Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Prostaglandins Other Lipid Mediat ; 153: 106524, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33418267

RESUMEN

The triplication of human chromosome 21 results in Down syndrome (DS), the most common genetic form of intellectual disability. This aneuploid condition also results in an enhanced risk of a spectrum of comorbid conditions, such as leukemia, early onset Alzheimer's disease, and diabetes. Individuals with DS also display an increased incidence of wound healing complications and resistance to solid tumor development. Due to this unique phenotype and the involvement of eicosanoids in key comorbidities like poor healing and tumor development, we hypothesized that cells from DS individuals would display altered eicosanoid production. Using age- and sex-matched dermal fibroblasts we interrogated this hypothesis. Briefly, assessment of over 90 metabolites derived from cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome p450 systems revealed a possible deficiency in the COX system. Basal gene expression and Western blotting experiments showed significantly decreased gene expression of COX1 and 2, and COX2 protein abundance in DS fibroblasts compared to euploid controls. Further, using two different stressors, scratch wound or LPS, we found that DS fibroblasts could not upregulate COX2 abundance and prostaglandin E2 production. Together, these findings show that dermal fibroblasts from DS individuals have a deficient COX2 response, which may contribute to wound healing complications and tumor resistance in DS.


Asunto(s)
Dinoprostona , Ciclooxigenasa 1 , Ciclooxigenasa 2/metabolismo , Síndrome de Down , Humanos
2.
Hum Genomics ; 13(1): 65, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823815

RESUMEN

BACKGROUND: Chronic alcohol consumption is a significant cause of liver disease worldwide. Several biochemical mechanisms have been linked to the initiation and progression of alcoholic liver disease (ALD) such as oxidative stress, inflammation, and metabolic dysregulation, including the disruption of NAD+/NADH. Indeed, an ethanol-mediated reduction in hepatic NAD+ levels is thought to be one factor underlying ethanol-induced steatosis, oxidative stress, steatohepatitis, insulin resistance, and inhibition of gluconeogenesis. Therefore, we applied a NAD+ boosting supplement to investigate alterations in the pathogenesis of early-stage ALD. METHODS: To examine the impact of NAD+ therapy on the early stages of ALD, we utilized nicotinamide mononucleotide (NMN) at 500 mg/kg intraperitoneal injection every other day, for the duration of a Lieber-DeCarli 6-week chronic ethanol model in mice. Numerous strategies were employed to characterize the effect of NMN therapy, including the integration of RNA-seq, immunoblotting, and metabolomics analysis. RESULTS: Our findings reveal that NMN therapy increased hepatic NAD+ levels, prevented an ethanol-induced increase in plasma ALT and AST, and changed the expression of 25% of the genes that were modulated by ethanol metabolism. These genes were associated with a number of pathways including the MAPK pathway. Interestingly, our analysis revealed that NMN treatment normalized Erk1/2 signaling and prevented an induction of Atf3 overexpression. CONCLUSIONS: These findings reveal previously unreported mechanisms by which NMN supplementation alters hepatic gene expression and protein pathways to impact ethanol hepatotoxicity in an early-stage murine model of ALD. Overall, our data suggest further research is needed to fully characterize treatment paradigms and biochemical implications of NAD+-based interventions.


Asunto(s)
Perfilación de la Expresión Génica , Hepatopatías Alcohólicas/tratamiento farmacológico , Mononucleótido de Nicotinamida/uso terapéutico , ARN/genética , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Enfermedad Crónica , Modelos Animales de Enfermedad , Etanol , Regulación de la Expresión Génica/efectos de los fármacos , Hepatopatías Alcohólicas/sangre , Hepatopatías Alcohólicas/genética , Metaboloma , Metabolómica , Ratones Endogámicos C57BL , Mononucleótido de Nicotinamida/farmacología , Sustancias Protectoras/metabolismo , ARN/metabolismo , Transducción de Señal/efectos de los fármacos
3.
J Proteome Res ; 18(4): 1513-1531, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30644754

RESUMEN

Mitochondrial dysfunction is one of many key factors in the etiology of alcoholic liver disease (ALD). Lysine acetylation is known to regulate numerous mitochondrial metabolic pathways, and recent reports demonstrate that alcohol-induced protein acylation negatively impacts these processes. To identify regulatory mechanisms attributed to alcohol-induced protein post-translational modifications, we employed a model of alcohol consumption within the context of wild type (WT), sirtuin 3 knockout (SIRT3 KO), and sirtuin 5 knockout (SIRT5 KO) mice to manipulate hepatic mitochondrial protein acylation. Mitochondrial fractions were examined by label-free quantitative HPLC-MS/MS to reveal competition between lysine acetylation and succinylation. A class of proteins defined as "differential acyl switching proteins" demonstrate select sensitivity to alcohol-induced protein acylation. A number of these proteins reveal saturated lysine-site occupancy, suggesting a significant level of differential stoichiometry in the setting of ethanol consumption. We hypothesize that ethanol downregulates numerous mitochondrial metabolic pathways through differential acyl switching proteins. Data are available via ProteomeXchange with identifier PXD012089.


Asunto(s)
Acilación/efectos de los fármacos , Etanol/farmacología , Mitocondrias , Proteoma , Animales , Hepatopatías Alcohólicas/metabolismo , Masculino , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteoma/química , Proteoma/efectos de los fármacos , Proteoma/metabolismo , Sirtuina 3/genética , Sirtuina 3/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo
4.
Infect Immun ; 84(4): 1137-1142, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26857573

RESUMEN

Trypanosoma cruzi infection, which is the etiological agent of Chagas disease, is associated with intense inflammation during the acute and chronic phases. The pathological progression of Chagas disease is influenced by the infiltration and transmigration of inflammatory cells across the endothelium to infected tissues, which are carefully regulated processes involving several molecular mediators, including adhesion molecules and platelet-activating factor (PAF). We have shown that PAF production is dependent upon calcium-independent group VIA phospholipase A2ß (iPLA2ß) following infection of human coronary artery endothelial cells (HCAECs) with T. cruzi, suggesting that the absence of iPLA2ß may decrease the recruitment of inflammatory cells to the heart to manage parasite accumulation. Cardiac endothelial cells isolated from iPLA2ß-knockout (iPLA2ß-KO) mice infected withT. cruzi demonstrated decreased PAF production compared to that by cells isolated from wild-type (WT) mice but demonstrated increases in adhesion molecule expression similar to those seen in WT mice. Myocardial inflammation in iPLA2ß-KO mice infected with T. cruzi was similar in severity to that in WT mice, but the iPLA2ß-KO mouse myocardium contained more parasite pseudocysts. Upon activation, macrophages from iPLA2ß-KO mice produced significantly less nitric oxide (NO) and caused lessT. cruzi inhibition than macrophages from wild-type mice. Thus, the absence of iPLA2ß activity does not influence myocardial inflammation, but iPLA2ß is essential forT. cruzi clearance.


Asunto(s)
Cardiomiopatía Chagásica/metabolismo , Cardiomiopatía Chagásica/parasitología , Regulación Enzimológica de la Expresión Génica/fisiología , Fosfolipasas A2 Grupo VI/metabolismo , Macrófagos/fisiología , Animales , Línea Celular , Eliminación de Gen , Fosfolipasas A2 Grupo VI/genética , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nitritos , Carga de Parásitos
5.
Am J Physiol Renal Physiol ; 310(10): F1074-80, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26911855

RESUMEN

Loss of integrity of the protective impermeability barrier in the urothelium has been identified as significant in bladder dysfunction. In this study, we tested the theory that the luminal layer of glycosaminoglycans (GAG) serves as an important component of barrier function. The peptide polycation protamine sulfate (PS), 1 mg/ml, was instilled intravesically for 10 min into rat bladders. Chondroitinase ABC (ChABC), 63 IU/ml, was instilled into an additional six rats for 30 min to digest the GAG layer. Unmanipulated controls and sham-injected controls were also performed. After 24 h, the rats were euthanized, the bladders were removed, and permeability was assessed in the Ussing chamber and by diffusion of FITC-labeled dextran (4 kDa) to measure macromolecular permeability. The status of tight junctions was assessed by immunofluorescence and electron microscopy. In control and sham treated rat bladders, the transepithelial electrical resistance were means of 2.5 ± 1.1 vs. 2.6 ± 1.1 vs 1.2 ± 0.5 and 1.01 ± 0.7 kΩ·cm(2) in the PS-treated and ChABC-treated rat bladders (P = 0.0016 and P = 0.0039, respectively). Similar differences were seen in dextran permeability. Histopathology showed a mild inflammation following PS treatment, but the ChABC-treated bladders were indistinguishable from controls. Tight junctions generally remained intact. ChABC digestion alone induced bladder permeability, confirming the importance of the GAG layer to bladder barrier function and supports that loss of the GAG layer seen in bladder biopsies of interstitial cystitis patients could be a significant factor producing symptoms for at least some interstitial cystitis/painful bladder syndrome patients.


Asunto(s)
Cistitis Intersticial/metabolismo , Modelos Animales de Enfermedad , Glicosaminoglicanos/fisiología , Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Animales , Condroitina ABC Liasa , Cistitis Intersticial/patología , Femenino , Ovariectomía , Permeabilidad , Ratas Sprague-Dawley , Uniones Estrechas/metabolismo , Vejiga Urinaria/patología , Urotelio/patología
6.
Am J Physiol Cell Physiol ; 307(10): C951-6, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25186013

RESUMEN

Cancer deaths are primarily caused by distant metastases, rather than by primary tumor growth; however, the role of smoking in metastasis remains unclear. We demonstrated previously that endothelial cell platelet-activating factor (PAF) production results in enhanced inflammatory cell recruitment to the lung. We propose that endothelial cell PAF accumulation plays a role in cancer cell migration to distal locations. We used cigarette smoke extract (CSE) to inhibit the activity of endothelial cell PAF acetylhydrolase (PAF-AH), which hydrolyzes and inactivates PAF, and determined whether this results in increased endothelial cell PAF accumulation and breast cancer adherence. Incubation of human lung microvascular endothelial cells (HMVEC-L) with CSE resulted in a significant inhibition of PAF-AH activity that was accompanied by increased PAF production and adherence of highly invasive MDA-MB-231 breast cancer cells. Pretreatment of HMVEC-L with (S)-bromoenol lactone to inhibit calcium-independent phospholipase A2ß (iPLA2ß, which initiates endothelial cell PAF production) prior to CSE exposure resulted in complete inhibition of MDA-MB-231 cell adherence. Similarly, pretreatment of MDA-MB-231 cells with the PAF receptor antagonist Ginkgo biloba resulted in inhibition of adherence to the endothelium. Immunoblot analysis indicated an increase in MDA-MB-231 cell PAF receptor expression with CSE exposure. Taken together, our data indicate that CSE exposure increases endothelial cell PAF production, resulting in enhanced adherence of tumor cells to the endothelium. Our in vitro data indicate that increased tumor cell adherence would lead to enhanced metastasis formation in smokers. Potential therapeutic targets include endothelial cell iPLA2ß or the tumor cell PAF receptor.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/antagonistas & inhibidores , Neoplasias de la Mama/enzimología , Adhesión Celular/fisiología , Células Endoteliales/enzimología , Pulmón/enzimología , Fumar/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Células MCF-7 , Humo/efectos adversos , Fumar/patología
7.
Free Radic Biol Med ; 196: 11-21, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36638900

RESUMEN

Cellular redox state directs differentiation of induced pluripotent stem cells (iPSC) by energy metabolism control and ROS generation. As oxidative stress and mitochondrial dysfunction have been extensively reported in Down syndrome (DS), we evaluated mitochondrial phenotypes and energy metabolism during neural differentiation of DS iPSCs to neural progenitor cells (NPCs). Our results indicate early maturation of mitochondrial networks and elevated NADPH oxidase 4 (NOX4) expression in DS iPSCs. DS cells also fail to transition from glycolysis to oxidative phosphorylation during differentiation. Specifically, DS NPCs show an increased energetic demand that is limited in their mitochondrial and glycolytic response to mitochondrial distress. Additionally, DS iPSC and NPC non-mitochondrial oxygen consumption was significantly impacted by NOX inhibition. Together, these data build upon previous evidence of accelerated neural differentiation in DS that correlates with cellular redox state. We demonstrate the potential for mitochondrial and non-mitochondrial ROS sources to impact differentiation timing in the context of DS, which could contribute to developmental deficits in this condition.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Humanos , Síndrome de Down/genética , Síndrome de Down/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Diferenciación Celular/genética , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo
8.
Redox Biol ; 64: 102792, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37390786

RESUMEN

In the U.S., alcohol-associated liver disease (ALD) impacts millions of people and is a major healthcare burden. While the pathology of ALD is unmistakable, the molecular mechanisms underlying ethanol hepatotoxicity are not fully understood. Hepatic ethanol metabolism is intimately linked with alterations in extracellular and intracellular metabolic processes, specifically oxidation/reduction reactions. The xenobiotic detoxification of ethanol leads to significant disruptions in glycolysis, ß-oxidation, and the TCA cycle, as well as oxidative stress. Perturbation of these regulatory networks impacts the redox status of critical regulatory protein thiols throughout the cell. Integrating these key concepts, our goal was to apply a cutting-edge approach toward understanding mechanisms of ethanol metabolism in disrupting hepatic thiol redox signaling. Utilizing a chronic murine model of ALD, we applied a cysteine targeted click chemistry enrichment coupled with quantitative nano HPLC-MS/MS to assess the thiol redox proteome. Our strategy reveals that ethanol metabolism largely reduces the cysteine proteome, with 593 cysteine residues significantly reduced and 8 significantly oxidized cysteines. Ingenuity Pathway Analysis demonstrates that ethanol metabolism reduces specific cysteines throughout ethanol metabolism (Adh1, Cat, Aldh2), antioxidant pathways (Prx1, Mgst1, Gsr), as well as many other biochemical pathways. Interestingly, a sequence motif analysis of reduced cysteines showed a correlation for hydrophilic, charged amino acids lysine or glutamic acid nearby. Further research is needed to determine how a reduced cysteine proteome impacts individual protein activity across these protein targets and pathways. Additionally, understanding how a complex array of cysteine-targeted post-translational modifications (e.g., S-NO, S-GSH, S-OH) are integrated to regulate redox signaling and control throughout the cell is key to the development of redox-centric therapeutic agents targeted to ameliorate the progression of ALD.


Asunto(s)
Cisteína , Compuestos de Sulfhidrilo , Ratones , Animales , Cisteína/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Proteoma/metabolismo , Proteómica , Espectrometría de Masas en Tándem , Química Clic , Oxidación-Reducción , Etanol
9.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L47-55, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21984569

RESUMEN

An early event in the pathogenesis of emphysema is the development of inflammation associated with accumulation of polymorphonuclear leukocytes (PMN) in small airways, and inflammatory cell recruitment from the circulation involves migration across endothelial and epithelial cell barriers. Platelet-activating factor (PAF) promotes transendothelial migration in several vascular beds, and we postulated that increased PAF production in the airways of smokers might enhance inflammatory cell recruitment and exacerbate inflammation. To examine this possibility, we incubated human lung microvascular endothelial cells (HMVEC-L) with cigarette smoke extract (CSE) and found that CSE inhibits PAF-acetylhydrolase (PAF-AH) activity. This enhances HMVEC-L PAF production and PMN adherence, and adherence is blocked by PAF receptor antagonists (CV3988 or ginkgolide B). CSE also inhibited PAF-AH activity of lung endothelial cells isolated from wild-type (WT) and iPLA(2)ß knockout mice, and with WT cells, CSE enhanced PAF production and RAW 264.7 cell adherence. In contrast, CSE did not affect PAF production or RAW 264.7 cell adherence to iPLA(2)ß-null cells, suggesting that iPLA(2)ß plays an important role in PAF production by lung endothelial cells. These findings suggest that inhibition of PAF-AH by components of cigarette smoke may initiate or exacerbate inflammatory lung disease by enhancing PAF production and promoting accumulation of inflammatory cells in small airways. In addition, iPLA(2)ß is identified as a potential target for therapeutic interventions to reduce airway inflammation and the progression of chronic lung disease.


Asunto(s)
Células Endoteliales/metabolismo , Fosfolipasas A2 Grupo VI , Factor de Activación Plaquetaria/metabolismo , Contaminación por Humo de Tabaco/efectos adversos , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Fosfolipasas A2 Grupo VI/deficiencia , Fosfolipasas A2 Grupo VI/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Éteres Fosfolípidos/farmacología , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores
10.
Free Radic Biol Med ; 186: 32-42, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35537597

RESUMEN

The ubiquity of cognitive deficits and early onset Alzheimer's disease in Down syndrome (DS) has focused much DS iPSC-based research on neuron degeneration and regeneration. Despite reports of elevated oxidative stress in DS brains, few studies assess the impact of this oxidative burden on iPSC differentiation. Here, we evaluate cellular specific redox differences in DS and euploid iPSCs and neural progenitor cells (NPCs) during critical intermediate stages of differentiation. Despite successful generation of NPCs, our results indicate accelerated neuroectodermal differentiation of DS iPSCs compared to isogenic, euploid controls. Specifically, DS embryoid bodies (EBs) and neural rosettes prematurely develop with distinct morphological differences from controls. Additionally, we observed developmental stage-specific alterations in mitochondrial superoxide production and SOD1/2 abundance, coupled with modulations in thioredoxin, thioredoxin reductase, and peroxiredoxin isoforms. Disruption of intracellular redox state and its associated signaling has the potential to disrupt cellular differentiation and development in DS lending to DS-specific phenotypes.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Diferenciación Celular/genética , Células Cultivadas , Síndrome de Down/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Estrés Oxidativo
11.
Free Radic Biol Med ; 172: 201-212, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34129926

RESUMEN

Down syndrome (DS) is the most common genetic cause of intellectual disability. Mechanistically, oxidative stress and mitochondrial dysfunction are reported to be etiological factors for many of the DS-related comorbidities and have previously been reported in a number of in vitro and in vivo models of DS. The purpose of this study was to test for the presence of mitochondrial dysfunction in fibroblast cells obtained via skin biopsy from individuals with DS, and to assess the impact of trisomy 21 on central carbon metabolism. Using extracellular flux assays in matched dermal fibroblasts from euploid and DS individuals, we found that basal mitochondrial dysfunction is quite mild. Stressing the cells with a cocktail of mitochondrial stressors revealed a significant mitochondrial deficit in DS cells compared to euploid controls. Evaluation of extracellular acidification rate did not reveal a baseline abnormality in glycolysis; however, metabolomic assessments utilizing isotopically labeled glucose and glutamine revealed altered central carbon metabolism in DS cells. Specifically, we observed greater glucose dependency, uptake and flux into the oxidative phase of the pentose phosphate pathway in DS fibroblasts. Furthermore, using induced pluripotent stem cells (iPSC) we found that mitochondrial function in DS iPSCs was similar to the previously published studies employing fetal cells. Together, these data indicate that aberrant central carbon metabolism is a candidate mechanism for stress-related mitochondrial dysfunction in DS.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Carbono/metabolismo , Células Cultivadas , Síndrome de Down/genética , Síndrome de Down/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mitocondrias
12.
Free Radic Biol Med ; 162: 65-76, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33279619

RESUMEN

The dithiocarbamate fungicide maneb (MB) has attracted interest due to increasing concern of the negative health effects of pesticides, as well as its association with Parkinson's disease (PD). Our laboratory has previously reported distinct phenotypic changes of neuroblastoma cells exposed to acute, sub-toxic levels of MB, including decreased mitochondrial respiration, altered lactate dynamics, and metabolic stress. In this study, we aimed to further define the specific molecular mechanisms of MB toxicity through the comparison of several thiol-containing compounds and their effects on cellular energy metabolism and thiol redox nodes. Extracellular flux analyses and stable isotope labeled tracer metabolomics were employed to evaluate alterations in energy metabolism of SK-N-AS human neuroblastoma cells after acute exposure of an array of compounds, including dithiocarbamates (maneb, nabam, zineb) and other thiol-containing small molecules (glutathione, N-acetylcysteine). These studies revealed MB and its methylated form (MeDTC) as unique toxicants with significant alterations to mitochondrial respiration, proliferation, and glycolysis. We observed MB to significantly impact cellular thiol redox status by oxidizing cellular glutathione and altering the thiol redox status of peroxiredoxin 3 (Prx3, mitochondrial) after acute exposure. Redox Western blotting revealed a MB-specific modification of cellular Prx3, strengthening the argument that MB can preferentially target mitochondrial enzymes containing reactive cysteine thiols. Further, stable isotope tracer metabolomics confirmed our energetics assessments, and demonstrated that MB exposure results in acute derangement of central carbon metabolism. Specifically, we observed shunting of cellular glucose into the pentose-phosphate pathway and reduction of TCA intermediates derived from glucose and glutamine. Also, we report novel lactate utilization for TCA enrichment and glutathione synthesis after MB exposure. In summary, our results further confirm that MB exerts its toxic effects via thiol modification, and significantly transforms central carbon metabolism.


Asunto(s)
Maneb , Enfermedad de Parkinson , Carbono , Humanos , Maneb/toxicidad , Oxidación-Reducción , Compuestos de Sulfhidrilo
13.
Cell Chem Biol ; 27(2): 206-213.e6, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31767537

RESUMEN

Post-translational modifications (PTMs) regulate enzyme structure and function to expand the functional proteome. Many of these PTMs are derived from cellular metabolites and serve as feedback and feedforward mechanisms of regulation. We have identified a PTM that is derived from the glycolytic by-product, methylglyoxal. This reactive metabolite is rapidly conjugated to glutathione via glyoxalase 1, generating lactoylglutathione (LGSH). LGSH is hydrolyzed by glyoxalase 2 (GLO2), cycling glutathione and generating D-lactate. We have identified the non-enzymatic acyl transfer of the lactate moiety from LGSH to protein Lys residues, generating a "LactoylLys" modification on proteins. GLO2 knockout cells have elevated LGSH and a consequent marked increase in LactoylLys. Using an alkyne-tagged methylglyoxal analog, we show that these modifications are enriched on glycolytic enzymes and regulate glycolysis. Collectively, these data suggest a previously unexplored feedback mechanism that may serve to regulate glycolytic flux under hyperglycemic or Warburg-like conditions.


Asunto(s)
Glutatión/análogos & derivados , Lisina/metabolismo , Tioléster Hidrolasas/metabolismo , Alquinos/química , Glutatión/química , Glutatión/metabolismo , Glucólisis/efectos de los fármacos , Glicosilación , Células HEK293 , Humanos , Lactoilglutatión Liasa/deficiencia , Lactoilglutatión Liasa/genética , Lactoilglutatión Liasa/metabolismo , Piruvaldehído/análogos & derivados , Piruvaldehído/química , Piruvaldehído/farmacología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Tioléster Hidrolasas/deficiencia , Tioléster Hidrolasas/genética
14.
Sci Rep ; 9(1): 7776, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31123328

RESUMEN

Alcoholic liver disease (ALD) is a significant health hazard and economic burden affecting approximately 10 million people in the United States. ALD stems from the production of toxic-reactive metabolites, oxidative stress and fat accumulation in hepatocytes which ultimately results in hepatocyte death promoting hepatitis and fibrosis deposition. Monocyte-derived infiltrating Ly6Chi and Ly6Clow macrophages are instrumental in perpetuating and resolving the hepatitis and fibrosis associated with ALD pathogenesis. In the present study we isolated liver infiltrating macrophages from mice on an ethanol diet and subjected them to metabolomic and proteomic analysis to provide a broad assessment of the cellular metabolite and protein differences between infiltrating macrophage phenotypes. We identified numerous differentially regulated metabolites and proteins between Ly6Chi and Ly6Clow macrophages. Bioinformatic analysis for pathway enrichment of the differentially regulated metabolites showed a significant number of metabolites involved in the processes of glycerophospholipid metabolism, arachidonic acid metabolism and phospholipid biosynthesis. From analysis of the infiltrating macrophage proteome, we observed a significant enrichment in the biological processes of antigen presentation, actin polymerization and organization, phagocytosis and apoptotic regulation. The data presented herein could yield exciting new research avenues for the analysis of signaling pathways regulating macrophage polarization in ALD.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Etanol/farmacología , Hepatopatías Alcohólicas/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Animales , Biología Computacional , Hígado/efectos de los fármacos , Hígado/patología , Hepatopatías Alcohólicas/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Estrés Oxidativo/efectos de los fármacos , Proteómica
15.
PLoS One ; 14(11): e0223254, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31714914

RESUMEN

Down syndrome (DS) is a chromosomal disorder caused by trisomy of chromosome 21 (Ts21). Unbalanced karyotypes can lead to dysfunction of the proteostasis network (PN) and disrupted proteostasis is mechanistically associated with multiple DS comorbidities. Autophagy is a critical component of the PN that has not previously been investigated in DS. Based on our previous observations of PN disruption in DS, we investigated possible dysfunction of the autophagic machinery in human DS fibroblasts and other DS cell models. Following induction of autophagy by serum starvation, DS fibroblasts displayed impaired autophagic flux indicated by autophagolysosome accumulation and elevated p62, NBR1, and LC3-II abundance, compared to age- and sex-matched, euploid (CTL) fibroblasts. While lysosomal physiology was unaffected in both groups after serum starvation, we observed decreased basal abundance of the Soluble N-ethylmaleimide-sensitive-factor Attachment protein Receptor (SNARE) family members syntaxin 17 (STX17) and Vesicle Associated Membrane Protein 8 (VAMP8) indicating that decreased autophagic flux in DS is due at least in part to a possible impairment of autophagosome-lysosome fusion. This conclusion was further supported by the observation that over-expression of either STX17 or VAMP8 in DS fibroblasts restored autophagic degradation and reversed p62 accumulation. Collectively, our results indicate that impaired autophagic clearance is a characteristic of DS cells that can be reversed by enhancement of SNARE protein expression and provides further evidence that PN disruption represents a candidate mechanism for multiple aspects of pathogenesis in DS and a possible future target for therapeutic intervention.


Asunto(s)
Autofagia , Síndrome de Down/metabolismo , Síndrome de Down/patología , Proteínas SNARE/metabolismo , Línea Celular , Femenino , Regulación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Transporte de Proteínas , Proteínas R-SNARE/metabolismo , Proteínas de Unión al ARN/metabolismo
16.
PLoS One ; 10(6): e0129466, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26057882

RESUMEN

PURPOSE: The differentiated superficial cells of the urothelium restrict urine flow into the bladder wall. We have demonstrated that urothelial cells isolated from bladders of patients with interstitial cystitis/painful bladder syndrome (IC/PBS) fail to release PGE2 in response to tryptase. This study examines the expression of PGE2 synthesis and degradation enzymes in urothelial cells during differentiation. MATERIALS AND METHODS: We measured immunoprotein expression of cyclooxygenase-2 (COX-2), prostaglandin E2 synthase (PGES) and 15-hydroxyprostaglandin dehydrogenase (PGDH) in human urothelial cells and in immortalized urothelial cells isolated from the bladders of IC/PBS patients or normal subjects during stratification and differentiation produced by increased calcium and fetal bovine serum (Ca/FBS) in the culture medium for 1, 3 and 7 days. RESULTS: PGES immunoprotein expression increased during differentiation in normal and IC/PBS urothelial cells. COX-2 expression also increased in cells from normal patients following differentiation. Remarkably, no COX-2 expression was detectable in urothelial cells isolated from 3 out of 4 IC/PBS patients. PGDH immunoprotein expression decreased in normal cells after 1 and 3 days of Ca/FBS addition, but returned to normal after 7 days. PGDH expression was unchanged during differentiation at 1 and 3 days, but was more than 2-fold higher at 7 days compared to day 0 in the IC/PBS cells. Urothelial cells isolated from IC/PBS patients demonstrated no PGE2 release in response to tryptase under any of the experimental conditions studied. CONCLUSIONS: Taken together, our results indicate that PGE2 release is compromised during stratification and differentiation in IC/PBS urothelium and may contribute to impaired barrier function.


Asunto(s)
Diferenciación Celular/fisiología , Cistitis Intersticial/metabolismo , Dinoprostona/metabolismo , Vejiga Urinaria/metabolismo , Urotelio/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Cistitis Intersticial/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Triptasas/metabolismo , Vejiga Urinaria/patología , Urotelio/patología
17.
Physiol Rep ; 2(1): e00196, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24744875

RESUMEN

Both acute and chronic phases of Trypanosoma cruzi (T. cruzi) infection are characterized by tissue inflammation, mainly in the heart. A key step in the inflammatory process is the transmigration of inflammatory cells across the endothelium to underlying infected tissues. We observed increased arachidonic acid release and platelet-activating factor (PAF) production in human coronary artery endothelial cells (HCAEC) at up to 96 h of T. cruzi infection. Arachidonic acid release is mediated by activation of the calcium-independent phospholipase A2 (iPLA2) isoforms iPLA2 ß and iPLA2 γ, whereas PAF production was dependent upon iPLA2 ß activation alone. Trypanosoma cruzi infection also resulted in increased cell surface expression of adhesion molecules. Increased adherence of inflammatory cells to T. cruzi-infected endothelium was blocked by inhibition of endothelial cell iPLA2 ß or by blocking the PAF receptor on inflammatory cells. This suggests that PAF, in combination with adhesion molecules, might contribute to parasite clearing in the heart by recruiting inflammatory cells to the endothelium.

18.
PLoS One ; 8(7): e69948, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922867

RESUMEN

The pathogenesis of interstitial cystitis/painful bladder syndrome (IC/PBS) is multifactorial, but likely involves urothelial cell dysfunction and mast cell accumulation in the bladder wall. Activated mast cells in the bladder wall release several inflammatory mediators, including histamine and tryptase. We determined whether mitogen-activated protein (MAP) kinases are activated in response to tryptase stimulation of urothelial cells derived from human normal and IC/PBS bladders. Tryptase stimulation of normal urothelial cells resulted in a 2.5-fold increase in extracellular signal regulated kinase 1/2 (ERK 1/2). A 5.5-fold increase in ERK 1/2 activity was observed in urothelial cells isolated from IC/PBS bladders. No significant change in p38 MAP kinase was observed in tryptase-stimulated normal urothelial cells but a 2.5-fold increase was observed in cells isolated from IC/PBS bladders. Inhibition of ERK 1/2 with PD98059 or inhibition of p38 MAP kinase with SB203580 did not block tryptase-stimulated iPLA2 activation. Incubation with the membrane phospholipid-derived PLA2 hydrolysis product lysoplasmenylcholine increased ERK 1/2 activity, suggesting the iPLA2 activation is upstream of ERK 1/2. Real time measurements of impedance to evaluate wound healing of cell cultures indicated increased healing rates in normal and IC/PBS urothelial cells in the presence of tryptase, with inhibition of ERK 1/2 significantly decreasing the wound healing rate of IC/PBS urothelium. We conclude that activation of ERK 1/2 in response to tryptase stimulation may facilitate wound healing or cell motility in areas of inflammation in the bladder associated with IC/PBS.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Triptasas/metabolismo , Urotelio/citología , Urotelio/metabolismo , Línea Celular , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , Immunoblotting , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación , Piridinas/farmacología , Triptasas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA