Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Genet ; 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38910033

RESUMEN

The emergence of aerobic respiration created unprecedented bioenergetic advantages, while imposing the need to protect critical genetic information from reactive byproducts of oxidative metabolism (i.e., reactive oxygen species, ROS). The evolution of histone proteins fulfilled the need to shield DNA from these potentially damaging toxins, while providing the means to compact and structure massive eukaryotic genomes. To date, several metabolism-linked histone post-translational modifications (PTMs) have been shown to regulate chromatin structure and gene expression. However, whether and how PTMs enacted by metabolically produced ROS regulate adaptive chromatin remodeling remain relatively unexplored. Here, we review novel mechanistic insights into the interactions of ROS with histones and their consequences for the control of gene expression regulation, cellular plasticity, and behavior.

2.
Nature ; 568(7751): 254-258, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30842661

RESUMEN

Mitochondrial metabolism is an attractive target for cancer therapy1,2. Reprogramming metabolic pathways could improve the ability of metabolic inhibitors to suppress cancers with limited treatment options, such as triple-negative breast cancer (TNBC)1,3. Here we show that BTB and CNC homology1 (BACH1)4, a haem-binding transcription factor that is increased in expression in tumours from patients with TNBC, targets mitochondrial metabolism. BACH1 decreases glucose utilization in the tricarboxylic acid cycle and negatively regulates transcription of electron transport chain (ETC) genes. BACH1 depletion by shRNA or degradation by hemin sensitizes cells to ETC inhibitors such as metformin5,6, suppressing growth of both cell line and patient-derived tumour xenografts. Expression of a haem-resistant BACH1 mutant in cells that express a short hairpin RNA for BACH1 rescues the BACH1 phenotype and restores metformin resistance in hemin-treated cells and tumours7. Finally, BACH1 gene expression inversely correlates with ETC gene expression in tumours from patients with breast cancer and in other tumour types, which highlights the clinical relevance of our findings. This study demonstrates that mitochondrial metabolism can be exploited by targeting BACH1 to sensitize breast cancer and potentially other tumour tissues to mitochondrial inhibitors.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Hemina/uso terapéutico , Metformina/uso terapéutico , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Ciclo del Ácido Cítrico/fisiología , Transporte de Electrón/genética , Femenino , Glucosa/metabolismo , Hemina/metabolismo , Xenoinjertos , Humanos , Metformina/metabolismo , Ratones , Ratones Desnudos , Mitocondrias/genética , Proteolisis , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 119(29): e2110348119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858297

RESUMEN

The dichotomous behavior of superoxide dismutase-2 (SOD2) in cancer biology has long been acknowledged and more recently linked to different posttranslational forms of the enzyme. However, a distinctive activity underlying its tumor-promoting function is yet to be described. Here, we report that acetylation, one of such posttranslational modifications (PTMs), increases SOD2 affinity for iron, effectively changing the biochemical function of this enzyme from that of an antioxidant to a demethylase. Acetylated, iron-bound SOD2 localizes to the nucleus, promoting stem cell gene expression via removal of suppressive epigenetic marks such as H3K9me3 and H3K927me3. Particularly, H3K9me3 was specifically removed from regulatory regions upstream of Nanog and Oct-4, two pluripotency factors involved in cancer stem cell reprogramming. Phenotypically, cells expressing nucleus-targeted SOD2 (NLS-SOD2) have increased clonogenicity and metastatic potential. FeSOD2 operating as H3 demethylase requires H2O2 as substrate, which unlike cofactors of canonical demethylases (i.e., oxygen and 2-oxoglutarate), is more abundant in tumor cells than in normal tissue. Therefore, our results indicate that FeSOD2 is a demethylase with unique activities and functions in the promotion of cancer evolution toward metastatic phenotypes.


Asunto(s)
Neoplasias de la Mama , Núcleo Celular , Histona Demetilasas , Hierro , Células Madre Neoplásicas , Superóxido Dismutasa , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Núcleo Celular/enzimología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Procesamiento Proteico-Postraduccional , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
4.
Cancer Causes Control ; 34(7): 621-624, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37081154

RESUMEN

Engagement of community participation is an innovative driver of modern research. However, to benefit the communities being studied, it is imperative to continuously evaluate ethical considerations, the relationship dynamic between researchers and community members, and the responsiveness of research teams to the needs and preferences of communities. Northwestern University's Center for Health Equity Transformation founded a community scientist program in 2018 that implemented a study using the Community-Based Participatory Research (CBPR) model. This project is an ongoing study of heavy metal exposure by geographic location in Chicago. Community scientists from various backgrounds, communities, and organizations formed an advisory panel, partnering with the cancer research team. This commentary describes lessons learned in structuring meaningful community involvement and benefit in CBPR, with a focus on three lessons learned that relate to ethics, relationships, and responsiveness. Our findings lay new groundwork for iteratively shaping best practices in CBPR.


Asunto(s)
Investigación Participativa Basada en la Comunidad , Médicos , Humanos , Proyectos de Investigación , Chicago
5.
Semin Cancer Biol ; 76: 287-291, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34563651

RESUMEN

There are several sources of heavy metal exposures whether occupational or environmental. These are connected both with the existence of natural reservoirs of metal toxicants or human activity such as mining, welding and construction. In general, exposure to heavy metals, such as cadmium (Cd), mercury (Hg), nickel (Ni), lead (Pb) and metalloids, such as arsenic (As), has been associated with diseases including neurodegenerative diseases, diabetes and cancer. Common to these diseases is the loss of cellular physiologic performance and phenotype required for proper function. On the metal side, electrophilic behavior that disrupts the electronic (or redox) state of cells is a common feature. This suggests that there may be a connection between changes to the redox equilibrium of cells caused by environmental exposures to heavy metals and the pathogenic effects of such exposures. In this mini-review, we will focus on two environmental contaminants cadmium (a metal) and arsenic (a metalloid) and explore their interactions with living organisms from the perspective of their electrophilic chemical reactivity that underlies both their potential as carcinogens and as drivers of more aggressive tumor phenotypes.


Asunto(s)
Arsénico/efectos adversos , Cadmio/efectos adversos , Carcinogénesis/inducido químicamente , Animales , Humanos , Fenotipo
6.
Proc Natl Acad Sci U S A ; 116(47): 23534-23541, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31591207

RESUMEN

Mitochondrial superoxide dismutase (SOD2) suppresses tumor initiation but promotes invasion and dissemination of tumor cells at later stages of the disease. The mechanism of this functional switch remains poorly defined. Our results indicate that as SOD2 expression increases acetylation of lysine 68 ensues. Acetylated SOD2 promotes hypoxic signaling via increased mitochondrial reactive oxygen species (mtROS). mtROS, in turn, stabilize hypoxia-induced factor 2α (HIF2α), a transcription factor upstream of "stemness" genes such as Oct4, Sox2, and Nanog. In this sense, our findings indicate that SOD2K68Ac and mtROS are linked to stemness reprogramming in breast cancer cells via HIF2α signaling. Based on these findings we propose that, as tumors evolve, the accumulation of SOD2K68Ac turns on a mitochondrial pathway to stemness that depends on HIF2α and may be relevant for the progression of breast cancer toward poor outcomes.


Asunto(s)
Neoplasias de la Mama/patología , Autorrenovación de las Células/fisiología , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/fisiología , Superóxido Dismutasa/fisiología , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Neoplasias de la Mama/metabolismo , Reprogramación Celular , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Peróxido de Hidrógeno/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/enzimología , Invasividad Neoplásica , Proteínas de Neoplasias/química , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Superóxido Dismutasa/química
7.
FASEB J ; 34(12): 16034-16048, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047385

RESUMEN

Inorganic arsenic (iAs/As2 O32- ) is an environmental toxicant found in watersheds around the world including in densely populated areas. iAs is a class I carcinogen known to target the skin, lungs, bladder, and digestive organs, but its role as a primary breast carcinogen remains controversial. Here, we examined a different possibility: that exposure to iAs promotes the transition of well-differentiated epithelial breast cancer cells characterized by estrogen and progesterone receptor expression (ER+/PR+), to more basal phenotypes characterized by active proliferation, and propensity to metastasis in vivo. Our results indicate two clear phenotypic responses to low-level iAs that depend on the duration of the exposure. Short-term pulses of iAs activate ER signaling, consistent with its reported pseudo-estrogen activity, but longer-term, chronic treatments for over 6 months suppresses both ER and PR expression and signaling. In fact, washout of these chronically exposed cells for up to 1 month failed to fully reverse the transcriptional and phenotypic effects of prolonged treatments, indicating durable changes in cellular physiologic identity. RNA-seq studies found that chronic iAs drives the transition toward more basal phenotypes characterized by impaired hormone receptor signaling despite the conservation of estrogen receptor expression. Because treatments for breast cancer patients are largely designed based on the detection of hormone receptor expression, our results suggest greater scrutiny of ER+ cancers in patients exposed to iAs, because these tumors may spawn more aggressive phenotypes than unexposed ER+ tumors, in particular, basal subtypes that tend to develop therapy resistance and metastasis.


Asunto(s)
Arsénico/fisiología , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/patología , Mama/efectos de los fármacos , Mama/patología , Animales , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Trends Biochem Sci ; 41(7): 568-577, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27180143

RESUMEN

ATP and reactive oxygen species (ROS) are signaling molecules that control cellular function and phenotype. Mitochondria produce both ATP and ROS. Since the electrons needed to generate either ATP or ROS originate from NADH/FADH2, the mechanism through which electrons flow towards oxygen determines yields and whether ATP or ROS prevails. Alterations in the electron flow impact cells dramatically, such as by supporting specialization (which requires high ATP) or imposing dedifferentiation. High ROS, facilitated by enzymes such as superoxide dismutase 2 (SOD2) that enhance mitochondrial hydrogen peroxide (mtH2O2), are normally linked to dedifferentiation of somatic cells. Here we propose that combined high mtH2O2 and mitochondrial unfolded protein response (UPR(mt)) activation are essential for somatic dedifferentiation programs and the acquisition of stem-like properties in reparative processes and disease.


Asunto(s)
Mitocondrias/metabolismo , Superóxido Dismutasa/metabolismo , Respuesta de Proteína Desplegada , Animales , Humanos , Peróxido de Hidrógeno/metabolismo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
9.
J Neurosci ; 39(43): 8576-8583, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31527120

RESUMEN

Type 2 diabetes mellitus (T2DM) is a risk factor for the development of late-onset Alzheimer's disease (AD). However, the mechanism underlying the development of late-onset AD is largely unknown. Here we show that levels of the endothelial-enriched protein caveolin-1 (Cav-1) are reduced in the brains of T2DM patients compared with healthy aging, and inversely correlated with levels of ß-amyloid (Aß). Depletion of Cav-1 is recapitulated in the brains of db/db (Leprdb ) diabetic mice and corresponds with recognition memory deficits as well as the upregulation of amyloid precursor protein (APP), BACE-1, a trending increase in ß-amyloid Aß42/40 ratio and hyperphosphorylated tau (p-tau) species. Importantly, we show that restoration of Cav-1 levels in the brains of male db/db mice using adenovirus overexpressing Cav-1 (AAV-Cav-1) rescues learning and memory deficits and reduces pathology (i.e., APP, BACE-1 and p-tau levels). Knocking down Cav-1 using shRNA in HEK cells expressing the familial AD-linked APPswe mutant variant upregulates APP, APP carboxyl terminal fragments, and Aß levels. In turn, rescue of Cav-1 levels restores APP metabolism. Together, these results suggest that Cav-1 regulates APP metabolism, and that depletion of Cav-1 in T2DM promotes the amyloidogenic processing of APP and hyperphosphorylation of tau. This may suggest that depletion of Cav-1 in T2DM underlies, at least in part, the development of AD and imply that restoration of Cav-1 may be a therapeutic target for diabetic-associated sporadic AD.SIGNIFICANCE STATEMENT More than 95% of the Alzheimer's patients have the sporadic late-onset form (LOAD). The cause for late-onset Alzheimer's disease is unknown. Patients with Type 2 diabetes mellitus have considerably higher incidence of cognitive decline and AD compared with the general population, suggesting a common mechanism. Here we show that the expression of caveolin-1 (Cav-1) is reduced in the brain in Type 2 diabetes mellitus. In turn, reduced Cav-1 levels induce AD-associated neuropathology and learning and memory deficits. Restoration of Cav-1 levels rescues these deficits. This study unravels signals underlying LOAD and suggests that restoration of Cav-1 may be an effective therapeutic target.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/patología , Caveolina 1/genética , Diabetes Mellitus Tipo 2/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Caveolina 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Fosforilación
10.
Arterioscler Thromb Vasc Biol ; 39(6): 1191-1202, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30943774

RESUMEN

Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-ß (transforming growth factor-ß) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-ß production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-ß, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-ß-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-ß signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.


Asunto(s)
Caveolina 1/deficiencia , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular , Adolescente , Adulto , Anciano , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Estudios de Casos y Controles , Caveolina 1/genética , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Vesículas Extracelulares/patología , Femenino , Humanos , Hipoxia/complicaciones , Indoles , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Hipertensión Arterial Pulmonar/etiología , Hipertensión Arterial Pulmonar/patología , Pirroles , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Smad/metabolismo , Adulto Joven
11.
Am J Physiol Heart Circ Physiol ; 317(1): H164-H167, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31172808

RESUMEN

Cardio-oncology has emerged as an exciting new field at the intersection of cardiology and oncology. While improved oncology treatment efficacy has increased survival rates in cancer patients, the long-term cardiovascular consequences of this life-saving treatment have become more clinically relevant. Both traditional and newer (targeted) cancer therapies can have cardiovascular and metabolic sequelae, resulting in heart failure, coronary artery disease, myocarditis, pericardial disease, hypertension, and vascular and metabolic perturbations (Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. N Engl J Med 375: 1457-1467, 2016). Both acute and chronic cardiovascular toxicities have proven challenging for clinicians and patients, significantly contributing to morbidity and mortality. Although chronic cardiovascular disease affects a growing number of cancer survivors (~17 million in the United States in 2019), cardiovascular toxicities associated with cancer and cancer therapies are poorly understood mechanistically. To balance potential damage to the cardiovascular system with effective and efficient cancer treatment, novel strategies are sorely needed. This perspective focuses on an assembly of articles that discuss novel means of counteracting adverse cardiovascular events in response to anticancer therapy. In light of new clinical syndromes in cardiology due to cancer therapies, we hope to highlight promising research opportunities offered by cardio-oncology (Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, Moslehi JJ. Cardio-oncology: how new targeted cancer therapies and precision medicine can inform cardiovascular discovery. Circulation 132: 2248-2258, 2015.).


Asunto(s)
Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Sistema Cardiovascular/efectos de los fármacos , Cardiopatías/inducido químicamente , Animales , Cardiotoxicidad , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Cardiopatías/prevención & control , Humanos , Sustancias Protectoras/uso terapéutico
12.
Anesthesiology ; 131(6): 1301-1315, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31658116

RESUMEN

BACKGROUND: Sevoflurane with its antiinflammatory properties has shown to decrease mortality in animal models of sepsis. However, the underlying mechanism of its beneficial effect in this inflammatory scenario remains poorly understood. Macrophages play an important role in the early stage of sepsis as they are tasked with eliminating invading microbes and also attracting other immune cells by the release of proinflammatory cytokines such as interleukin-1ß, interleukin-6, and tumor necrosis factor-α. Thus, the authors hypothesized that sevoflurane mitigates the proinflammatory response of macrophages, while maintaining their bactericidal properties. METHODS: Murine bone marrow-derived macrophages were stimulated in vitro with lipopolysaccharide in the presence and absence of 2% sevoflurane. Expression of cytokines and inducible NO synthase as well as uptake of fluorescently labeled Escherichia coli (E. coli) were measured. The in vivo endotoxemia model consisted of an intraperitoneal lipopolysaccharide injection after anesthesia with either ketamine and xylazine or 4% sevoflurane. Male mice (n = 6 per group) were observed for a total of 20 h. During the last 30 min fluorescently labeled E. coli were intraperitoneally injected. Peritoneal cells were extracted by peritoneal lavage and inducible NO synthase expression as well as E. coli uptake by peritoneal macrophages was determined using flow cytometry. RESULTS: In vitro, sevoflurane enhanced lipopolysaccharide-induced inducible NO synthase expression after 8 h by 466% and increased macrophage uptake of fluorescently labeled E. coli by 70% compared with vehicle-treated controls. Inhibiting inducible NO synthase expression pharmacologically abolished this increase in bacteria uptake. In vivo, inducible NO synthase expression was increased by 669% and phagocytosis of E. coli by 49% compared with the control group. CONCLUSIONS: Sevoflurane enhances phagocytosis of bacteria by lipopolysaccharide-challenged macrophages in vitro and in vivo via an inducible NO synthase-dependent mechanism. Thus, sevoflurane potentiates bactericidal and antiinflammatory host-defense mechanisms in endotoxemia.


Asunto(s)
Antiinflamatorios/farmacología , Regulación Enzimológica de la Expresión Génica , Macrófagos/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fagocitosis/fisiología , Sevoflurano/farmacología , Animales , Actividad Bactericida de la Sangre/efectos de los fármacos , Actividad Bactericida de la Sangre/fisiología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/genética , Fagocitosis/efectos de los fármacos , Células RAW 264.7
13.
Am J Physiol Regul Integr Comp Physiol ; 314(2): R181-R190, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046313

RESUMEN

Skeletal muscle insulin resistance is a hallmark of Type 2 diabetes (T2DM) and may be exacerbated by protein modifications by methylglyoxal (MG), known as dicarbonyl stress. The glyoxalase enzyme system composed of glyoxalase 1/2 (GLO1/GLO2) is the natural defense against dicarbonyl stress, yet its protein expression, activity, and regulation remain largely unexplored in skeletal muscle. Therefore, this study investigated dicarbonyl stress and the glyoxalase enzyme system in the skeletal muscle of subjects with T2DM (age: 56 ± 5 yr.; BMI: 32 ± 2 kg/m2) compared with lean healthy control subjects (LHC; age: 27 ± 1 yr.; BMI: 22 ± 1 kg/m2). Skeletal muscle biopsies obtained from the vastus lateralis at basal and insulin-stimulated states of the hyperinsulinemic (40 mU·m-2·min-1)-euglycemic (5 mM) clamp were analyzed for proteins related to dicarbonyl stress and glyoxalase biology. At baseline, T2DM had increased carbonyl stress and lower GLO1 protein expression (-78.8%), which inversely correlated with BMI, percent body fat, and HOMA-IR, while positively correlating with clamp-derived glucose disposal rates. T2DM also had lower NRF2 protein expression (-31.6%), which is a positive regulator of GLO1, while Keap1 protein expression, a negative regulator of GLO1, was elevated (207%). Additionally, insulin stimulation during the clamp had a differential effect on NRF2, Keap1, and MG-modified protein expression. These data suggest that dicarbonyl stress and the glyoxalase enzyme system are dysregulated in T2DM skeletal muscle and may underlie skeletal muscle insulin resistance. Whether these phenotypic differences contribute to the development of T2DM warrants further investigation.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Resistencia a la Insulina , Lactoilglutatión Liasa/metabolismo , Carbonilación Proteica , Músculo Cuádriceps/enzimología , Adulto , Aldehído Reductasa/metabolismo , Glucemia/metabolismo , Estudios de Casos y Controles , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Insulina/sangre , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Masculino , Persona de Mediana Edad , Factor 2 Relacionado con NF-E2/metabolismo , Triosa-Fosfato Isomerasa/metabolismo
14.
Biochim Biophys Acta Bioenerg ; 1858(8): 628-632, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28087256

RESUMEN

Reactive oxygen species (ROS) largely originating in the mitochondria play essential roles in the metabolic and (epi)genetic reprogramming of cancer cell evolution towards more aggressive phenotypes. Recent studies have indicated that the activity of superoxide dismutase (SOD2) may promote tumor progression by serving as a source of hydrogen peroxide (H2O2). H2O2 is a form of ROS that is particularly active as a redox agent affecting cell signaling due to its ability to freely diffuse out of the mitochondria and alter redox active amino acid residues on regulatory proteins. Therefore, there is likely a dichotomy whereas SOD2 can be considered a protective antioxidant, as well as a pro-oxidant during cancer progression, with these effects depending on the accumulation and detoxification of H2O2. Glutathione peroxidase-1 GPX1, is a selenium-dependent scavenger of H2O2 which partitions between the mitochondria and the cytosol. Epidemiologic studies indicated that allelic variations in the SOD2 and GPX1 genes alter the distribution and relative concentrations of SOD2 and GPX1 in mitochondria, thereby affecting the dynamic between the production and elimination of H2O2. Experimental and epidemiological evidence supporting a conflicting role of SOD2 in tumor biology, and epidemiological evidence that SOD2 and GPX1 can interact to affect cancer risk and progression indicated that it is the net accumulation of mitochondrial H2O2 (mtH2O2) resulting from of the balance between the activities SOD2 and anti-oxidants such as GPX1 that determines whether SOD2 prevents or promotes oncogenesis. In this review, research supporting the idea that GPX1 is a gatekeeper restraining the oncogenic power of mitochondrial ROS generated by SOD2 is presented. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Asunto(s)
Transformación Celular Neoplásica , Glutatión Peroxidasa/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología , Neoplasias/enzimología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/fisiología , Alelos , Progresión de la Enfermedad , Glutatión Peroxidasa/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Proteínas Mitocondriales/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/epidemiología , Oxidación-Reducción , Superóxido Dismutasa/genética , Glutatión Peroxidasa GPX1
15.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L760-L771, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28188225

RESUMEN

Endothelial cell (EC) activation and vascular injury are hallmark features of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Caveolin-1 (Cav-1) is highly expressed in pulmonary microvascular ECs and plays a key role in maintaining vascular homeostasis. The aim of this study was to determine if the lung inflammatory response to Escherichia coli lipopolysaccharide (LPS) promotes priming of ECs via Cav-1 depletion and if this contributes to the onset of pulmonary vascular remodeling. To test the hypothesis that depletion of Cav-1 primes ECs to respond to profibrotic signals, C57BL6 wild-type (WT) mice (Tie2.Cre-;Cav1fl/fl ) were exposed to nebulized LPS (10 mg; 1 h daily for 4 days) and compared with EC-specific Cav1-/- (Tie2.Cre+;Cav1fl/fl ). After 96 h of LPS exposure, total lung Cav-1 and bone morphogenetic protein receptor type II (BMPRII) expression were reduced in WT mice. Moreover, plasma albumin leakage, infiltration of immune cells, and levels of IL-6/IL-6R and transforming growth factor-ß (TGF-ß) were elevated in both LPS-treated WT and EC-Cav1-/- mice. Finally, EC-Cav1-/- mice exhibited a modest increase in microvascular thickness basally and even more so on exposure to LPS (96 h). EC-Cav1-/- mice and LPS-treated WT mice exhibited reduced BMPRII expression and endothelial nitric oxide synthase uncoupling, which along with increased TGF-ß promoted TGFßRI-dependent SMAD-2/3 phosphorylation. Finally, human lung sections from patients with ARDS displayed reduced EC Cav-1 expression, elevated TGF-ß levels, and severe pulmonary vascular remodeling. Thus EC Cav-1 depletion, oxidative stress-mediated reduction in BMPRII expression, and enhanced TGF-ß-driven SMAD-2/3 signaling promote pulmonary vascular remodeling in inflamed lungs.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Caveolina 1/metabolismo , Células Endoteliales/patología , Inflamación/patología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular , Actinas/metabolismo , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Adulto , Anciano , Animales , Líquido del Lavado Bronquioalveolar , Forma de la Célula/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/farmacología , Lipopolisacáridos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Modelos Biológicos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteolisis/efectos de los fármacos , Arteria Pulmonar/patología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/patología , Factores de Tiempo , Remodelación Vascular/efectos de los fármacos
16.
Breast Cancer Res Treat ; 163(2): 321-330, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28251385

RESUMEN

INTRODUCTION: Non-Latina black breast cancer patients experience a shorter survival from breast cancer than their non-Latina white counterparts. We compared breast cancer-specific survival for the subset of black and white patients with estrogen and/or progesterone receptor-positive tumors that are generally targeted with endocrine therapy. METHODS: Using data collected from a population-based cohort of breast cancer patients from Chicago, IL, Kaplan-Meier survival curves and hazard functions were generated and proportional hazards models were estimated to determine the black/white disparity in time to death from breast cancer while adjusting for age at diagnosis, patient characteristics, treatment-related variables, and tumor grade and stage. RESULTS: In regression models, hazard of breast cancer death among ER/PR-positive patients was at least 4 times higher for black than for white patients in all models tested. Notably, even after adjusting for stage at diagnosis, tumor grade, and treatment variables (including initiation of systemic adjuvant therapies), the hazard ratio for death from ER/PR-positive breast cancer between black and white women was 4.39 (95% CI 1.76, 10.9, p = 0.001). CONCLUSIONS: We observed a racial disparity in breast cancer survival for patients diagnosed with ER/PR-positive tumors that did not appear to be due to differences in tumor stage, grade, or therapy initiation in black patients, suggesting that there may be racial differences in the molecular characteristics of hormone receptor-positive tumors, such that ER/PR-positive tumors in black patients may be less responsive to standard treatments.


Asunto(s)
Neoplasias de la Mama/mortalidad , Disparidades en el Estado de Salud , Negro o Afroamericano , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/etnología , Neoplasias de la Mama/metabolismo , Chicago/epidemiología , Femenino , Hispánicos o Latinos , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
17.
J Bioenerg Biomembr ; 49(4): 325-333, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28616679

RESUMEN

It is becoming increasingly clear that mitochondria drive cellular functions and in vivo phenotypes by directing the production rate and abundance of metabolites that are proposed to function as signaling molecules (Chandel 2015; Selak et al. 2005; Etchegaray and Mostoslavsky 2016). Many of these metabolites are intermediates that make up cellular metabolism, part of which occur in mitochondria (i.e. the TCA and urea cycles), while others are produced "on demand" mainly in response to alterations in the microenvironment in order to participate in the activation of acute adaptive responses (Mills et al. 2016; Go et al. 2010). Reactive oxygen species (ROS) are well suited for the purpose of executing rapid and transient signaling due to their short lived nature (Bae et al. 2011). Hydrogen peroxide (H2O2), in particular, possesses important characteristics including diffusibility and faster reactivity with specific residues such as methionine, cysteine and selenocysteine (Bonini et al. 2014). Therefore, it is reasonable to propose that H2O2 functions as a relatively specific redox signaling molecule. Even though it is now established that mtH2O2 is indispensable, at least for hypoxic adaptation and energetic and/or metabolic homeostasis (Hamanaka et al. 2016; Guzy et al. 2005), the question of how H2O2 is produced and regulated in the mitochondria is only partially answered. In this review, some roles of this indispensable signaling molecule in driving cellular metabolism will be discussed. In addition, we will discuss how H2O2 formation in mitochondria depends on and is controlled by MnSOD. Finally, we will conclude this manuscript by highlighting why a better understanding of redox hubs in the mitochondria will likely lead to new and improved therapeutics of a number of diseases, including cancer.


Asunto(s)
Mitocondrias/metabolismo , Transducción de Señal , Superóxido Dismutasa/fisiología , Animales , Humanos , Peróxido de Hidrógeno/metabolismo , Oxidación-Reducción
18.
Circ Res ; 114(3): 469-79, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24337049

RESUMEN

RATIONALE: Oxidants generated by activated endothelial cells are known to induce apoptosis, a pathogenic feature of vascular injury and inflammation from multiple pathogeneses. The melastatin-family transient receptor potential 2 (TRPM2) channel is an oxidant-sensitive Ca2+ permeable channel implicated in mediating apoptosis; however, the mechanisms of gating of the supranormal Ca2+ influx required for initiating of apoptosis are not understood. OBJECTIVE: Here, we addressed the role of TRPM2 and its interaction with the short splice variant TRPM2 short variant (TRPM2-S) in mediating the Ca2+ entry burst required for induction of endothelial cell apoptosis. METHODS AND RESULTS: We observed that TRPM2-S was basally associated with TRPM2 in the endothelial plasmalemma, and this interaction functioned to suppress TRPM2-dependent Ca2+ gating constitutively. Reactive oxygen species production in endothelial cells or directly applying reactive oxygen species induced protein kinase C-α activation and phosphorylation of TRPM2 at Ser 39. This in turn stimulated a large entry of Ca2+ and activated the apoptosis pathway. A similar TRPM2-dependent endothelial apoptosis mechanism was seen in intact vessels. The protein kinase C-α-activated phosphoswitch opened the TRPM2 channel to allow large Ca2+ influx by releasing TRPM2-S inhibition of TRPM2, which in turn activated caspase-3 and cleaved the caspase substrate poly(ADP-ribose) polymerase. CONCLUSIONS: Here, we describe a fundamental mechanism by which activation of the trp superfamily TRPM2 channel induces apoptosis of endothelial cells. The signaling mechanism involves reactive oxygen species-induced protein kinase C-α activation resulting in phosphorylation of TRPM2-S that allows enhanced TRPM2-mediated gating of Ca2+ and activation of the apoptosis program. Strategies aimed at preventing the uncoupling of TRPM2-S from TRPM2 and subsequent Ca2+ gating during oxidative stress may mitigate endothelial apoptosis and its consequences in mediating vascular injury and inflammation.


Asunto(s)
Apoptosis/genética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Isoformas de Proteínas/metabolismo , Empalme del ARN/genética , Canales Catiónicos TRPM/metabolismo , Animales , Señalización del Calcio/genética , Células Cultivadas , Variación Genética , Humanos , Ratones , Ratones Noqueados , Familia de Multigenes , Unión Proteica/genética , Isoformas de Proteínas/genética , Canales Catiónicos TRPM/genética
19.
Glia ; 63(3): 423-40, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25331559

RESUMEN

Redox-signaling is implicated in deleterious microglial activation underlying CNS disease, but how ROS program aberrant microglial function is unknown. Here, the oxidation of NF-κB p50 to a free radical intermediate is identified as a marker of dysfunctional M1 (pro-inflammatory) polarization in microglia. Microglia exposed to steady fluxes of H2 O2 showed altered NF-κB p50 protein-protein interactions, decreased NF-κB p50 DNA binding, and augmented late-stage TNFα expression, indicating that H2 O2 impairs NF-κB p50 function and prolongs amplified M1 activation. NF-κB p50(-/-) mice and cultures exhibited a disrupted M2 (alternative) response and impaired resolution of the M1 response. Persistent neuroinflammation continued 1 week after LPS (1 mg/kg, IP) administration in the NF-κB p50(-/-) mice. However, peripheral inflammation had already resolved in both strains of mice. Treatment with the spin-trap DMPO mildly reduced LPS-induced 22 h TNFα in the brain in NF-κB p50(+/+) mice. Interestingly, DMPO failed to reduce and strongly augmented brain TNFα production in NF-κB p50(-/-) mice, implicating a fundamental role for NF-κB p50 in the regulation of chronic neuroinflammation by free radicals. These data identify NF-κB p50 as a key redox-signaling mechanism regulating the M1/M2 balance in microglia, where loss of function leads to a CNS-specific vulnerability to chronic inflammation.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Microglía/fisiología , Subunidad p50 de NF-kappa B/metabolismo , Animales , Encéfalo/inmunología , Línea Celular , Células Cultivadas , ADN/metabolismo , Modelos Animales de Enfermedad , Inflamación/fisiopatología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Neuroinmunomodulación/fisiología , Oxidación-Reducción , Ratas , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L208-20, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25416384

RESUMEN

Pulmonary vascular remodeling, mainly attributable to enhanced pulmonary arterial smooth muscle cell proliferation and migration, is a major cause for elevated pulmonary vascular resistance and pulmonary arterial pressure in patients with pulmonary hypertension. The signaling cascade through Akt, comprised of three isoforms (Akt1-3) with distinct but overlapping functions, is involved in regulating cell proliferation and migration. This study aims to investigate whether the Akt/mammalian target of rapamycin (mTOR) pathway, and particularly which Akt isoform, contributes to the development and progression of pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension (HPH). Compared with the wild-type littermates, Akt1(-/-) mice were protected against the development and progression of chronic HPH, whereas Akt2(-/-) mice did not demonstrate any significant protection against the development of HPH. Furthermore, pulmonary vascular remodeling was significantly attenuated in the Akt1(-/-) mice, with no significant effect noted in the Akt2(-/-) mice after chronic exposure to normobaric hypoxia (10% O2). Overexpression of the upstream repressor of Akt signaling, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), and conditional and inducible knockout of mTOR in smooth muscle cells were also shown to attenuate the rise in right ventricular systolic pressure and the development of right ventricular hypertrophy. In conclusion, Akt isoforms appear to have a unique function within the pulmonary vasculature, with the Akt1 isoform having a dominant role in pulmonary vascular remodeling associated with HPH. The PTEN/Akt1/mTOR signaling pathway will continue to be a critical area of study in the pathogenesis of pulmonary hypertension, and specific Akt isoforms may help specify therapeutic targets for the treatment of pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar/genética , Hipoxia/patología , Proteínas Proto-Oncogénicas c-akt/genética , Remodelación Vascular , Animales , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Movimiento Celular , Proliferación Celular , Humanos , Hipertrofia Ventricular Derecha , Pulmón/irrigación sanguínea , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Fosfohidrolasa PTEN/biosíntesis , Fosforilación , Arteria Pulmonar/patología , Interferencia de ARN , ARN Interferente Pequeño , Serina-Treonina Quinasas TOR/genética , Tamoxifeno/farmacología , Resistencia Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA