heart were also reduced. The treatment with vitamin C preserved the left kidney weight, restored renal function, reduced NO levels, decreased iNOS expression, elevated constitutive NOS isoforms, and improved oxygen consumption. In the heart, oxygen consumption and NO levels were improved after vitamin C treatment, whereas the three NOS isoforms were overexpressed. These data indicate that vitamin C provides protection to the kidneys and some beneficial effects to the heart after IR, indicating it may be a preventive approach against cardiorenal insults.


Asunto(s)
Ácido Ascórbico/farmacología , Síndrome Cardiorrenal/patología , Riñón/patología , Mitocondrias/patología , Animales , Respiración de la Célula/efectos de los fármacos , Isoenzimas/metabolismo , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Dinámicas Mitocondriales/efectos de los fármacos , Modelos Biológicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología
9.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34769217

RESUMEN

Amiodarone is a potent antiarrhythmic drug and displays substantial liver toxicity in humans. It has previously been demonstrated that amiodarone and its metabolite (desethylamiodarone, DEA) can inhibit mitochondrial function, particularly complexes I (CI) and II (CII) of the electron transport system in various animal tissues and cell types. The present study, performed in human peripheral blood cells, and one liver-derived human cell line, is primarily aimed at assessing the concentration-dependent effects of these drugs on mitochondrial function (respiration and cellular ATP levels). Furthermore, we explore the efficacy of a novel cell-permeable succinate prodrug in alleviating the drug-induced acute mitochondrial dysfunction. Amiodarone and DEA elicit a concentration-dependent impairment of mitochondrial respiration in both intact and permeabilized platelets via the inhibition of both CI- and CII-supported respiration. The inhibitory effect seen in human platelets is also confirmed in mononuclear cells (PBMCs) and HepG2 cells. Additionally, amiodarone elicits a severe concentration-dependent ATP depletion in PBMCs, which cannot be explained solely by mitochondrial inhibition. The succinate prodrug NV118 alleviates the respiratory deficit in platelets and HepG2 cells acutely exposed to amiodarone. In conclusion, amiodarone severely inhibits metabolism in primary human mitochondria, which can be counteracted by increasing mitochondrial function using intracellular delivery of succinate.


Asunto(s)
Amiodarona/toxicidad , Antiarrítmicos/toxicidad , Mitocondrias/efectos de los fármacos , Sustancias Protectoras/farmacología , Ácido Succínico/farmacología , Adenosina Trifosfato/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Respiración de la Célula/efectos de los fármacos , Células Hep G2 , Humanos , Mitocondrias/metabolismo , Profármacos/farmacología
10.
Cell Death Dis ; 12(10): 893, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593753

RESUMEN

Uncontrolled mitosis is one of the most important features of cancer, and mitotic kinases are thought to be ideal targets for anticancer therapeutics. However, despite numerous clinical attempts spanning decades, clinical trials for mitotic kinase-targeting agents have generally stalled in the late stages due to limited therapeutic effectiveness. Alisertib (MLN8237) is a promising oral mitotic aurora kinase A (AURKA, Aurora-A) selective inhibitor, which is currently under several clinical evaluations but has failed in its first Phase III trial due to inadequate efficacy. In this study, we performed genome-wide CRISPR/Cas9-based screening to identify vulnerable biological processes associated with alisertib in breast cancer MDA-MB-231 cells. The result indicated that alisertib treated cancer cells are more sensitive to the genetic perturbation of oxidative phosphorylation (OXPHOS). Mechanistic investigation indicated that alisertib treatment, as well as other mitotic kinase inhibitors, rapidly reduces the intracellular ATP level to generate a status that is highly addictive to OXPHOS. Furthermore, the combinational inhibition of mitotic kinase and OXPHOS by alisertib, and metformin respectively, generates severe energy exhaustion in mitotic cells that consequently triggers cell death. The combination regimen also enhanced tumor regression significantly in vivo. This suggests that targeting OXPHOS by metformin is a potential strategy for promoting the therapeutic effects of mitotic kinase inhibitors through the joint targeting of mitosis and cellular energy homeostasis.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Mitosis , Fosforilación Oxidativa , Adenosina Trifosfato/metabolismo , Animales , Aurora Quinasa A/metabolismo , Azepinas/farmacología , Neoplasias de la Mama/patología , Sistemas CRISPR-Cas/genética , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Citosol/metabolismo , Sinergismo Farmacológico , Metabolismo Energético/efectos de los fármacos , Femenino , Homeostasis/efectos de los fármacos , Humanos , Metformina/farmacología , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitosis/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Pirimidinas/farmacología
11.
Cell Death Dis ; 12(11): 953, 2021 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-34657120

RESUMEN

Many cell death pathways, including apoptosis, regulated necrosis, and ferroptosis, are relevant for neuronal cell death and share common mechanisms such as the formation of reactive oxygen species (ROS) and mitochondrial damage. Here, we present the role of the actin-regulating protein cofilin1 in regulating mitochondrial pathways in oxidative neuronal death. Cofilin1 deletion in neuronal HT22 cells exerted increased mitochondrial resilience, assessed by quantification of mitochondrial ROS production, mitochondrial membrane potential, and ATP levels. Further, cofilin1-deficient cells met their energy demand through enhanced glycolysis, whereas control cells were metabolically impaired when challenged by ferroptosis. Further, cofilin1 was confirmed as a key player in glutamate-mediated excitotoxicity and associated mitochondrial damage in primary cortical neurons. Using isolated mitochondria and recombinant cofilin1, we provide a further link to toxicity-related mitochondrial impairment mediated by oxidized cofilin1. Our data revealed that the detrimental impact of cofilin1 on mitochondria depends on the oxidation of cysteine residues at positions 139 and 147. Overall, our findings show that cofilin1 acts as a redox sensor in oxidative cell death pathways of ferroptosis, and also promotes glutamate excitotoxicity. Protective effects by cofilin1 inhibition are particularly attributed to preserved mitochondrial integrity and function. Thus, interfering with the oxidation and pathological activation of cofilin1 may offer an effective therapeutic strategy in neurodegenerative diseases.


Asunto(s)
Cofilina 1/metabolismo , Mitocondrias/patología , Neuronas/patología , Estrés Oxidativo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cofilina 1/deficiencia , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácido Glutámico/toxicidad , Glucólisis/efectos de los fármacos , Humanos , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piperazinas/toxicidad , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/farmacología
12.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681781

RESUMEN

In this paper, we examined the effects of melittin, a bee venom membrane-active peptide, on mitochondrial respiration and cell viability of healthy human lymphocytes (HHL) and Jurkat cells, as well as on lymphoblasts from acute human T cell leukemia. The viability of melittin-treated cells was related to changes in O2 consumption and in the respiratory control index (RCI) of mitochondria isolated from melittin-pretreated cells as well as of mitochondria first isolated from cells and then directly treated with melittin. It was shown that melittin is three times more cytotoxic to Jurkat cells than to HHL, but O2 consumption and RCI values of mitochondria from both cell types were equally affected by melittin when melittin was directly added to mitochondria. To elucidate the molecular mechanism of melittin's cytotoxicity to healthy and cancer cells, the effects of melittin on lipid-packing and on the dynamics in model plasma membranes of healthy and cancer cells, as well as of the inner mitochondrial membrane, were studied by EPR spin probes. The affinity of melittin binding to phosphatidylcholine, phosphatidylserine, phosphatidic acid and cardiolipin, and binding sites of phospholipids on the surface of melittin were studied by 31P-NMR, native PAGE and AutoDock modeling. It is suggested that the melittin-induced decline of mitochondrial bioenergetics contributes primarily to cell death; the higher cytotoxicity of melittin to cancer cells is attributed to its increased permeability through the plasma membrane.


Asunto(s)
Linfocitos/efectos de los fármacos , Meliteno/farmacología , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Venenos de Abeja/química , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Humanos , Células Jurkat , Membrana Dobles de Lípidos/química , Linfocitos/metabolismo , Meliteno/aislamiento & purificación , Mitocondrias/fisiología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Modelos Biológicos , Permeabilidad/efectos de los fármacos
13.
Plant Physiol ; 186(3): 1487-1506, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34624108

RESUMEN

Because it is the precursor for various essential cellular components, the amino acid serine is indispensable for every living organism. In plants, serine is synthesized by two major pathways: photorespiration and the phosphorylated pathway of serine biosynthesis (PPSB). However, the importance of these pathways in providing serine for plant development is not fully understood. In this study, we examine the relative contributions of photorespiration and PPSB to providing serine for growth and metabolism in the C3 model plant Arabidopsis thaliana. Our analyses of cell proliferation and elongation reveal that PPSB-derived serine is indispensable for plant growth and its loss cannot be compensated by photorespiratory serine biosynthesis. Using isotope labeling, we show that PPSB-deficiency impairs the synthesis of proteins and purine nucleotides in plants. Furthermore, deficiency in PPSB-mediated serine biosynthesis leads to a strong accumulation of metabolites related to nitrogen metabolism. This result corroborates 15N-isotope labeling in which we observed an increased enrichment in labeled amino acids in PPSB-deficient plants. Expression studies indicate that elevated ammonium uptake and higher glutamine synthetase/glutamine oxoglutarate aminotransferase (GS/GOGAT) activity causes this phenotype. Metabolic analyses further show that elevated nitrogen assimilation and reduced amino acid turnover into proteins and nucleotides are the most likely driving forces for changes in respiratory metabolism and amino acid catabolism in PPSB-deficient plants. Accordingly, we conclude that even though photorespiration generates high amounts of serine in plants, PPSB-derived serine is more important for plant growth and its deficiency triggers the induction of nitrogen assimilation, most likely as an amino acid starvation response.


Asunto(s)
Arabidopsis/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Nitrógeno/metabolismo , Desarrollo de la Planta/fisiología , Reguladores del Crecimiento de las Plantas/metabolismo , Serina/biosíntesis , Vías Biosintéticas , Fosforilación
14.
Oxid Med Cell Longev ; 2021: 5577541, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707777

RESUMEN

Mitochondrial dysfunction and oxidative stress are extensively linked to Parkinson's disease (PD) pathogenesis. Melatonin is a pleiotropic molecule with antioxidant and neuroprotective effects. The aim of this study was to evaluate the effect of melatonin on oxidative stress markers, mitochondrial complex 1 activity, and mitochondrial respiratory control ratio in patients with PD. A double-blind, cross-over, placebo-controlled randomized clinical trial study was conducted in 26 patients who received either 25 mg of melatonin or placebo at noon and 30 min before bedtime for three months. At the end of the trial, in patients who received melatonin, we detected a significant diminution of lipoperoxides, nitric oxide metabolites, and carbonyl groups in plasma samples from PD patients compared with the placebo group. Conversely, catalase activity was increased significantly in comparison with the placebo group. Compared with the placebo group, the melatonin group showed significant increases of mitochondrial complex 1 activity and respiratory control ratio. The fluidity of the membranes was similar in the melatonin group and the placebo group at baseline and after three months of treatment. In conclusion, melatonin administration was effective in reducing the levels of oxidative stress markers and restoring the rate of complex I activity and respiratory control ratio without modifying membrane fluidity. This suggests that melatonin could play a role in the treatment of PD.


Asunto(s)
Antioxidantes/uso terapéutico , Antiparkinsonianos/uso terapéutico , Melatonina/uso terapéutico , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Antioxidantes/efectos adversos , Antiparkinsonianos/efectos adversos , Biomarcadores/sangre , Respiración de la Célula/efectos de los fármacos , Estudios Cruzados , Método Doble Ciego , Complejo I de Transporte de Electrón/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Melatonina/efectos adversos , México , Mitocondrias/metabolismo , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/diagnóstico , Factores de Tiempo , Resultado del Tratamiento
15.
PLoS One ; 16(10): e0258845, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34699527

RESUMEN

The ectoparasite Varroa destructor Anderson and Trueman is the most important parasites of the western honey bee, Apis mellifera L. The most widely currently used treatment uses formic acid (FA), but the understanding of its effects on V. destructor is limited. In order to understand the mechanism of action of FA, its effect on Varroa mites was investigated using proteomic analysis by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). V. destructor was collected from honey bee colonies with natural mite infestation before and 24 h after the initiation of FA treatment and subjected to proteome analysis. A total of 2637 proteins were identified. Quantitative analysis of differentially expressed candidate proteins (fold change ≥ 1.5; p ≤ 0.05) revealed 205 differentially expressed proteins: 91 were induced and 114 repressed in the FA-treated group compared to the untreated control group. Impaired protein synthesis accompanied by increased protein and amino acid degradation suggest an imbalance in proteostasis. Signs of oxidative stress included significant dysregulation of candidate proteins of mitochondrial cellular respiration, increased endocytosis, and induction of heat shock proteins. Furthermore, an increased concentration of several candidate proteins associated with detoxification was observed. These results suggest dysregulated cellular respiration triggered by FA treatment as well as an increase in cellular defense mechanisms, including induced heat shock proteins and detoxification enzymes.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Formiatos/farmacología , Proteómica/métodos , Varroidae/metabolismo , Animales , Abejas/parasitología , Respiración de la Célula/efectos de los fármacos , Cromatografía Liquida , Regulación de la Expresión Génica/efectos de los fármacos , Estrés Oxidativo , Espectrometría de Masas en Tándem , Varroidae/efectos de los fármacos
16.
Cells ; 10(10)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34685775

RESUMEN

IL-2 is a cytokine released from CD4+T cells with dual actions and can either potentiate the inflammatory response or quell a chronic inflammatory response depending on its circulating concentration. IL-2 is elevated in many chronic inflammatory conditions and is increased during preeclampsia (PE). PE is characterized by new-onset hypertension during pregnancy and organ dysfunction and increasing evidence indicates that proinflammatory cytokines cause hypertension and mitochondrial (mt) dysfunction during pregnancy. The reduced uterine perfusion pressure (RUPP) model of placental ischemia is a rat model of PE that we commonly use in our laboratory and we have previously shown that low doses of recombinant IL-2 can decrease blood pressure in RUPP rats. The objective of this study was to determine the effects of a low dose of recombinant IL-2 on multi-organ mt dysfunction in the RUPP rat model of PE. We tested our hypothesis by infusing recombinant IL-2 (0.05 ng/mL) into RUPP rats on GD14 and examined mean arterial pressure (MAP), renal, placental and endothelial cell mt function compared to control RUPP. MAP was elevated in RUPP rats (n = 6) compared to controls (n = 5) (122 ± 5 vs. 102 ± 3 mmHg, p < 0.05), but was reduced by administration of LD recombinant IL-2 (107 ± 1 vs. 122 ± 5 mmHg, n = 9, p < 0.05). Renal, placental and endothelial mt ROS were significantly increased in RUPP rats compared to RUPP+ IL-2 and controls. Placental and renal respiration rates were reduced in RUPP rats compared to control rats but were normalized with IL-2 administration to RUPPs. These data indicate that low-dose IL-2 normalized multi-organ mt function and hypertension in response to placental ischemia.


Asunto(s)
Hipertensión/complicaciones , Interleucina-2/farmacología , Isquemia/complicaciones , Mitocondrias/metabolismo , Placenta/patología , Animales , Presión Sanguínea/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hipertensión/sangre , Hipertensión/fisiopatología , Interleucina-2/sangre , Isquemia/sangre , Mitocondrias/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Preeclampsia/sangre , Preeclampsia/fisiopatología , Embarazo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
17.
Clin Sci (Lond) ; 135(19): 2243-2263, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34569605

RESUMEN

The protein tyrosine kinase inhibitor imatinib is used in the treatment of various malignancies but may also promote beneficial effects in the treatment of diabetes. The aim of the present investigation was to characterize the mechanisms by which imatinib protects insulin producing cells. Treatment of non-obese diabetic (NOD) mice with imatinib resulted in increased beta-cell AMP-activated kinase (AMPK) phosphorylation. Imatinib activated AMPK also in vitro, resulting in decreased ribosomal protein S6 phosphorylation and protection against islet amyloid polypeptide (IAPP)-aggregation, thioredoxin interacting protein (TXNIP) up-regulation and beta-cell death. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) mimicked and compound C counteracted the effect of imatinib on beta-cell survival. Imatinib-induced AMPK activation was preceded by reduced glucose/pyruvate-dependent respiration, increased glycolysis rates, and a lowered ATP/AMP ratio. Imatinib augmented the fractional oxidation of fatty acids/malate, possibly via a direct interaction with the beta-oxidation enzyme enoyl coenzyme A hydratase, short chain, 1, mitochondrial (ECHS1). In non-beta cells, imatinib reduced respiratory chain complex I and II-mediated respiration and acyl-CoA carboxylase (ACC) phosphorylation, suggesting that mitochondrial effects of imatinib are not beta-cell specific. In conclusion, tyrosine kinase inhibitors modestly inhibit mitochondrial respiration, leading to AMPK activation and TXNIP down-regulation, which in turn protects against beta-cell death.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Hipoglucemiantes/farmacología , Mesilato de Imatinib/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Animales , Proteínas Portadoras/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Diabetes Mellitus/enzimología , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Enoil-CoA Hidratasa/metabolismo , Activación Enzimática , Humanos , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Masculino , Ratones Endogámicos NOD , Mitocondrias/enzimología , Mitocondrias/patología , Fosforilación , Ratas Sprague-Dawley , Proteína S6 Ribosómica/metabolismo
18.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34575875

RESUMEN

Mitochondrial dysfunction during ischemic stroke ultimately manifests as ATP depletion. Mitochondrial ATP synthase upon loss of mitochondrial membrane potential during ischemia rapidly hydrolyses ATP and thus contributes to ATP depletion. Increasing evidence suggests that inhibition of ATP synthase limits ATP depletion and is protective against ischemic tissue damage. Bedaquiline (BDQ) is an anti-microbial agent, approved for clinical use, that inhibits ATP synthase of Mycobacteria; however recently it has been shown to act on mitochondrial ATP synthase, inhibiting both ATP synthesis and hydrolysis in low micromolar concentrations. In this study, we investigated whether preconditioning with BDQ can alleviate ischemia/reperfusion-induced brain injury in Wistar rats after middle cerebral artery occlusion-reperfusion and whether it affects mitochondrial functions. We found that BDQ was effective in limiting necrosis and neurological dysfunction during ischemia-reperfusion. BDQ also caused inhibition of ATPase activity, mild uncoupling of respiration, and stimulated mitochondrial respiration both in healthy and ischemic mitochondria. Mitochondrial calcium retention capacity was unaffected by BDQ preconditioning. We concluded that BDQ has neuroprotective properties associated with its action on mitochondrial respiration and ATPase activity.


Asunto(s)
Diarilquinolinas/farmacología , Inhibidores Enzimáticos/farmacología , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/metabolismo , Accidente Cerebrovascular/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Respiración de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuroprotección/efectos de los fármacos , Ratas , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología
19.
PLoS One ; 16(8): e0255164, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34343196

RESUMEN

Glutamate plays diverse roles in neuronal cells, affecting cell energetics and reactive oxygen species (ROS) generation. These roles are especially vital for neuronal cells, which deal with high amounts of glutamate as a neurotransmitter. Our analysis explored neuronal glutamate implication in cellular energy metabolism and ROS generation, using a kinetic model that simulates electron transport details in respiratory complexes, linked ROS generation and metabolic reactions. The analysis focused on the fact that glutamate attenuates complex II inhibition by oxaloacetate, stimulating the latter's transformation into aspartate. Such a mechanism of complex II activation by glutamate could cause almost complete reduction of ubiquinone and deficiency of oxidized form (Q), which closes the main stream of electron transport and opens a way to massive ROS generating transfer in complex III from semiquinone radicals to molecular oxygen. In this way, under low workload, glutamate triggers the respiratory chain (RC) into a different steady state characterized by high ROS generation rate. The observed stepwise dependence of ROS generation on glutamate concentration experimentally validated this prediction. However, glutamate's attenuation of oxaloacetate's inhibition accelerates electron transport under high workload. Glutamate-oxaloacetate interaction in complex II regulation underlies the observed effects of uncouplers and inhibitors and acceleration of Ca2+ uptake. Thus, this theoretical analysis uncovered the previously unknown roles of oxaloacetate as a regulator of ROS generation and glutamate as a modifier of this regulation. The model predicted that this mechanism of complex II activation by glutamate might be operative in situ and responsible for excitotoxicity. Spatial-time gradients of synthesized hydrogen peroxide concentration, calculated in the reaction-diffusion model with convection under a non-uniform local approximation of nervous tissue, have shown that overproduction of H2O2 in a cell causes excess of its level in neighbor cells.


Asunto(s)
Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Ácido Oxaloacético/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sinapsis/metabolismo , Adenosina Trifosfato/metabolismo , Antimicina A/análogos & derivados , Antimicina A/farmacología , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Respiración de la Célula/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Cinética , Metacrilatos/farmacología , Mitocondrias/efectos de los fármacos , Fantasmas de Imagen , Sinapsis/efectos de los fármacos , Tiazoles/farmacología , Factores de Tiempo
20.
J Endocrinol ; 251(1): 111-123, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34370682

RESUMEN

Supplementation with precursors of NAD has been shown to prevent and reverse insulin resistance, mitochondrial dysfunction, and liver damage in mouse models of diet-induced obesity. We asked whether the beneficial effects of supplementation with the NAD precursor nicotinamide riboside (NR) are dependent on mouse strain. We compared the effects of NR supplementation on whole-body energy metabolism and mitochondrial function in mildly obese C57BL/6N and C57BL/6J mice, two commonly used strains to investigate metabolism. Male C57BL/6N and C57BL/6J mice were fed a high-fat diet (HFD) or standard chow with or without NR supplementation for 8 weeks. Body and organ weights, glucose tolerance, and metabolic parameters as well as mitochondrial O2 flux in liver and muscle fibers were assessed. We found that NR supplementation had no influence on body or organ weight, glucose metabolism or hepatic lipid accumulation, energy expenditure, or metabolic flexibility but increased mitochondrial respiration in soleus muscle in both mouse strains. Strain-dependent differences were detected for body and fat depot weight, fasting blood glucose, hepatic lipid accumulation, and energy expenditure. We conclude that, in mild obesity, NR supplementation does not alter metabolic phenotype in two commonly used laboratory mouse strains.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Niacinamida/análogos & derivados , Obesidad/tratamiento farmacológico , Compuestos de Piridinio/uso terapéutico , Animales , Respiración de la Célula/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Evaluación de Medicamentos , Intolerancia a la Glucosa/prevención & control , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Niacinamida/uso terapéutico , Obesidad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA