42 results on '"Guzmán-Ruiz R"'
Search Results
2. Optimization of a MALDI-Imaging protocol for studying adipose tissue-associated disorders
- Author
-
Fernández-Vega, A., Chicano-Gálvez, E., Prentice, B.M., Anderson, D., Priego-Capote, F., López-Bascón, M.A., Calderón-Santiago, M., Avendaño, M.S., Guzmán-Ruiz, R., Tena-Sempere, M., Fernández, J.A., Caprioli, R.M., and Malagón, M.M.
- Published
- 2020
- Full Text
- View/download PDF
3. Influence of sample preparation on lipidomics analysis of polar lipids in adipose tissue
- Author
-
López-Bascón, M.A., Calderón-Santiago, M., Sánchez-Ceinos, J., Fernández-Vega, A., Guzmán-Ruiz, R., López-Miranda, J., Malagon, M.M., and Priego-Capote, F.
- Published
- 2018
- Full Text
- View/download PDF
4. Effects of the interaction of single-walled carbon nanotubes with 4-nonylphenol on their in vitro toxicity
- Author
-
Caballero-Díaz, E., Guzmán-Ruiz, R., Malagón, M.M., Simonet, B.M., and Valcárcel, M.
- Published
- 2014
- Full Text
- View/download PDF
5. Alarmin high-mobility group B1 (HMGB1) is regulated in human adipocytes in insulin resistance and influences insulin secretion in β-cells
- Author
-
Guzmán-Ruiz, R, Ortega, F, Rodríguez, A, Vázquez-Martínez, R, Díaz-Ruiz, A, Garcia-Navarro, S, Giralt, M, Garcia-Rios, A, Cobo-Padilla, D, Tinahones, F J, López-Miranda, J, Villarroya, F, Frühbeck, G, Fernández-Real, J M, and Malagón, M M
- Published
- 2014
- Full Text
- View/download PDF
6. Impaired mRNA splicing and proteostasis in preadipocytes in obesity related metabolic disease
- Author
-
Universitat Rovira i Virgili, Sánchez-Ceinos J; Guzmán-Ruiz R; Rangel-Zúñiga OA; López-Alcalá J; Moreno-Caño E; Del Río-Moreno M; Romero-Cabrera JL; Pérez-Martínez P; Maymo-Masip E; Vendrell J; Fernández-Veledo S; Fernández-Real JM; Laurencikiene J; Rydén M; Membrives A; Luque RM; López-Miranda J; Malagón MM, Universitat Rovira i Virgili, and Sánchez-Ceinos J; Guzmán-Ruiz R; Rangel-Zúñiga OA; López-Alcalá J; Moreno-Caño E; Del Río-Moreno M; Romero-Cabrera JL; Pérez-Martínez P; Maymo-Masip E; Vendrell J; Fernández-Veledo S; Fernández-Real JM; Laurencikiene J; Rydén M; Membrives A; Luque RM; López-Miranda J; Malagón MM
- Abstract
Preadipocytes are crucial for healthy adipose tissue expansion. Preadipocyte differentiation is altered in obese individuals, which has been proposed to contribute to obesity-associated metabolic disturbances. Here, we aimed at identifying the pathogenic processes underlying impaired adipocyte differentiation in obese individuals with insulin resistance (IR)/type 2 diabetes (T2D). We report that down-regulation of a key member of the major spliceosome, PRFP8/PRP8, as observed in IR/T2D preadipocytes from subcutaneous (SC) fat, prevented adipogenesis by altering both the expression and splicing patterns of adipogenic transcription factors and lipid droplet-related proteins, while adipocyte differentiation was restored upon recovery of PRFP8/PRP8 normal levels. Adipocyte differentiation was also compromised under conditions of endoplasmic reticulum (ER)-associated protein degradation (ERAD) hyperactivation, as occurs in SC and omental (OM) preadipocytes in IR/T2D obesity. Thus, targeting mRNA splicing and ER proteostasis in preadipocytes could improve adipose tissue function and thus contribute to metabolic health in obese individuals.
- Published
- 2021
7. Cardiac carnitine palmitoyltransferase is a target for leptin
- Author
-
Guzmán-Ruiz, R, Somoza, B, Fernández-Alfonso, M S, and Ruiz-Gayo, M
- Published
- 2008
8. Experiences in flipped learning
- Author
-
Burón, Isabel, Avendaño, S., Cantarero, Irene, Gahete Ortiz, Manuel D., Guzmán Ruiz, R., Vázquez, M.J., Luque, Rafael, Malagón, María M., Villalba, José Manuel, and Calzado Canale, Marco Antonio
- Subjects
Clase invertida ,Teaching innovation ,Flipped class ,Flipped learning ,Innovación docente - Abstract
El presente trabajo describe unas experiencias de innovación docente, de igual perfil y metodología, desarrolladas por un conjunto de profesores noveles participantes bajo la tutela experta de profesorado senior. El tipo de experiencia seleccionada ha sido el diseño y ejecución de una flipped classroom, o clase invertida. Además, se incluye la correspondiente evaluación de la acción con una valoración académica o impacto en los aprendizajes, y una valoración de la experiencia por parte del alumnado y profesorado responsable. Los resultados de cinco experiencias con impacto a mas de 270 alumnos de distinto perfil académico (tres asignaturas de titulo de Grado y dos de Máster) y las evaluaciones cuantificadas indican una valoracion muy positiva de esta metodología flipped learning (FL) por los alumnos y su deseo de aplicabilidad. Por su parte, el profesorado constata que las experiencias FL han incrementado satisfactoriamente el aprendizaje de sus alumnos y declaran una alta valoración de la experiencia para la mejora de sus competencia docentes. The present work describes experiences of teaching innovation, of the same profile and methodology, developed by a group of participating junior teachers under the expert guidance of senior teaching staff. The type of experience selected has been the design and execution of a flipped classroom, or inverted class. In addition, the corresponding evaluation of the action is included with an academic assessment or impact on learning, and an assessment of the experience by the students and teaching staff responsible. The results of five experiences with impact to more than 270 students of different academic profiles (three degrees and two Masters) and the quantified evaluations indicate a very positive assessment of this flipped learning methodology by the students and their desire for applicability. On the other hand, the teachers note that the FL experiences have successfully increased the learning of their students and declare a high evaluation of the experience for the improvement of their teaching competences.
- Published
- 2018
9. EXPERIENCIAS EN FLIPPED LEARNING
- Author
-
Burón, M.I., primary, Avendaño, S., additional, Cantarero, I., additional, Gahete, M.D., additional, Guzmán-Ruiz, R., additional, Vazquez, M.J., additional, Luque, R., additional, Malagón, M.M., additional, Villalba, J.M., additional, and Calzado, M.A., additional
- Published
- 2018
- Full Text
- View/download PDF
10. Defective glucose and lipid metabolism in rheumatoid arthritis is determined by chronic inflammation in metabolic tissues
- Author
-
Arias de la Rosa, I., primary, Escudero-Contreras, A., additional, Rodríguez-Cuenca, S., additional, Ruiz-Ponce, M., additional, Jiménez-Gómez, Y., additional, Ruiz-Limón, P., additional, Pérez-Sánchez, C., additional, Ábalos-Aguilera, M. C., additional, Cecchi, I., additional, Ortega, R., additional, Calvo, J., additional, Guzmán-Ruiz, R., additional, Malagón, M. M., additional, Collantes-Estevez, E., additional, Vidal-Puig, A., additional, López-Pedrera, Ch., additional, and Barbarroja, N., additional
- Published
- 2018
- Full Text
- View/download PDF
11. Proteasome Dysfunction Associated to Oxidative Stress and Proteotoxicity in Adipocytes Compromises Insulin Sensitivity in Human Obesity
- Author
-
Díaz-Ruiz, A., Guzmán-Ruiz, R., Moreno, N. R., García-Rios, A., Delgado-Casado, N., Membrives, A., Túnez, I., El Bekay, R., Fernández-Real, J. M., Tovar Carro, Sulay, Diéguez González, Carlos, Tinahones, F. J., Vázquez-Martínez, R., López-Miranda, J., and Malagón, M
- Subjects
Adult ,Male ,Obesity, Metabolically Benign ,Proteasome Endopeptidase Complex ,Palmitic Acid ,Subcutaneous Fat ,Endoplasmic Reticulum Stress ,Disease Models, Animal ,Mice ,Oxidative Stress ,Gene Expression Regulation ,3T3-L1 Cells ,Adipocytes ,Unfolded Protein Response ,Animals ,Humans ,Female ,Insulin Resistance ,Omentum - Abstract
AIMS: Obesity is characterized by a low-grade systemic inflammatory state and adipose tissue (AT) dysfunction, which predispose individuals to the development of insulin resistance (IR) and metabolic disease. However, a subset of obese individuals, referred to as metabolically healthy obese (MHO) individuals, are protected from obesity-associated metabolic abnormalities. Here, we aim at identifying molecular factors and pathways in adipocytes that are responsible for the progression from the insulin-sensitive to the insulin-resistant, metabolically unhealthy obese (MUHO) phenotype. RESULTS: Proteomic analysis of paired samples of adipocytes from subcutaneous (SC) and omental (OM) human AT revealed that both types of cells are altered in the MUHO state. Specifically, the glutathione redox cycle and other antioxidant defense systems as well as the protein-folding machinery were dysregulated and endoplasmic reticulum stress was increased in adipocytes from IR subjects. Moreover, proteasome activity was also compromised in adipocytes of MUHO individuals, which was associated with enhanced accumulation of oxidized and ubiquitinated proteins in these cells. Proteasome activity was also impaired in adipocytes of diet-induced obese mice and in 3T3-L1 adipocytes exposed to palmitate. In line with these data, proteasome inhibition significantly impaired insulin signaling in 3T3-L1 adipocytes. INNOVATION: This study provides the first evidence of the occurrence of protein homeostasis deregulation in adipocytes in human obesity, which, together with oxidative damage, interferes with insulin signaling in these cells. CONCLUSION: Our results suggest that proteasomal dysfunction and impaired proteostasis in adipocytes, resulting from protein oxidation and/or misfolding, constitute major pathogenic mechanisms in the development of IR in obesity. IMIBIC/Universidad de Córdoba-SCAI (ProteoRed, PRB2-ISCIII) MINECO/FEDER Junta de Andalucía/FEDER CIBERobn(Instituto de Salud Carlos III)
- Published
- 2015
12. O58 Un traitement chronique à l’apeline stimule l’oxydation des acides gras dans le muscle de souris normopondérales et insulino-résistantes
- Author
-
Attané, C., Guzman-Ruiz, R., Bézaire, V., Daviaud, D., Dray, C., Ruiz-Gayo, M., Valet, P., and Castan-Laurell, I.
- Published
- 2009
- Full Text
- View/download PDF
13. Identification of biomarkers of adipose tissue dysfunction in insulin resistance conditions
- Author
-
Navarro-Ruiz, Carmen, Malagón, María M., and Guzmán Ruiz, R.
- Subjects
Enfermedades metabólicas ,Tejido adiposo ,Biomarcadores ,Proteómica ,Resistencia a insulina ,Obesidad ,Marcadores proteómicos ,Acetiloma - Abstract
El constante incremento de la prevalencia de la obesidad a nivel mundial, que está alcanzando dimensiones epidémicas, constituye un grave problema de salud pública, debido al desarrollo de comorbilidades graves asociadas a esta patología, como la resistencia a insulina (IR), diabetes tipo 2 (T2D), enfermedad de hígado graso no alcohólico (NAFLD), dislipemias, enfermedades cardiovasculares, asma, hipertensión arterial, hipercolesterolemia, accidentes cerebrovasculares, artritis, enfermedades neurodegenerativas e incluso ciertos tipos de cáncer. Durante el desarrollo de la obesidad, ocurre un desequilibrio entre el gasto energético y la ingesta calórica producido por múltiples factores complejos y variables entre los que encuentran el sexo, la edad, el ambiente socio-cultural, una alimentación descontrolada –asociada, o no, a procesos emocionales–, y la actividad física. Independientemente del/los factor/es que provocan el desarrollo de obesidad, la disfunción del tejido adiposo subyace como causa biológica principal asociada a esta enfermedad. El tejido adiposo es un órgano de gran complejidad, siendo esencial para el mantenimiento de la homeostasis energética del organismo por su papel como responsable del almacenamiento del exceso de energía y su posterior movilización según demandas energéticas. Además, tras el descubrimiento de la leptina en 1994, el tejido adiposo pasó a ser reconocido como un órgano endocrino productor de hormonas, citoquinas, factores de crecimiento y factores vasoactivos, denominados en su conjunto como adipoquinas, que regulan desde la homeostasis energética, hasta la ingesta, la inflamación, la reproducción o el crecimiento celular. Si nos centramos en el tejido adiposo blanco (de aquí en adelante, denominado tejido adiposo), se distribuye fundamentalmente en dos localizaciones principales: depósito subcutáneo (SC) y depósito omental (OM), con diferencias celulares, fisiológicas, metabólicas y funcionales. Por ello, hay que destacar la importancia del estudio de las características de cada depósito del tejido adiposo ya que su distribución −independientemente de la cantidad de grasa corporal total− es un factor de riesgo para el desarrollo de las comorbilidades de la obesidad. Así, el incremento del depósito OM se asocia con un mayor riesgo cardiovascular, mientras que la acumulación de grasa en el depósito SC se relaciona con un perfil protector desde el punto de vista metabólico. No obstante, la remodelación y disfunción de cada depósito también puede contribuir a la fisiopatología de la enfermedad metabólica relacionada con la obesidad. En concreto, las alteraciones en la adipogénesis del depósito SC se han relacionado con la falta de expansibilidad de dicho tejido y, con ello, al desarrollo de las complicaciones metabólicas asociadas a la obesidad. En este contexto, el exceso de energía existente en condiciones de obesidad conlleva a una expansión patológica del tejido adiposo que conduce a otros procesos patogénicos en este tejido, entre los que se encuentran una desregulación del metabolismo glucídico y lipídico, aparición de estrés oxidativo y del retículo endoplasmático y un estado inflamatorio. Además, cuando se alcanza el límite de expansión del tejido adiposo, no es posible acumular más lípidos en este órgano, induciendo el depósito ectópico de grasa en otros tejidos involucrados en el mantenimiento de la homeostasis de la glucosa, un evento comúnmente definido como lipotoxicidad. La acumulación excesiva de lípidos en tejidos ectópicos conduce, en gran medida, al desarrollo de resistencia a insulina, siendo probablemente el contribuyente principal −junto a la inflamación− de la aparición de complicaciones metabólicas en los sujetos con obesidad. A pesar de las múltiples evidencias que se han encontrado sobre la relación entre la disfunción del tejido adiposo y la aparición de enfermedades metabólicas –como la obesidad, la resistencia a insulina y la diabetes tipo 2–, aún se desconocen muchos de los mecanismos subyacentes y rutas biológicas que se alteran en el tejido adiposo y promueven el desarrollo de estas condiciones patológicas. Los últimos avances en la identificación de marcadores de disfunción del tejido adiposo en obesidad y su asociación con enfermedades metabólicas han sido posibles gracias, en parte, a las técnicas ómicas, incluyendo estudios genómicos, transcriptómicos y proteómicos en modelos animales y humanos en condiciones de obesidad. En el grupo donde se ha desarrollado esta Tesis Doctoral, se han llevado a cabo varios estudios proteómicos de tejido adiposo y de sus principales componentes: fracción del estroma vascular, precursores y adipocitos que se alteran en obesidad con o sin resistencia a insulina (IR) o diabetes tipo 2 (T2D) o también, en repuesta a la pérdida de peso tras cirugía bariátrica. Sin embargo, a pesar de los avances realizados en la identificación de nuevos marcadores de la disfunción del tejido adiposo en obesidad, son necesarios nuevos estudios que estén centrados no solo en los posibles cambios cuantitativos que puedan sufrir las proteínas, sino también en su localización y, especialmente, en el estado funcional/disfuncional de dichas proteínas. En este sentido, el estudio de las modificaciones postraduccionales de las proteínas puede proporcionar vías adicionales de identificación de biomarcadores potenciales que puedan ser útiles para el establecimiento de nuevas alternativas biomédicas. Actualmente, se acepta que la diferencia en complejidad que existe entre el genoma y el proteoma se debe, en gran parte, a las modificaciones postraduccionales (MPTs) que sufren las proteínas, ya que de los 25.000 genes aproximadamente que tiene el ser humano, se codifica un número final estimado de 1 millón de proteínas. La gran mayoría de proteínas están reguladas por MPTs (fosforilación, acetilación, carbonilación…) de las que poco se sabe en el tejido adiposo en obesidad. Por ello, esta Tesis Doctoral ha tenido como objetivo explorar, identificar y caracterizar nuevos posibles marcadores del tejido adiposo regulados funcionalmente por modificaciones postraduccionales, una reversible –acetilación− y otra, irreversible – carbonilación−, e implicados en el desarrollo y progresión de enfermedad metabólica (IR y T2D) en condiciones de obesidad. En concreto, la acetilación es una de las MPTs que más se ha relacionado con la obesidad, diabetes y cáncer, así como con enfermedades cardiovasculares y neurodegenerativas. En esta Tesis Doctoral se describe, por primera vez, el perfil de proteínas acetiladas (acetiloma) del tejido adiposo humano de los dos principales depósitos (subcutáneo y omental) bajo condiciones fisiológicas (individuos delgados) y en condiciones patológicas (obesidad y resistencia a insulina). Estos estudios muestran que el 25% del proteoma total del tejido adiposo humano está regulado por acetilación, demostrando la gran importancia que tiene esta MPT en el funcionamiento de este órgano. Al analizar el perfil de acetilación de proteínas específicas en los distintos grupos de sujetos estudiados, se observó que la obesidad y la resistencia a insulina afectan de manera depósito-dependiente a la acetilación de proteínas en el tejido adiposo. La reducción total del estado de acetilación, así como la alteración de los niveles de acetilación en enzimas clave del metabolismo energético observadas entre individuos delgados y obesos con normoglucemia (Ob-NG) o resistencia a insulina (Ob-IR), podrían contribuir a la disfunción de los adipocitos y al desarrollo de enfermedades metabólicas asociadas a la obesidad. Como se ha mencionado, el análisis del acetiloma mostró la existencia de diferentes patrones de acetilación entre los dos depósitos principales de tejido adiposo, SC y OM, destacando la complejidad molecular de este tejido. En concreto, este análisis mostró cambios en la acetilación de la chaperona lipídica y adipoquina, FABP4, en el depósito SC en relación con la resistencia a la insulina, revelando así un posible biomarcador de expansión disfuncional específica de depósito. Estudios previos han demostrado que FABP4 podría ser un marcador de resistencia a insulina debido a sus altos niveles circulantes en individuos obesos con resistencia a insulina. FABP4 interviene en la captación, almacenamiento y movilización de ácidos grasos, además de participar en la regulación transcripcional mediada por lípidos en el núcleo y ejercer una acción señalizadora como adipoquina. La alteración de FABP4 en obesidad podría relacionarse con la acumulación alterada de lípidos que ocurre en el tejido adiposo en estas condiciones, debido a su papel en la modulación del metabolismo lipídico en los adipocitos. Sin embargo, se desconoce si esta función podría estar modulada por acetilación, lo que podría ayudar a definir realmente a FABP4 como biomarcador de estas patologías. Nuestros resultados indicaron que los cambios en el perfil de acetilación de FABP4 regulan su ubicación celular y alteran las funciones intracelulares de FABP4 (control de adipogénesis y de las rutas lipolítica/lipogénica), indicando la existencia de una regulación cualitativa, más allá de la cantidad de proteína disponible, que impacta en su función. Por tanto, no solo la proteína en sí, sino también su estado de acetilación, podrían constituir dianas terapéuticas en obesidad y resistencia a insulina. or otra parte, en la patogénesis de la obesidad, resistencia a insulina y diabetes tipo 2, juega un papel fundamental el estrés oxidativo, el cual se produce como consecuencia de un acúmulo citotóxico de especies reactivas de oxígeno (ROS). El exceso de ROS provoca daño celular como consecuencia de la inducción de procesos como la peroxidación de lípidos o la carbonilación de proteínas. Así, en esta Tesis Doctoral hemos explorado este último proceso, describiendo, por primera vez, el perfil diferencial de proteínas carboniladas (carboniloma) del tejido adiposo humano de los dos depósitos de grasa, subcutáneo y omental, en sujetos con obesidad y diferente grado de sensibilidad a insulina –sujetos con obesidad y normoglucemia (Ob-NG), con obesidad y resistencia a insulina (Ob-IR) o con obesidad y diabetes tipo 2 (Ob-T2D)–. Nuestros resultados proteómicos evidenciaron diferencias específicas de depósito en el carboniloma del tejido adiposo, proporcionando nuevos conocimientos sobre la participación del tejido adiposo omental en el desarrollo de resistencia a insulina asociada a la obesidad. En concreto, nuestros resultados proteómicos mostraron un patrón de carbonilación diferencial de la transferrina entre los sujetos Ob-NG vs. Ob-IR y Ob-T2D en el depósito SC y OM, encontrando un aumento significativo de la transferrina carbonilada en pacientes Ob-IR vs. Ob-NG en el depósito OM. Adicionalmente, debido a la relación entre el metabolismo glucídico y del hierro analizamos la homeostasis del hierro en el tejido adiposo de nuestros sujetos de estudio, no encontrando diferencias significativas en el contenido de hierro total entre los grupos. Sin embargo, se encontraron cambios en la distribución del receptor de transferrina en adipocitos condiciones de exposición al ambiente existente en diabetes (i.e., exposición a medios condicionados procedentes de pacientes que desarrollan T2D) que podrían asociarse a una alteración en su unión con la transferrina. También identificamos los sitios potenciales de interacción entre transferrina y su receptor susceptibles de modificar dicha interacción. Así, nuestros estudios mostraron que la internalización del receptor de transferrina se altera en respuesta a un ambiente inductor de T2D, lo que sugiere que en el desarrollo de T2D podría contribuir, al menos en parte, la carbonilación de un residuo específico de la transferrina, P74, modificando sus propiedades electrostáticas y, por tanto, comprometiendo su unión con su receptor. En conjunto, estos resultados muestran la posible relación entre la carbonilación de proteínas y la desregulación de la homeostasis del hierro que tiene lugar en condiciones de obesidad y enfermedad metabólica. Por último, los estudios proteómicos en general, y de MPTs en particular, tienen limitaciones en cuanto a la falta de herramientas (por ejemplo, anticuerpos específicos de residuos, o inducción de MPTs en residuos específicos de proteínas concretas) que permitan profundizar para el diseño de estrategias de prevención o terapéuticas. En este contexto, la nanomedicina aparece como una línea de trabajo futuro para el diseño de estas estrategias, una línea que aún no ha sido muy explorada en relación al tejido adiposo en particular. Existe, por tanto, la necesidad de buscar nuevas estrategias metodológicas en este ámbito, tales como el uso de los quantum dots. En concreto, ya hay quantum dots conjugadas con proteínas comerciales (Qdot™ Streptavidin Sampler Kit, por ejemplo) para aplicaciones como Western blot, citometría de flujo, adquisición de imágenes por microscopía confocal e incluso en flujos de trabajo de purificación para lograr el fraccionamiento deseado. En esta Tesis Doctoral se llevó a cabo, por primera vez, un estudio comparativo in vitro totalmente paralelo del comportamiento de dos tipos de quantum dots, CQDs (carbon-based quantum dots) y SQDs (semiconductor quantum dots) en preadipocitos, como precursor del tipo celular maduro del tejido adiposo y por tanto, elemento fundamental en la expansión de dicho tejido. Nuestros resultados mostraron que las CQDs podrían ser una buena herramienta para la caracterización y posible aplicación biomédica de proteínas de interés en condiciones de obesidad y resistencia a insulina, como FABP4 o transferrina, las cuales son reguladas funcionalmente por modificaciones postraduccionales. Más aún, independientemente del tipo de nanopartícula, hay que tener en cuenta la influencia de los distintos componentes del microentorno celular (i.e, medio de cultivo), ya que pueden alterar las características sintéticas de las mismas, otorgándoles nuevas propiedades biológicas que repercuten en su comportamiento a nivel celular. Estos resultados, por tanto, abren una nueva línea de futuro para el desarrollo no solo de estudios de caracterización básicos, sino para la puesta a punto de nuevas herramientas con aplicación potencial en el campo de la obesidad.
- Published
- 2022
14. Mediators of the cellular response to the development of obesity and metabolic disease
- Author
-
Sánchez Ceinos, Julia, Malagón, María M., and Guzmán Ruiz, R.
- Subjects
Preadipocytes ,Adipogenesis ,Molecular biology ,miRNAs ,Adipose tissue ,Metabolic diseases ,Insulin resistance ,Type 2 diabetes ,Obesity ,Biomarkers - Abstract
Obesity is the pandemic of the 21st century. Overall, about 15% of the world’s adult population is obese, and if the rising trend continues, it is estimated that global prevalence reaches 20% by 2025. Obesity poses a major public health issue due to its elevated risks for adverse health consequences, including several serious chronic diseases, such as insulin resistance (IR), type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), cardiovascular disease, dyslipidemia, hypertension, stroke, hypercholesterolemia, hypertriglyceridemia, arthritis, asthma, neurodegenerative diseases, and even certain forms of cancer. Many factors and mechanisms have been implicated in obesity pathogenesis, but their trigger is uncertain, and the causal relationship between them and the complications of obesity remains in question. However, there is little doubt that obesity is closely associated with adipose tissue dysfunction, and that this condition leads to the development of metabolic diseases. Adipose tissue is a complex organ with primary roles in energy homeostasis control. Thus, adipose tissue not only acts as a reservoir for energy storage and utilization, but also it senses energy demands and secretes signalling factors to regulate other metabolic tissues. However, in obesity, adipose tissue may become severely dysfunctional and not expand properly to store the energy excess. This induces ectopic fat deposition in other tissues involved in the maintenance of glucose homeostasis, an event commonly defined as “lipotoxicity”. It has been extensively demonstrated that excessive lipid accumulation in ectopic tissues leads to local inflammation and IR. Numerous pathogenic processes have been associated with the unhealthy expansion of the adipose tissue, including inflammation, fibrosis, hypoxia, altered adipokines secretion, mitochondrial dysfunction, hyperinsulinemia, endoplasmic reticulum (ER), and oxidative stress. Dietary and lifestyle, as well as therapeutic interventions, can be adequate to treat obesity and prevent metabolic alterations. Nevertheless, the exact molecular and cellular mechanisms underlying adipose tissue dysfunction and its implications in the development of metabolic disturbances are broadly unknown. Improving our understanding in this field might lead to the development of new approaches and the identification of therapeutic targets for treating obesity. In this Doctoral Thesis, we aimed at identifying potential factors and pathway markers relevant to the loss of adipose tissue function in obesity and their relationship to obesityrelated IR/T2D. To this end, we analysed human adipose tissue samples in combination with both human and murine cell lines, using state-of-the-art methodologies to investigate adipose tissue functionality in obesity and in response to weight loss upon bariatric surgery (BS), and their corresponding mechanisms associated with metabolic alterations or recovery, respectively. Specifically, to reach this general goal, we have carried out two separate studies, which are depicted in detail below. 1. Study 1: Identification of pathogenic markers of metabolic disease in preadipocytes of obese individuals Adipose precursor cells, the preadipocytes, are essential for the maintenance of adipose tissue homeostasis, regeneration, and expansion. Preadipocytes differentiation into adipocytes (i.e., adipogenesis) enables adipocyte turnover and adipose tissue growth, and ensures adipose tissue plasticity to accommodate surplus energy. It has been proposed that the inability for recruiting new adipose cells, together with the functional impairment of hypertrophied adipocytes that occur in obesity, contributes to lipid spillover from the adipose tissue. Thus, increasing adipogenesis appears as a valuable strategy to facilitate healthy adipose tissue expansion and ensure metabolic health. Adipocyte differentiation relies on major changes in gene expression programs regulating mRNA and protein production. An increasing body of evidence shows that mRNA processing and, in particular, alternative splicing, is crucial for genome reprogramming during cell differentiation. However, the splicing components relevant to adipogenesis and the cellular events regulated by alternative splicing during adipocyte differentiation have been scarcely explored, and it is yet to be established whether alternative splicing is modified in human obesity. Another crucial mechanism preserving precursor cell function relates to protein homeostasis (i.e., proteostasis), which maintains the capacity of cells to expand in order to sustain tissue growth and regeneration. Several lines of evidence support an important role for the endoplasmic reticulum (ER) protein quality control system in the regulation of adipogenesis. In fact, the unfolded protein response (UPR) is perturbed in the obese adipose tissue, and it has been proposed to contribute to the pathology of obesity. By contrast, it is still unknown whether the other component of the protein control system, the ER-associated protein degradation (ERAD), which is crucial for protecting cells against the accumulation of misfolded/unfolded proteins and proteotoxicity, is altered in the obese adipose tissue. Here, in order to identify altered molecular pathways that may contribute to metabolic disease in obesity, we set out an iTRAQ-LC-MS/MS proteomic approach for the analysis of subcutaneous (SC) and omental (OM) preadipocytes from obese individuals with normoglycaemia (NG) and T2D. Down-regulation of multiple components of the splicing machinery was observed in SC preadipocytes from obese individuals with insulin resistance (IR) or T2D, as compared to NG obesity. This, together with the observation that adipogenesis can be modulated by regulating the expression levels of a key spliceosome component, PRFP8/PRP8, supports a role for alternative splicing in the development of obesity-associated metabolic complications. In addition, our studies show that not only the UPR is altered in human SC and OM preadipocytes from IR/T2D obese subjects, but also that the ERAD system is hyperactivated. This condition, when mimicked in vitro, prevented adipogenesis. Our results provide novel mechanistic explanations for the impaired adipogenic capacity observed in IR/T2D obesity that relates to both mRNA and ER-proteostasis disturbances. 2. Study 2: Characterization of miRNAs as markers of dysfunctional adipose tissue in type 2 diabetes (T2D) It is increasingly accepted that, besides adipokines, the adipose tissue is a major physiological source of circulating microRNAs (miRNAs). miRNAs, a class of small noncoding RNAs that post-transcriptionally regulate gene expression, have emerged as key molecules for cell function. Anomalous miRNA levels and alterations in their biogénesis machinery have been related to several metabolic diseases, including obesity as well as T2D, and dyslipidemia. miRNAs are also actively secreted into the circulation and have been proposed to act as messengers for intercellular communication. This characteristic has pointed out circulating miRNAs as potential biomarkers for disease and altered circulating levels of numerous miRNAs have been associated with metabolic disorders. In this scenario, in order to design targeted preventive therapeutic strategies, it is critical to identify the early mechanisms that precede disease onset. Recently, the predictive value of a number of miRNAs for the diagnosis of T2D incidence was assessed in the CORonary Diet Intervention with Olive oil and cardiovascular PREVention (CORDIOPREV) study (ClinicalTrial.gov ID: NTC00924937), a prospective study carried out in 1,002 patients with coronary heart disease and high cardiovascular risk. These studies demonstrated that, when combined with HbA1c, a group of nine miRNAs (miR-9, miR-28-3p, miR-29a, miR-30a-5p, miR- 103, miR-126, miR-150, miR-223-3p, and miR-375), provided a higher predictive value in T2D diagnosis than clinical parameters. The relationship between baseline levels of these miRNAs with markers of beta-cell function and systemic and peripheral IR was also investigated. However, their potential association with adipose tissue deregulation was not analyzed. This is of interest as adipose tissue dysfunction has been proposed as a major contributing factor for the development of T2D, and adipose tissue miRNA expression profile is altered in obesity and T2D. In this line, several cross-sectional human studies have shown that the obesity-related expression pattern of specific circulating miRNAs reflects their miRNA adipose tissue expression profiles, supporting a role for circulating miRNAs as adipose tissue biomarkers. However, the relationship between circulating miRNAs and adipose tissue functional state is not fully understood, yet it might be useful to identify adipose tissue-related metabolic complications. In this scenario, we aimed at analyzing the relationship between previously established predictive miRNAs for T2D onset in the CORDIOPREV cohort (Jiménez- Lucena et al., 2018) and the loss of adipose tissue insulin sensitivity. Once established that circulating miR-223-3p was dysregulated in relation to adipose tissue function, we performed functional analysis to elucidate both the potential of preadipocytes and adipocytes as miR-223-3p secreting cells, and the consequences of miR-223-3p dysregulation on adipocyte biology. Our results indicate that miR-223-3p secretion by preadipocytes and adipocytes is prevented under inflammatory conditions, and that its pathogenic accumulation leads to alterations in both glucose and lipid metabolism in these cells. These observations could answer to the lower circulating levels on this miRNA found in those patients who are going to develop T2D, as well as explain its underlying role as a potential predictor of adipose dysfunction related to T2D development. We have also demonstrated that the changes that occur in the circulating milieu after BS-induced weight-loss modulate miR-223-3p expression by adipocytes. The relevance of these observations was further supported by our functional studies in adipocytes exposed to pre- and post-BS serum, where the post-BS serum improved the insulininduced glucose uptake in adipocytes, in comparison with those cells exposed to pre- BS. These findings further support the notion that inflammatory mediators present in either the serum, or locally in the adipose tissue, may be responsible for the alterations in miR-223-3p regulation. In sum, when viewed together, our studies indicate that impaired splicing and disturbed ER-proteostasis are components of the pathogenic molecular fingerprint of preadipocytes that could be targeted to prevent and/or improve adipose tissue dysfunction in obesity and its related metabolic disorders. Likewise, our data support the notion that miR-223-3p may have a role as a potential predictor of adipose dysfunction related to T2D development, thus unveiling a novel molecular target that may be helpful to design novel therapeutic strategies to prevent T2D. La obesidad es la pandemia del siglo XXI. En conjunto, alrededor del 15% de la población adulta mundial es obesa. Si la tendencia al alza continúa, se estima que la prevalencia mundial alcanzará el 20% para el 2025. Así, la obesidad plantea un problema importante de salud pública debido a sus elevados riesgos para la salud, entre ellas varias enfermedades crónicas graves, como resistencia a insulina (IR), diabetes tipo 2 (T2D), enfermedad de hígado graso no alcohólico (NAFLD), enfermedad cardiovascular, dislipidemia, hipertensión, accidente cerebrovascular, hipercolesterolemia, hipertrigliceridemia, artritis, asma, enfermedades neurodegenerativas e incluso ciertas formas de cáncer. Diversos factores y mecanismos han sido implicados en la patogénesis de la obesidad, pero su desencadenante es incierto. Además, la relación causal entre ellos y las complicaciones de la obesidad permanece sin resolver. Sin embargo, hay pocas dudas de que la obesidad está estrechamente asociada con la disfunción del tejido adiposo, y que esta condición conduce al desarrollo de enfermedades metabólicas. El tejido adiposo es un órgano complejo con funciones principales en el control de la homeostasis energética. Por lo tanto, el tejido adiposo no solo actúa como un depósito para el almacenamiento y la utilización de energía, sino que también detecta las demandas de energía, y secreta factores de señalización para regular otros tejidos metabólicos. Sin embargo, en obesidad, el tejido adiposo puede volverse gravemente disfuncional, y no expandirse adecuadamente para almacenar el exceso de energía. Esto induce el depósito ectópico de grasa en otros tejidos involucrados en el mantenimiento de la homeostasis de la glucosa, un evento comúnmente definido como "lipotoxicidad". Se ha demostrado ampliamente que la acumulación excesiva de lípidos en tejidos ectópicos provoca inflamación local e IR. Numerosos procesos patogénicos se han asociado con la expansión no saludable del tejido adiposo, que incluyen: inflamación, fibrosis, hipoxia, secreción alterada de adipoquinas, disfunción mitocondrial, hiperinsulinemia, y estrés de retículo endoplásmico (ER) y oxidativo. Las intervenciones dietéticas y de estilo de vida, así como las terapéuticas, pueden ser adecuadas para tratar la obesidad y prevenir alteraciones metabólicas. Sin embargo, los mecanismos moleculares y celulares exactos que subyacen a la disfunción del tejido adiposo, y sus implicaciones en el desarrollo de alteraciones metabólicas son ampliamente desconocidos. Mejorar nuestra comprensión en este campo podría conducir al desarrollo de nuevos enfoques y a la identificación de dianas terapéuticas para tratar la obesidad. En esta Tesis Doctoral, nuestro objetivo fue identificar los posibles factores y marcadores de las vías relevantes para la pérdida de la función del tejido adiposo en obesidad, y su relación con IR/T2D asociadas. Para ello, analizamos muestras de tejido adiposo humano en combinación con líneas celulares, tanto humanas como murinas, utilizando metodologías de vanguardia para investigar la funcionalidad del tejido adiposo en obesidad, y su respuesta a la pérdida de peso tras cirugía bariátrica (BS). Así como, sus correspondientes mecanismos asociados con alteraciones metabólicas o de recuperación, respectivamente. Específicamente, para alcanzar este objetivo general, hemos llevado a cabo dos estudios separados, que se describen en detalle a continuación. 1. Estudio 1: Identificación de marcadores patogénicos de enfermedad metabólica en preadipocitos de individuos obesos Las células adiposas precursoras, los preadipocitos, son esenciales para el mantenimiento de la homeostasis, regeneración y expansión del tejido adiposo. La diferenciación de los preadipocitos en adipocitos (adipogénesis) permite el recambio de adipocitos y el crecimiento del tejido adiposo, y asegura la plasticidad de este tejido para acomodar el exceso de energía. Se ha propuesto que la incapacidad para reclutar nuevas células adiposas, junto con el deterioro funcional de los adipocitos hipertróficos que ocurre en obesidad, contribuye al derrame de lípidos del tejido adiposo. Por lo tanto, el aumento de la adipogénesis aparece como una estrategia valiosa para facilitar la expansión saludable del tejido adiposo y salvaguardar la salud metabólica. La diferenciación de adipocitos se basa en cambios importantes en los programas de expresión génica que regulan la producción de ARNm y proteínas. Un creciente número de evidencias muestra que el procesamiento del ARNm y, en particular, el empalme alternativo, es crucial para la reprogramación del genoma durante la diferenciación celular. Sin embargo, los componentes de empalme relevantes para la adipogénesis y los eventos celulares regulados por este empalme alternativo durante la diferenciación de adipocitos apenas se han explorado. Así mismo, aún no se ha establecido si el empalme alternativo se modifica en la obesidad humana. Otro mecanismo crucial que preserva la función de las células precursoras se relaciona con la homeostasis de las proteínas (proteostasis), que mantiene la capacidad de las células de expandirse para mantener el crecimiento y la regeneración de los tejidos. Varias líneas de evidencia apoyan un papel importante para el sistema de control de calidad proteico del retículo endoplásmico (ER) en la regulación de la adipogénesis. De hecho, la respuesta a proteínas mal plegadas (UPR) se perturba en el tejido adiposo obeso, y se ha propuesto que contribuye a la patología de la obesidad. Por el contrario, todavía se desconoce si otro componente del sistema de control de proteínas, la degradación de proteínas asociada a ER (ERAD), que es crucial para proteger las células contra la acumulación de proteínas mal plegadas/desplegadas y la proteotoxicidad, está alterado en el tejido adiposo obeso. Aquí, con el fin de identificar vías moleculares alteradas que pueden contribuir a la enfermedad metabólica en obesidad, realizamos un estudio proteómico de iTRAQ-LCMS/ MS para el análisis de preadipocitos subcutáneos (SC) y omentales (OM) de individuos obesos con normoglucemia (NG) y T2D. Se observó una disminución de múltiples componentes de la maquinaria de empalme en preadipocitos SC de individuos obesos con IR o T2D, en comparación con los obesos NG. Esto, junto con la observación de que la adipogénesis puede modularse regulando los niveles de expresión de un componente clave del espliceosoma, PRPF8/PRP8, respalda el papel de este proceso en el desarrollo de complicaciones metabólicas asociadas a la obesidad. Además, nuestros estudios muestran que no solo el sistema UPR está alterado en preadipocitos humanos SC y OM de sujetos obesos con IR/T2D, sino que también existe una hiperactivación del sistema ERAD. Esta condición, cuando se imita in vitro, previene la adipogénesis. Nuestros resultados proporcionan novedosas explicaciones mecanicistas para la capacidad adipogénica deteriorada observada en los obesos IR/T2D que se relaciona tanto con la alteración del procesamiento de ARNm como con la de la proteostasis del ER. 2. Estudio 2: Caracterización de miRNAs como marcadores de tejido adiposo disfuncional en la diabetes tipo 2 (T2D) Cada vez se acepta más que, además de las adipoquinas, el tejido adiposo es una importante fuente fisiológica de microRNAs (miRNAs) circulantes. Los miRNAs, una clase de pequeños ARN no codificantes que regulan la expresión génica posttranscripcionalmente, han surgido como moléculas clave para la función celular. Los niveles de miRNAs anómalos y las alteraciones en su maquinaria de biogénesis se han relacionado con varias enfermedades metabólicas, incluida la obesidad, así como la T2D y la dislipidemia. Los miRNAs también se secretan activamente a la circulación y se ha propuesto que actúen como mensajeros de la comunicación intercelular. Esta característica ha señalado a los miRNAs circulantes como posibles biomarcadores de enfermedades, y los niveles circulantes alterados de numerosos miRNAs se han asociado con trastornos metabólicos. En este escenario, para diseñar estrategias terapéuticas preventivas específicas, es fundamental identificar los mecanismos tempranos que preceden al inicio de la enfermedad. Recientemente, en el estudio CORonary Diet Intervention with Olive oil and cardiovascular PREVention (CORDIOPREV, ClinicalTrial.gov ID: NTC00924937), se evaluó el valor predictivo de varios miRNAs para el diagnóstico de incidencia de T2D, un estudio prospectivo realizado en 1,002 pacientes con enfermedad coronaria y alto riesgo cardiovascular. Estos estudios demostraron que, cuando se combina con HbA1c, un grupo de nueve miRNAs (miR-9, miR-28-3p, miR-29a, miR-30a-5p, miR-103, miR- 126, miR-150, miR-223-3p, y miR-375), proporcionaron un valor predictivo más alto en el diagnóstico de T2D que los parámetros clínicos. También se investigó la relación entre los niveles basales de estos miRNAs con marcadores de la función de las células beta y la IR sistémica y periférica. Sin embargo, su posible asociación con la desregulación del tejido adiposo no se analizó. Esto es de interés ya que se ha propuesto que la disfunción del tejido adiposo es un factor importante que contribuye al desarrollo de T2D, y el perfil de expresión de miRNAs del tejido adiposo está alterado en obesidad y T2D. En esta línea, varios estudios transversales en humanos han demostrado que el patrón de expresión de específicos miRNAs circulantes, relacionados con obesidad, refleja los perfiles de expresión de estos miRNAs en el tejido adiposo, lo que respalda el papel de los miRNAs circulantes como biomarcadores del tejido adiposo. Sin embargo, la relación entre los miRNAs circulantes y el estado funcional del tejido adiposo no se comprende completamente, lo cual podría ser útil para identificar las complicaciones metabólicas relacionadas con el tejido adiposo. En este escenario, nuestro objetivo fue analizar la relación entre los miRNAs predictivos para el inicio de T2D, previamente establecidos en la cohorte CORDIOPREV (Jiménez-Lucena et al., 2018), y la pérdida de sensibilidad a insulina del tejido adiposo. Una vez establecido que el miR-223-3p circulante estaba desregulado con relación a la función del tejido adiposo, realizamos un análisis funcional para dilucidar tanto el potencial de los preadipocitos y los adipocitos como células secretoras de miR-223-3p, como las consecuencias de la desregulación de este miRNA sobre la biología de los adipocitos. Así, nuestros resultados indican que la secreción de miR-223-3p por preadipocitos y adipocitos es inhibida en condiciones inflamatorias, y que su acumulación patológica provoca alteraciones tanto en el metabolismo glucídico como lipídico de estas células. Estas observaciones podrían dar respuesta a los bajos niveles de este miRNA encontrados en aquellos pacientes que van a desarrollar T2D, así como explicar su potencial predictor de la disfunción adiposa relacionada con el desarrollo de esta enfermedad. También hemos demostrado que los cambios que ocurren en el medio circulante después de la pérdida de peso inducida por BS modulan la expresión de miR-223-3p en adipocitos. La relevancia de estas observaciones fue apoyada además por nuestros estudios funcionales en adipocitos expuestos a suero pre- y post-BS, donde el suero post-BS mejoró la captación de glucosa estimulada por insulina en adipocitos, en comparación con aquellos expuestos a sueros pre-BS. Estos hallazgos refuerzan aún más la idea de que los mediadores inflamatorios presentes en el suero, o localmente en el tejido adiposo, pueden ser responsables de las alteraciones en la regulación de miR- 223-3p. En resumen, cuando se ven en conjunto, nuestros estudios indican que el empalme deficiente y la alteración de la proteostasis del ER son componentes de la huella molecular patogénica de los preadipocitos que podrían ayudar a prevenir y/o mejorar la disfunción del tejido adiposo en obesidad y sus trastornos metabólicos relacionados. Del mismo modo, nuestros datos respaldan la noción de que miR-223-3p puede tener un papel como potencial predictor de la disfunción adiposa relacionada con el desarrollo de T2D, revelando así una nueva diana molecular que puede ser útil para diseñar nuevas estrategias terapéuticas para prevenir la T2D.
- Published
- 2021
15. Characterization of novel pathways in the regulation of lipid metabolism in adipocytes. Alterations in obesity
- Author
-
Fernández Vega, Alejandro, Malagón, María M., and Guzmán Ruiz, R.
- Subjects
Lipidoma ,Tejido adiposo ,Metabolismo lipídico ,Resistencia a insulina ,Adipocitos ,Obesidad ,Rutas metabólicas ,Biosíntesis - Published
- 2020
16. Caracterización de nuevas rutas reguladoras del metabolismo lipídico en adipocitos. Alteraciones en obesidad
- Author
-
Fernández Vega, Alejandro, Malagón, María M., and Guzmán Ruiz, R.
- Subjects
Lipidoma ,Tejido adiposo ,Metabolismo lipídico ,Resistencia a insulina ,Adipocitos ,Obesidad ,Rutas metabólicas ,Biosíntesis - Published
- 2020
17. Interrelation of adipose tissue macrophages and fibrosis in obesity.
- Author
-
Dahdah N, Tercero-Alcázar C, Malagón MM, Garcia-Roves PM, and Guzmán-Ruiz R
- Subjects
- Humans, Animals, Obesity metabolism, Obesity pathology, Obesity immunology, Macrophages metabolism, Macrophages immunology, Macrophages pathology, Macrophages physiology, Adipose Tissue metabolism, Adipose Tissue pathology, Adipose Tissue immunology, Fibrosis
- Abstract
Obesity is characterized by adipose tissue expansion, extracellular matrix remodelling and unresolved inflammation that contribute to insulin resistance and fibrosis. Adipose tissue macrophages represent the most abundant class of immune cells in adipose tissue inflammation and could be key mediators of adipocyte dysfunction and fibrosis in obesity. Although macrophage activation states are classically defined by the M1/M2 polarization nomenclature, novel studies have revealed a more complex range of macrophage phenotypes in response to external condition or the surrounding microenvironment. Here, we discuss the plasticity of adipose tissue macrophages (ATMs) in response to their microenvironment in obesity, with special focus on macrophage infiltration and polarization, and their contribution to adipose tissue fibrosis. A better understanding of the role of ATMs as regulators of adipose tissue remodelling may provide novel therapeutic strategies against obesity and associated metabolic diseases., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Localization, traffic and function of Rab34 in adipocyte lipid and endocrine functions.
- Author
-
López-Alcalá J, Gordon A, Trávez A, Tercero-Alcázar C, Correa-Sáez A, González-Rellán MJ, Rangel-Zúñiga OA, Rodríguez A, Membrives A, Frühbeck G, Nogueiras R, Calzado MA, Guzmán-Ruiz R, and Malagón MM
- Subjects
- Humans, Adipocytes, Adipokines, GTP Phosphohydrolases, Obesity, Lipids, Fatty Acid-Binding Proteins, Adiponectin, Proteomics
- Abstract
Background: Excessive lipid accumulation in the adipose tissue in obesity alters the endocrine and energy storage functions of adipocytes. Adipocyte lipid droplets represent key organelles coordinating lipid storage and mobilization in these cells. Recently, we identified the small GTPase, Rab34, in the lipid droplet proteome of adipocytes. Herein, we have characterized the distribution, intracellular transport, and potential contribution of this GTPase to adipocyte physiology and its regulation in obesity., Methods: 3T3-L1 and human primary preadipocytes were differentiated in vitro and Rab34 distribution and trafficking were analyzed using markers of cellular compartments. 3T3-L1 adipocytes were transfected with expression vectors and/or Rab34 siRNA and assessed for secretory activity, lipid accumulation and expression of proteins regulating lipid metabolism. Proteomic and protein interaction analyses were employed for the identification of the Rab34 interactome. These studies were combined with functional analysis to unveil the role played by the GTPase in adipocytes, with a focus on the actions conveyed by Rab34 interacting proteins. Finally, Rab34 regulation in response to obesity was also evaluated., Results: Our results show that Rab34 localizes at the Golgi apparatus in preadipocytes. During lipid droplet biogenesis, Rab34 translocates from the Golgi to endoplasmic reticulum-related compartments and then reaches the surface of adipocyte lipid droplets. Rab34 exerts distinct functions related to its intracellular location. Thus, at the Golgi, Rab34 regulates cisternae integrity as well as adiponectin trafficking and oligomerization. At the lipid droplets, this GTPase controls lipid accumulation and lipolysis through its interaction with the E1-ubiquitin ligase, UBA1, which induces the ubiquitination and proteasomal degradation of the fatty acid transporter and member of Rab34 interactome, FABP5. Finally, Rab34 levels in the adipose tissue and adipocytes are regulated in response to obesity and related pathogenic insults (i.e., fibrosis)., Conclusions: Rab34 plays relevant roles during adipocyte differentiation, including from the regulation of the oligomerization (i.e., biological activity) and secretion of a major adipokine with insulin-sensitizing actions, adiponectin, to lipid storage and mobilization from lipid droplets. Rab34 dysregulation in obesity may contribute to the altered adipokine secretion and lipid metabolism that characterize adipocyte dysfunction in conditions of excess adiposity., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. Rab18 Drift in Lipid Droplet and Endoplasmic Reticulum Interactions of Adipocytes under Obesogenic Conditions.
- Author
-
López-Alcalá J, Soler-Vázquez MC, Tercero-Alcázar C, Sánchez-Ceinos J, Guzmán-Ruiz R, Malagón MM, and Gordon A
- Subjects
- Humans, Adipocytes metabolism, Endoplasmic Reticulum metabolism, Lipid Droplets metabolism, Obesity metabolism
- Abstract
The adipose tissue stores excess energy in the form of neutral lipids within adipocyte lipid droplets (LDs). The correct function of LDs requires the interaction with other organelles, such as the endoplasmic reticulum (ER) as well as with LD coat-associated proteins, including Rab18, a mediator of intracellular lipid trafficking and ER-LD interaction. Although perturbations of the inter-organelle contact sites have been linked to several diseases, such as cancer, no information regarding ER-LD contact sites in dysfunctional adipocytes from the obese adipose tissue has been published to date. Herein, the ER-LD connection and Rab18 distribution at ER-LD contact sites are examined in adipocytes challenged with fibrosis and inflammatory conditions, which represent known hallmarks of the adipose tissue in obesity. Our results show that adipocytes differentiated in fibrotic conditions caused ER fragmentation, the expansion of ER-LD contact sites, and modified Rab18 dynamics. Likewise, adipocytes exposed to inflammatory conditions favored ER-LD contact, Rab18 accumulation in the ER, and Rab18 redistribution to large LDs. Finally, our studies in human adipocytes supported the suggestion that Rab18 transitions to the LD coat from the ER. Taken together, our results suggest that obesity-related pathogenic processes alter the maintenance of ER-LD interactions and interfere with Rab18 trafficking through these contact sites.
- Published
- 2023
- Full Text
- View/download PDF
20. Exploring the role of the inflammasomes on prostate cancer: Interplay with obesity.
- Author
-
Pérez-Gómez JM, Montero-Hidalgo AJ, Fuentes-Fayos AC, Sarmento-Cabral A, Guzmán-Ruiz R, Malagón MM, Herrera-Martínez AD, Gahete MD, and Luque RM
- Subjects
- Male, Humans, Aged, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Signal Transduction, Inflammation metabolism, Obesity metabolism, Inflammasomes metabolism, Prostatic Neoplasms
- Abstract
Obesity is a weight-related disorder characterized by excessive adipose tissue growth and dysfunction which leads to the onset of a systemic chronic low-grade inflammatory state. Likewise, inflammation is considered a classic cancer hallmark affecting several steps of carcinogenesis and tumor progression. In this regard, novel molecular complexes termed inflammasomes have been identified which are able to react to a wide spectrum of insults, impacting several metabolic-related disorders, but their contribution to cancer biology remains unclear. In this context, prostate cancer (PCa) has a markedly inflammatory component, and patients frequently are elderly individuals who exhibit weight-related disorders, being obesity the most prevalent condition. Therefore, inflammation, and specifically, inflammasome complexes, could be crucial players in the interplay between PCa and metabolic disorders. In this review, we will: 1) discuss the potential role of each inflammasome component (sensor, molecular adaptor, and targets) in PCa pathophysiology, placing special emphasis on IL-1β/NF-kB pathway and ROS and hypoxia influence; 2) explore the association between inflammasomes and obesity, and how these molecular complexes could act as the cornerstone between the obesity and PCa; and, 3) compile current clinical trials regarding inflammasome targeting, providing some insights about their potential use in the clinical practice., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
21. LRP10, PGK1 and RPLP0: Best Reference Genes in Periprostatic Adipose Tissue under Obesity and Prostate Cancer Conditions.
- Author
-
Pérez-Gómez JM, Porcel-Pastrana F, De La Luz-Borrero M, Montero-Hidalgo AJ, Gómez-Gómez E, Herrera-Martínez AD, Guzmán-Ruiz R, Malagón MM, Gahete MD, and Luque RM
- Subjects
- Male, Humans, Obesity genetics, Software, Adipose Tissue pathology, Reference Standards, LDL-Receptor Related Proteins, Phosphoglycerate Kinase, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Abstract
Obesity (OB) is a metabolic disorder characterized by adipose tissue dysfunction that has emerged as a health problem of epidemic proportions in recent decades. OB is associated with multiple comorbidities, including some types of cancers. Specifically, prostate cancer (PCa) has been postulated as one of the tumors that could have a causal relationship with OB. Particularly, a specialized adipose tissue (AT) depot known as periprostatic adipose tissue (PPAT) has gained increasing attention over the last few years as it could be a key player in the pathophysiological interaction between PCa and OB. However, to date, no studies have defined the most appropriate internal reference genes (IRGs) to be used in gene expression studies in this AT depot. In this work, two independent cohorts of PPAT samples ( n = 20/ n = 48) were used to assess the validity of a battery of 15 literature-selected IRGs using two widely used techniques (reverse transcription quantitative PCR [RT-qPCR] and microfluidic-based qPCR array). For this purpose, ΔCt method, GeNorm (v3.5), BestKeeper (v1.0), NormFinder (v.20.0), and RefFinder software were employed to assess the overall trends of our analyses. LRP10 , PGK1 , and RPLP0 were identified as the best IRGs to be used for gene expression studies in human PPATs, specifically when considering PCa and OB conditions.
- Published
- 2023
- Full Text
- View/download PDF
22. Influence of Protein Carbonylation on Human Adipose Tissue Dysfunction in Obesity and Insulin Resistance.
- Author
-
Navarro-Ruiz MC, Soler-Vázquez MC, Díaz-Ruiz A, Peinado JR, Nieto Calonge A, Sánchez-Ceinos J, Tercero-Alcázar C, López-Alcalá J, Rangel-Zuñiga OA, Membrives A, López-Miranda J, Malagón MM, and Guzmán-Ruiz R
- Abstract
Background: Obesity is characterized by adipose tissue dysregulation and predisposes individuals to insulin resistance and type 2 diabetes. At the molecular level, adipocyte dysfunction has been linked to obesity-triggered oxidative stress and protein carbonylation, considering protein carbonylation as a link between oxidative stress and metabolic dysfunction. The identification of specific carbonylated proteins in adipose tissue could provide novel biomarkers of oxidative damage related to metabolic status (i.e prediabetes). Thus, we aimed at characterizing the subcutaneous and omental human adipose tissue carbonylome in obesity-associated insulin resistance., Methods: 2D-PAGE was used to identify carbonylated proteins, and clinical correlations studies and molecular biology approaches including intracellular trafficking, reactive oxygen species assay, and iron content were performed using in vitro models of insulin resistance., Results: The carbonylome of human adipose tissue included common (serotransferrin, vimentin, actin, and annexin A2) and depot-specific (carbonic anhydrase and α-crystallin B in the subcutaneous depot; and α-1-antitrypsin and tubulin in the omental depot) differences that point out the complexity of oxidative stress at the metabolic level, highlighting changes in carbonylated transferrin expression. Posterior studies using in vitro prediabetic model evidence alteration in transferrin receptor translocation, linked to the prediabetic environment. Finally, ligand-receptor molecular docking studies showed a reduced affinity for carbonylated transferrin binding to its receptor compared to wild-type transferrin, emphasizing the role of transferrin carbonylation in the link between oxidative stress and metabolic dysfunction., Conclusions: The adipose tissue carbonylome contributes to understanding the molecular mechanism driving adipocyte dysfunction and identifies possible adipose tissue carbonylated targets in obesity-associated insulin resistance.
- Published
- 2022
- Full Text
- View/download PDF
23. Understanding the adipose tissue acetylome in obesity and insulin resistance.
- Author
-
Navarro-Ruiz MDC, López-Alcalá J, Díaz-Ruiz A, Moral SDD, Tercero-Alcázar C, Nieto-Calonge A, López-Miranda J, Tinahones FJ, Malagón MM, and Guzmán-Ruiz R
- Subjects
- Adipocytes metabolism, Adipose Tissue pathology, Humans, Lipids, Obesity pathology, Insulin Resistance
- Abstract
Obesity is a widely prevalent pathology with a high exponential growth worldwide. Altered lipid accumulation by adipose tissue is one of the main causes of obesity and exploring lipid homeostasis in this tissue may represent a source for the identification of possible therapeutic targets. The study of the proteome and the post-translational modifications of proteins, specifically acetylation due to its involvement in energy metabolism, may be of great interest to understand the molecular mechanisms involved in adipose tissue dysfunction in obesity. The objective of this study was to characterize the subcutaneous and omental adipose tissue acetylome in conditions of obesity and insulin resistance and to describe the importance of acetylation of key molecules in adipose tissue to use them as therapeutic targets. The results describe for the first time the acetylome of subcutaneous and omental adipose tissue under physiological and physiopathological conditions such as obesity and insulin resistance. New evidence showed different acetylation patterns between two main depots and highlight the molecular complexity of adipose tissue. Results showed changes in FABP4 acetylation in subcutaneous fat in relation to insulin resistance, thus unveiling a potential marker of depot-specific dysfunctional expansion in obesity-associated metabolic disease. Furthermore, it is shown that the acetylation of FABP4 affects its function, modulating the capacity of differentiation in adipocytes. In conclusion, this study demonstrates a profound, depot-specific alteration of adipose tissue acetylome, wherein the acetylation of FABP4 may play a key role in adipocyte differentiation and lipid accumulation., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. The clinical and molecular cardiometabolic fingerprint of an exploratory psoriatic arthritis cohort is associated with the disease activity and differentially modulated by methotrexate and apremilast.
- Author
-
Arias de la Rosa I, López-Montilla MD, Román-Rodríguez C, Pérez-Sánchez C, Gómez-García I, López-Medina C, Ladehesa-Pineda ML, Ábalos-Aguilera MDC, Ruiz D, Patiño-Trives AM, Luque-Tévar M, Añón-Oñate I, Pérez-Galán MJ, Guzmán-Ruiz R, Malagón MM, López-Pedrera C, Escudero-Contreras A, Collantes-Estévez E, and Barbarroja N
- Subjects
- Cross-Sectional Studies, Humans, Leukocytes, Mononuclear, Methotrexate therapeutic use, Thalidomide analogs & derivatives, Antirheumatic Agents therapeutic use, Arthritis, Psoriatic diagnosis, Arthritis, Psoriatic drug therapy, Cardiovascular Diseases drug therapy
- Abstract
Objectives: (1) To evaluate clinical and molecular cardiovascular disease (CVD) signs and their relationship with psoriatic arthritis (PsA) features and (2) to identify a clinical patient profile susceptible to benefit from methotrexate (MTX) and/or apremilast regarding CVD risk., Methods: This cross-sectional study included 100 patients with PsA and 100 age-matched healthy donors. In addition, an exploratory cohort of 45 biologically naïve patients treated for 6 months with apremilast, MTX or combined therapy according to routine clinical practice was recruited. Extensive clinical and metabolic profiles were obtained. Ninety-nine surrogate CVD-related molecules were analysed in plasma and peripheral blood mononuclear cells (PBMCs). Hard cluster analysis was performed to identify the clinical and molecular phenotypes. Mechanistic studies were performed on adipocytes., Results: Cardiometabolic comorbidities were associated with disease activity and long-term inflammatory status. Thirty-five CVD-related proteins were altered in the plasma and PBMCs of PsA patients and were associated with the key clinical features of the disease. Plasma levels of some of the CVD-related molecules might distinguish insulin-resistant patients (MMP-3, CD163, FABP-4), high disease activity (GAL-3 and FABP-4) and poor therapy outcomes (CD-163, LTBR and CNTN-1). Hard cluster analysis identified two phenotypes of patients according to the rates of cardiometabolic comorbidities with distinctive clinical and molecular responses to each treatment., Conclusions: (1) Novel CVD-related proteins associated with clinical features could be emerging therapeutic targets in the context of PsA and (2) the pleiotropic action of apremilast could make it an excellent choice for the management of PsA patients with high CVD risk, targeting metabolic alterations and CVD-related molecules., (© 2022 The Authors. Journal of Internal Medicine published by John Wiley & Sons Ltd on behalf of Association for Publication of The Journal of Internal Medicine.)
- Published
- 2022
- Full Text
- View/download PDF
25. Molecular Changes in the Adipose Tissue Induced by Rheumatoid Arthritis: Effects of Disease-Modifying Anti-Rheumatic Drugs.
- Author
-
Arias de la Rosa I, Escudero-Contreras A, Ruiz-Ponce M, Román-Rodríguez C, Pérez-Sánchez C, Ábalos-Aguilera MDC, Ortega-Castro R, Alcaide J, Murri M, Font P, Calvo-Gutiérrez J, Luque-Tevar M, Patiño-Trives AM, Guzmán-Ruiz R, Malagón MDM, Tinahones FJ, Collantes-Estévez E, López-Pedrera C, and Barbarroja N
- Subjects
- Adipose Tissue drug effects, Adult, Animals, Cross-Sectional Studies, Female, Humans, Male, Mice, Middle Aged, Adipose Tissue metabolism, Antirheumatic Agents therapeutic use, Arthritis, Rheumatoid complications, Arthritis, Rheumatoid drug therapy, Obesity complications
- Abstract
Disease severity, progression and response to therapy might be worse in obese rheumatoid arthritis (RA) patients, but paradoxically, obesity also might protect from radiographic joint damage. Thus, the intricate relationship between obesity and RA needs urgent clarification. The aim of this study was to assess the influence of obesity on the onset and development of RA and to determine whether arthritis could modify the adipose tissue biology and whether conventional Disease Modifying Anti-Rheumatic Drugs (cDMARDs) can modulate these alterations. Two strategies were followed: (1) clinical profiling of two cohorts of RA: non-obese and obese patients; and (2) mechanistic studies carried out in both a collagen-induced arthritis (CIA) in an obese mouse model and 3T3-L1 adipocytes treated with cDMARDs (leflunomide, methotrexate, and hydroxychloroquine). In our cohort of RA patients with low-moderate disease activity, the presence of obesity was not related to a higher activity of the disease; actually, disease activity score 28-erythrocyte sedimentation rate (DAS28-ESR) was reduced in the obese RA patients. However, the induction of arthritis promoted transcriptomic changes in the adipose tissue under obesity condition in the obese CIA model. Treatment with hydroxychloroquine reduced weight and insulin resistance, accompanied by beneficial metabolic effects in the adipose tissue. These molecular changes in adipose tissue were also observed after methotrexate administration. In sum, arthritis might affect directly the inflammatory burden and metabolic alterations associated with obesity in adipose tissue. Clinicians should be cautious measuring the activity of the disease in obesity and managing the best therapeutic options for the metabolic comorbidities of these patients, where the combination of hydroxychloroquine and methotrexate should be considered to improve adipose tissue dysfunction in obese RA., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Arias de la Rosa, Escudero-Contreras, Ruiz-Ponce, Román-Rodríguez, Pérez-Sánchez, Ábalos-Aguilera, Ortega-Castro, Alcaide, Murri, Font, Calvo-Gutiérrez, Luque-Tevar, Patiño-Trives, Guzmán-Ruiz, Malagón, Tinahones, Collantes-Estévez, López-Pedrera and Barbarroja.)
- Published
- 2021
- Full Text
- View/download PDF
26. Impaired mRNA splicing and proteostasis in preadipocytes in obesity-related metabolic disease.
- Author
-
Sánchez-Ceinos J, Guzmán-Ruiz R, Rangel-Zúñiga OA, López-Alcalá J, Moreno-Caño E, Del Río-Moreno M, Romero-Cabrera JL, Pérez-Martínez P, Maymo-Masip E, Vendrell J, Fernández-Veledo S, Fernández-Real JM, Laurencikiene J, Rydén M, Membrives A, Luque RM, López-Miranda J, and Malagón MM
- Subjects
- Adipocytes cytology, Adipocytes metabolism, Adipogenesis, Adult, Cell Differentiation, Cell Line, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Female, Humans, Male, Middle Aged, Obesity genetics, Obesity metabolism, Adipocytes physiology, Diabetes Mellitus, Type 2 complications, Obesity complications, Proteostasis, RNA, Messenger genetics
- Abstract
Preadipocytes are crucial for healthy adipose tissue expansion. Preadipocyte differentiation is altered in obese individuals, which has been proposed to contribute to obesity-associated metabolic disturbances. Here, we aimed at identifying the pathogenic processes underlying impaired adipocyte differentiation in obese individuals with insulin resistance (IR)/type 2 diabetes (T2D). We report that down-regulation of a key member of the major spliceosome, PRFP8 /PRP8, as observed in IR/T2D preadipocytes from subcutaneous (SC) fat, prevented adipogenesis by altering both the expression and splicing patterns of adipogenic transcription factors and lipid droplet-related proteins, while adipocyte differentiation was restored upon recovery of PRFP8 /PRP8 normal levels. Adipocyte differentiation was also compromised under conditions of endoplasmic reticulum (ER)-associated protein degradation (ERAD) hyperactivation, as occurs in SC and omental (OM) preadipocytes in IR/T2D obesity. Thus, targeting mRNA splicing and ER proteostasis in preadipocytes could improve adipose tissue function and thus contribute to metabolic health in obese individuals., Competing Interests: JS, RG, OR, JL, EM, MD, JR, PP, EM, JV, SF, JF, JL, MR, AM, RL, JL, MM No competing interests declared, (© 2021, Sánchez-Ceinos et al.)
- Published
- 2021
- Full Text
- View/download PDF
27. Crohn's Disease Increases the Mesothelial Properties of Adipocyte Progenitors in the Creeping Fat.
- Author
-
Madeira A, Serena C, Ejarque M, Maymó-Masip E, Millan M, Navarro-Ruiz MC, Guzmán-Ruiz R, Malagón MM, Espin E, Martí M, Menacho M, Megía A, Vendrell J, and Fernández-Veledo S
- Subjects
- Adipose Tissue pathology, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Biomarkers, Computational Biology methods, Crohn Disease etiology, Gene Expression Profiling, Humans, Immunophenotyping, Intra-Abdominal Fat metabolism, Mesothelin, Proteomics methods, Subcutaneous Fat metabolism, Transcriptome, Adipose Tissue metabolism, Crohn Disease metabolism, Crohn Disease pathology, Stem Cells metabolism
- Abstract
Our understanding of the interplay between human adipose tissue and the immune system is limited. The mesothelium, an immunologically active structure, emerged as a source of visceral adipose tissue. After investigating the mesothelial properties of human visceral and subcutaneous adipose tissue and their progenitors, we explored whether the dysfunctional obese and Crohn's disease environments influence the mesothelial/mesenchymal properties of their adipocyte precursors, as well as their ability to mount an immune response. Using a tandem transcriptomic/proteomic approach, we evaluated the mesothelial and mesenchymal expression profiles in adipose tissue, both in subjects covering a wide range of body-mass indexes and in Crohn's disease patients. We also isolated adipose tissue precursors (adipose-derived stem cells, ASCs) to assess their mesothelial/mesenchymal properties, as well as their antigen-presenting features. Human visceral tissue presented a mesothelial phenotype not detected in the subcutaneous fat. Only ASCs from mesenteric adipose tissue, named creeping fat, had a significantly higher expression of the hallmark mesothelial genes mesothelin ( MSLN ) and Wilms' tumor suppressor gene 1 ( WT1 ), supporting a mesothelial nature of these cells. Both lean and Crohn's disease visceral ASCs expressed equivalent surface percentages of the antigen-presenting molecules human leucocyte antigen-DR isotype (HLA-DR) and CD86. However, lean-derived ASCs were predominantly HLA-DR
dim , whereas in Crohn's disease, the HLA-DRbright subpopulation was increased 3.2-fold. Importantly, the mesothelial-enriched Crohn's disease precursors activated CD4+ T-lymphocytes. Our study evidences a mesothelial signature in the creeping fat of Crohn's disease patients and its progenitor cells, the latter being able to present antigens and orchestrate an immune response.- Published
- 2021
- Full Text
- View/download PDF
28. miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset.
- Author
-
Sánchez-Ceinos J, Rangel-Zuñiga OA, Clemente-Postigo M, Podadera-Herreros A, Camargo A, Alcalá-Diaz JF, Guzmán-Ruiz R, López-Miranda J, and Malagón MM
- Abstract
Circulating microRNAs (miRNAs) have been proposed as biomarkers for type 2 diabetes (T2D). Adipose tissue (AT), for which dysfunction is widely associated with T2D development, has been reported as a major source of circulating miRNAs. However, the role of dysfunctional AT in the altered pattern of circulating miRNAs associated with T2D onset remains unexplored. Herein, we investigated the relationship between T2D-associated circulating miRNAs and AT function, as well as the role of preadipocytes and adipocytes as secreting cells of candidate circulating miRNAs. Among the plasma miRNAs related to T2D onset in the CORonary Diet Intervention with Olive oil and cardiovascular PREVention (CORDIOPREV) cohort, baseline miR-223-3p levels (diminished in patients who next developed T2D [incident-T2D]) were significantly related to AT insulin resistance (IR). Baseline serum from incident-T2D participants induced inflammation and IR in 3T3-L1 adipocytes. We demonstrated that tumor necrosis factor (TNF)-α inhibited miR-223-3p secretion while enhancing miR-223-3p intracellular accumulation in 3T3-L1 (pre)adipocytes. Overexpression studies showed that an intracellular increase of miR-223-3p impaired glucose and lipid metabolism in these cells. Our findings provide mechanistic insights into the alteration of circulating miRNAs preceding T2D, unveiling both preadipocytes and adipocytes as miR-223-3p -secreting cells and suggesting that inflammation promotes miR-223-3p intracellular accumulation, which might contribute to (pre)adipocyte dysfunction and body metabolic dysregulation., Competing Interests: The authors declare no competing interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
29. Subclinical cardiovascular risk signs in adults with juvenile idiopathic arthritis in sustained remission.
- Author
-
Aranda-Valera IC, Arias de la Rosa I, Roldán-Molina R, Ábalos-Aguilera MDC, Torres-Granados C, Patiño-Trives A, Luque-Tevar M, Ibáñez-Costa A, Guzmán-Ruiz R, Malagón MDM, Escudero-Contreras A, López-Pedrera C, Collantes-Estévez E, and Barbarroja N
- Subjects
- Adult, Age of Onset, Asymptomatic Diseases, Biomarkers blood, Carotid Intima-Media Thickness, Cross-Sectional Studies, Female, Heart Disease Risk Factors, Humans, Inflammation Mediators blood, Insulin Resistance, Male, Oxidative Stress, Patient Acuity, Spain, Time, Arthritis, Juvenile blood, Arthritis, Juvenile diagnosis, Arthritis, Juvenile epidemiology, Arthritis, Juvenile physiopathology, Cytokines blood, Endothelium, Vascular metabolism, Inflammation blood, Inflammation immunology
- Abstract
Background: Juvenile Idiopathic Arthritis (JIA) is one of the most common chronic diseases of childhood that often persists into adulthood and can result in significant long-term morbidity. As a long lasting chronic inflammatory disease, concern has been raised regarding the risk of premature development of cardiovascular disease (CVD) in JIA. This study aims to determine whether adults with JIA in clinical remission display clinical and subclinical signs of CVD risk: inflammatory mediators, adipokines, endothelial dysfunction and oxidative stress markers., Methods: This is a cross-sectional study including 25 patients diagnosed with JIA according to the International League of Associations for Rheumatology criteria (ILAR 2001) and 25 age- and sex-matched controls. Remission was determined by JADAS10 < 1 and according to Wallace criteria. The presence of traditional CVD risk factors was analyzed. An extensive clinical analysis including body mass index (BMI), lipid profile, homeostatic model assessment - insulin resistance (HOMA-IR) and arterial blood pressure was performed. Intima media thickness of the common carotid artery (CIMT) was measured as a marker of subclinical atherosclerosis. Several proinflammatory cytokines, molecules involved in the endothelial dysfunction, oxidative stress and adipokines were quantified on serum by ELISA and on peripheral blood mononuclear cells (PBMCs) by RT-PCR. In vitro studies were carried out in healthy PBMCs, adipocytes and endothelial cells which were treated with serum from JIA patients under sustained remission., Results: Mean duration of the disease was 13.47 ± 5.47 years. Mean age was 25.11 ± 7.21. Time in remission was 3.52 ± 3.33 years. Patients were in remission with no treatment (40%) and with treatments (60%). CVD risk factors and CIMT were similar in JIA patients and controls. However, cholesterol levels were significantly elevated in JIA patients. Levels of adipocytokines, oxidative stress and endothelial activation markers were elevated in serum and PBMCs from JIA patients. Serum of those JIA patients induced the activation of adipocytes, endothelial cells and healthy PBMCs., Conclusions: JIA adult patients in remission have subclinical signs of inflammation and CVD risk, showed by an increase in the levels of inflammatory cytokines, endothelial activation and oxidative stress markers and adipokines, molecules closely involved in the alteration of the vascular system.
- Published
- 2020
- Full Text
- View/download PDF
30. Adipose tissue depot-specific intracellular and extracellular cues contributing to insulin resistance in obese individuals.
- Author
-
Guzmán-Ruiz R, Tercero-Alcázar C, Rabanal-Ruiz Y, Díaz-Ruiz A, El Bekay R, Rangel-Zuñiga OA, Navarro-Ruiz MC, Molero L, Membrives A, Ruiz-Rabelo JF, Pandit A, López-Miranda J, Tinahones FJ, and Malagón MM
- Subjects
- Adipocytes metabolism, Adipocytes pathology, Adipogenesis physiology, Adipose Tissue metabolism, Adult, Cues, Extracellular Matrix metabolism, Female, Guanine Nucleotide Dissociation Inhibitors metabolism, Humans, Lumican metabolism, Male, Middle Aged, Obesity metabolism, Proteomics methods, Subcutaneous Fat metabolism, Adipose Tissue pathology, Extracellular Matrix pathology, Insulin Resistance physiology, Obesity pathology
- Abstract
Adipose tissue dysregulation in obesity strongly influences systemic metabolic homeostasis and is often linked to insulin resistance (IR). However, the molecular mechanisms underlying adipose tissue dysfunction in obesity are not fully understood. Herein, a proteomic analysis of subcutaneous (SC) and omental (OM) fat from lean subjects and obese individuals with different degrees of insulin sensitivity was performed to identify adipose tissue biomarkers related to obesity-associated metabolic disease. Our results suggest that dysregulation of both adipose tissue extracellular matrix (ECM) organization and intracellular trafficking processes may be associated with IR in obesity. Thus, abnormal accumulation of the small leucine-rich proteoglycan, lumican, as observed in SC fat of IR obese individuals, modifies collagen I organization, impairs adipogenesis and activates stress processes [endoplasmic reticulum and oxidative stress] in adipocytes. In OM fat, IR is associated with increased levels of the negative regulator of the Rab family of small GTPases, GDI2, which alters lipid storage in adipocytes by inhibiting insulin-stimulated binding of the Rab protein, Rab18, to lipid droplets. Together, these results indicate that lumican and GDI2 might play depot-dependent, pathogenic roles in obesity-associated IR. Our findings provide novel insights into the differential maladaptive responses of SC and OM adipose tissue linking obesity to IR., (© 2020 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)
- Published
- 2020
- Full Text
- View/download PDF
31. Ghrelin Reduces TNF-α-Induced Human Hepatocyte Apoptosis, Autophagy, and Pyroptosis: Role in Obesity-Associated NAFLD.
- Author
-
Ezquerro S, Mocha F, Frühbeck G, Guzmán-Ruiz R, Valentí V, Mugueta C, Becerril S, Catalán V, Gómez-Ambrosi J, Silva C, Salvador J, Colina I, Malagón MM, and Rodríguez A
- Subjects
- Adult, Bariatric Surgery, Cross-Sectional Studies, Female, Gene Expression Regulation drug effects, Ghrelin metabolism, Humans, Liver pathology, Liver Function Tests, Male, Middle Aged, Non-alcoholic Fatty Liver Disease etiology, Obesity, Morbid complications, Obesity, Morbid surgery, Treatment Outcome, Apoptosis drug effects, Autophagy drug effects, Ghrelin pharmacology, Hepatocytes drug effects, Non-alcoholic Fatty Liver Disease pathology, Obesity, Morbid pathology, Pyroptosis drug effects, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Context: Human obesity is associated with increased circulating TNF-α, a proinflammatory cytokine that induces hepatocyte cell death., Objective: The potential beneficial effects of acylated and desacyl ghrelin in the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis in obesity via the inhibition of TNF-α-induced hepatocyte apoptosis, autophagic cell death, and pyroptosis were investigated., Design, Settings, and Participants: Plasma ghrelin isoforms and TNF-α were measured in 158 participants, and hepatocyte cell death was evaluated in liver biopsies from 76 patients with morbid obesity undergoing bariatric surgery with available liver echography and pathology analysis. The effect of acylated and desacyl ghrelin on basal and TNF-α-induced cell death was determined in vitro in human HepG2 hepatocytes., Results: Circulating TNF-α and the acylated/desacyl ghrelin ratio were increased, whereas desacyl ghrelin levels were decreased in patients with obesity and NAFLD. Six months after bariatric surgery, decreased acylated/desacyl ghrelin levels, and improved hepatic function were found. Patients with obesity and type 2 diabetes showed increased hepatic ghrelin O-acyltransferase transcripts as well as an increased hepatic apoptosis, pyroptosis, and compromised autophagy. In HepG2 hepatocytes, acylated and desacyl ghrelin treatment reduced TNF-α-induced apoptosis, evidenced by lower caspase-8 and caspase-3 cleavage, as well as TUNEL-positive cells and pyroptosis, revealed by decreased caspase-1 activation and lower high-mobility group box 1 expression. Moreover, acylated ghrelin suppressed TNF-α-activated hepatocyte autophagy, as evidenced by a decreased LC3B-II/I ratio and increased p62 accumulation via AMPK/mTOR., Conclusions: Ghrelin constitutes a protective factor against hepatocyte cell death. The increased acylated/desacyl ghrelin ratio in patients with obesity and NAFLD might constitute a compensatory mechanism to overcome TNF-α-induced hepatocyte apoptosis, autophagy, and pyroptosis.
- Published
- 2019
- Full Text
- View/download PDF
32. The caveolae-associated coiled-coil protein, NECC2, regulates insulin signalling in Adipocytes.
- Author
-
Trávez A, Rabanal-Ruiz Y, López-Alcalá J, Molero-Murillo L, Díaz-Ruiz A, Guzmán-Ruiz R, Catalán V, Rodríguez A, Frühbeck G, Tinahones FJ, Gasman S, Vitale N, Jiménez-Gómez Y, and Malagón MM
- Subjects
- 3T3-L1 Cells, Adipocytes, Adipogenesis genetics, Adipose Tissue metabolism, Adipose Tissue pathology, Animals, Caveolae metabolism, Caveolin 1 genetics, Humans, Mice, Microtubule-Associated Proteins genetics, Mitogen-Activated Protein Kinase 1 genetics, Obesity metabolism, Obesity pathology, Phosphorylation, Proto-Oncogene Proteins c-akt genetics, Receptor, Insulin genetics, Signal Transduction, Insulin genetics, Insulin Resistance genetics, Membrane Proteins genetics, Microtubule-Associated Proteins physiology, Obesity genetics
- Abstract
Adipocyte dysfunction in obesity is commonly associated with impaired insulin signalling in adipocytes and insulin resistance. Insulin signalling has been associated with caveolae, which are coated by large complexes of caveolin and cavin proteins, along with proteins with membrane-binding and remodelling properties. Here, we analysed the regulation and function of a component of caveolae involved in growth factor signalling in neuroendocrine cells, neuroendocrine long coiled-coil protein-2 (NECC2), in adipocytes. Studies in 3T3-L1 cells showed that NECC2 expression increased during adipogenesis. Furthermore, NECC2 co-immunoprecipitated with caveolin-1 (CAV1) and exhibited a distribution pattern similar to that of the components of adipocyte caveolae, CAV1, Cavin1, the insulin receptor and cortical actin. Interestingly, NECC2 overexpression enhanced insulin-activated Akt phosphorylation, whereas NECC2 downregulation impaired insulin-induced phosphorylation of Akt and ERK2. Finally, an up-regulation of NECC2 in subcutaneous and omental adipose tissue was found in association with human obesity and insulin resistance. This effect was also observed in 3T3-L1 adipocytes exposed to hyperglycaemia/hyperinsulinemia. Overall, the present study identifies NECC2 as a component of adipocyte caveolae that is regulated in response to obesity and associated metabolic complications, and supports the contribution of this protein as a molecular scaffold modulating insulin signal transduction at these membrane microdomains., (© 2018 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)
- Published
- 2018
- Full Text
- View/download PDF
33. The Effects of Bariatric Surgery-Induced Weight Loss on Adipose Tissue in Morbidly Obese Women Depends on the Initial Metabolic Status.
- Author
-
Moreno-Castellanos N, Guzmán-Ruiz R, Cano DA, Madrazo-Atutxa A, Peinado JR, Pereira-Cunill JL, García-Luna PP, Morales-Conde S, Socas-Macias M, Vázquez-Martínez R, Leal-Cerro A, and Malagón MM
- Subjects
- Adult, Bariatric Surgery, Female, Humans, Obesity, Morbid metabolism, Women's Health, Biomarkers metabolism, Insulin Resistance, Metabolic Syndrome metabolism, Obesity, Morbid surgery, Subcutaneous Fat, Abdominal metabolism, Weight Loss
- Abstract
Background: Adipose tissue (AT) dysfunction in obesity is commonly linked to insulin resistance and promotes the development of metabolic disease. Bariatric surgery (BS) represents an effective strategy to reduce weight and to improve metabolic health in morbidly obese subjects. However, the mechanisms and pathways that are modified in AT in response to BS are not fully understood, and few information is still available as to whether these may vary depending on the metabolic status of obese subjects., Methods: Abdominal subcutaneous adipose tissue (SAT) samples were obtained from morbidly obese women (n = 18) before and 13.3 ± 0.37 months after BS. Obese women were stratified into two groups: normoglycemic (NG; Glu < 100 mg/dl, HbA1c <5.7 %) or insulin resistant (IR; Glu 100-126 mg/dl, HbA1c 5.7-6.4 %) (n = 9/group). A multi-comparative proteomic analysis was employed to identify differentially regulated SAT proteins by BS and/or the degree of insulin sensitivity. Serum levels of metabolic, inflammatory, and anti-oxidant markers were also analyzed., Results: Before surgery, NG and IR subjects exhibited differences in AT proteins related to inflammation, metabolic processes, the cytoskeleton, and mitochondria. BS caused comparable weight reductions and improved glucose homeostasis in both groups. However, BS caused dissimilar changes in metabolic enzymes, inflammatory markers, cytoskeletal components, mitochondrial proteins, and angiogenesis regulators in NG and IR women., Conclusions: BS evokes significant molecular rearrangements indicative of improved AT function in morbidly obese women at either low or high metabolic risk, though selective adaptive changes in key cellular processes occur depending on the initial individual's metabolic status.
- Published
- 2016
- Full Text
- View/download PDF
34. Proteasome Dysfunction Associated to Oxidative Stress and Proteotoxicity in Adipocytes Compromises Insulin Sensitivity in Human Obesity.
- Author
-
Díaz-Ruiz A, Guzmán-Ruiz R, Moreno NR, García-Rios A, Delgado-Casado N, Membrives A, Túnez I, El Bekay R, Fernández-Real JM, Tovar S, Diéguez C, Tinahones FJ, Vázquez-Martínez R, López-Miranda J, and Malagón MM
- Subjects
- 3T3-L1 Cells, Adipocytes metabolism, Adult, Animals, Disease Models, Animal, Endoplasmic Reticulum Stress, Female, Gene Expression Regulation, Humans, Male, Mice, Obesity, Metabolically Benign metabolism, Obesity, Metabolically Benign pathology, Omentum cytology, Omentum metabolism, Omentum pathology, Palmitic Acid pharmacology, Proteomics methods, Subcutaneous Fat metabolism, Subcutaneous Fat pathology, Unfolded Protein Response, Adipocytes pathology, Insulin Resistance, Obesity, Metabolically Benign physiopathology, Oxidative Stress, Proteasome Endopeptidase Complex metabolism
- Abstract
Aims: Obesity is characterized by a low-grade systemic inflammatory state and adipose tissue (AT) dysfunction, which predispose individuals to the development of insulin resistance (IR) and metabolic disease. However, a subset of obese individuals, referred to as metabolically healthy obese (MHO) individuals, are protected from obesity-associated metabolic abnormalities. Here, we aim at identifying molecular factors and pathways in adipocytes that are responsible for the progression from the insulin-sensitive to the insulin-resistant, metabolically unhealthy obese (MUHO) phenotype., Results: Proteomic analysis of paired samples of adipocytes from subcutaneous (SC) and omental (OM) human AT revealed that both types of cells are altered in the MUHO state. Specifically, the glutathione redox cycle and other antioxidant defense systems as well as the protein-folding machinery were dysregulated and endoplasmic reticulum stress was increased in adipocytes from IR subjects. Moreover, proteasome activity was also compromised in adipocytes of MUHO individuals, which was associated with enhanced accumulation of oxidized and ubiquitinated proteins in these cells. Proteasome activity was also impaired in adipocytes of diet-induced obese mice and in 3T3-L1 adipocytes exposed to palmitate. In line with these data, proteasome inhibition significantly impaired insulin signaling in 3T3-L1 adipocytes., Innovation: This study provides the first evidence of the occurrence of protein homeostasis deregulation in adipocytes in human obesity, which, together with oxidative damage, interferes with insulin signaling in these cells., Conclusion: Our results suggest that proteasomal dysfunction and impaired proteostasis in adipocytes, resulting from protein oxidation and/or misfolding, constitute major pathogenic mechanisms in the development of IR in obesity.
- Published
- 2015
- Full Text
- View/download PDF
35. Remodeling of energy metabolism and absence of electrophysiological changes in the heart of obese hyperleptinemic mice. New insights into the pleiotropic role of leptin.
- Author
-
Guzmán-Ruiz R, Gómez-Hurtado N, Gil-Ortega M, Somoza B, González MC, Aránguez I, Martín-Ramos M, González-Martín C, Delgado C, Fernández-Alfonso M, and Ruiz-Gayo M
- Abstract
Dietary treatment with high-fat diets (HFD) triggers diabetes and hyperleptinemia, concomitantly with a partial state of leptin resistance that affects hepatic and adipose tissue but not the heart. In this context, characterized by widespread steatosis, cardiac lipid content remains unchanged. As previously reported, HFD-evoked hyperleptinemia could be a pivotal element contributing to increase fatty-acid (FA) metabolism in the heart and to prevent cardiac steatosis. This metabolic adaptation might theoretically reduce energy efficiency in cardiomyocytes and lead to cardiac electrophysiological remodeling. Therefore the aim of the current study has been to investigate the impact of long-term HFD on cardiac metabolism and electrophysiological properties of the principal ionic currents responsible of the action potential duration in mouse cardiomyocytes. Male C57BL/6J mice were fed a control (10 kcal% from fat) or HFD (45 kcal% from fat) during 32 weeks. Quantification of enzymatic activities regulating mitochondrial uptake of pyruvate and FA showed an increase of both carnitine-palmitoyltransferase and citrate synthase activities together with a decrease of lactate dehydrogenase and pyruvate dehydrogenase activities. Increased expression of uncoupling protein-3, Mn-, and Cu/Zn-superoxide dismutases and catalase were also detected. Total glutathione/oxidized glutathione ratios were unaffected by HFD. These data suggest that HFD triggers adaptive mechanisms aimed at (i) facilitating FA catabolism, and (ii) preventing oxidative stress. All these changes did not affect the duration of action potentials in cardiomyocytes and only slightly modified electrocardiographic parameters.
- Published
- 2013
- Full Text
- View/download PDF
36. Adipobiology for novel therapeutic approaches in metabolic syndrome.
- Author
-
Malagón MM, Díaz-Ruiz A, Guzmán-Ruiz R, Jiménez-Gómez Y, Moreno NR, García-Navarro S, Vázquez-Martínez R, and Peinado JR
- Subjects
- Animals, Energy Metabolism physiology, Humans, Insulin Resistance physiology, Metabolic Syndrome epidemiology, Obesity blood, Obesity epidemiology, Obesity therapy, Adipokines blood, Adipose Tissue metabolism, Metabolic Syndrome blood, Metabolic Syndrome therapy
- Abstract
Obesity is dramatically increasing virtually worldwide, which has been linked to the rising prevalence of metabolic syndrome. Excess fat accumulation causes severe alterations in adipose tissue function. Actually, adipose tissue is now recognized as a major endocrine and secretory organ that releases a wide variety of signaling molecules (hormones, growth factors, cytokines, chemokines, etc.), the adipokines, which play central roles in the regulation of energy metabolism and homeostasis, immunity and inflammation. In addition, adipose tissue is no longer regarded as a passive lipid storage site but as a highly dynamic energy depot which stores excess energy during periods of positive energy balance and mobilizes it in periods of nutrient deficiency in a tightly regulated manner. Altered lipid release and adipokine production and signaling, as occurs in obesity, are linked to insulin resistance and the associated comorbidities of metabolic syndrome (dyslipidemia, hypertension), which confer an increased risk for the development of type 2 diabetes and cardiovascular disease. Here we summarize current knowledge on adipose tissue and review the contribution of novel techniques and experimental approaches in adipobiology to the identification of novel biomarkers and potential targets for dietary or pharmacological intervention to prevent and treat adipose tissue-associated diseases.
- Published
- 2013
- Full Text
- View/download PDF
37. Leptin drives fat distribution during diet-induced obesity in mice.
- Author
-
Guzmán-Ruiz R, Stucchi P, Ramos MP, Sevillano J, Somoza B, Fernández-Alfonso M, and Ruiz-Gayo M
- Subjects
- Animals, Diet, High-Fat, Male, Mice, Mice, Inbred C57BL, Body Fat Distribution, Leptin physiology, Lipid Metabolism, Obesity metabolism, Receptors, Leptin physiology
- Abstract
Objective: Desensitization of leptin receptors is a process that specifically occurs in some tissues. We have hypothesized that during the development of obesity tissue lipids would increase gradually in particular organs depending on leptin responsiveness. Our aim was to establish a relationship between leptin resistance and lipid deposition by using a model of diet-induced obesity (DIO) and we have characterized, in mice undergoing a dietary treatment with a high-fat (HF) diet, the evolution of lipid content and leptin responsiveness in white adipose tissue and liver., Methods: Four-week-old male C57BL/6J mice were divided into two groups and assigned either to a low-fat or to a high-fat diet. Dietary treatment lasted 8, 20 or 32 weeks. The last day animals received 1mg/kg leptin and then tissues were weighed and processed for Western-blotting and lipid determination., Results: We observed an initial increase of the relative weight of adipose pads that was blunted after 32-week HF. In contrast, liver size exhibited an initial decrease followed by a progressive increase, which was coincident with the increase of hepatic triglycerides and with the impairment of leptin receptor signalling., Conclusion: Our data show that leptin resistance within white adipose tissue does not deal with an increase of the size of adipose pads and suggest that consequences of leptin resistance, in terms of fat accumulation, are tissue-dependent., (Copyright © 2011 SEEN. Published by Elsevier Espana. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
38. Apelin treatment increases complete Fatty Acid oxidation, mitochondrial oxidative capacity, and biogenesis in muscle of insulin-resistant mice.
- Author
-
Attané C, Foussal C, Le Gonidec S, Benani A, Daviaud D, Wanecq E, Guzmán-Ruiz R, Dray C, Bezaire V, Rancoule C, Kuba K, Ruiz-Gayo M, Levade T, Penninger J, Burcelin R, Pénicaud L, Valet P, and Castan-Laurell I
- Subjects
- AMP-Activated Protein Kinases physiology, Adipokines, Animals, Apelin, Cyclic AMP-Dependent Protein Kinases physiology, Diet, High-Fat, Energy Metabolism drug effects, Lipid Metabolism drug effects, Mice, Mice, Inbred C57BL, Oxidation-Reduction, Fatty Acids metabolism, Insulin Resistance, Intercellular Signaling Peptides and Proteins pharmacology, Mitochondria, Muscle metabolism, Muscle, Skeletal metabolism
- Abstract
Both acute and chronic apelin treatment have been shown to improve insulin sensitivity in mice. However, the effects of apelin on fatty acid oxidation (FAO) during obesity-related insulin resistance have not yet been addressed. Thus, the aim of the current study was to determine the impact of chronic treatment on lipid use, especially in skeletal muscles. High-fat diet (HFD)-induced obese and insulin-resistant mice treated by an apelin injection (0.1 μmol/kg/day i.p.) during 4 weeks had decreased fat mass, glycemia, and plasma levels of triglycerides and were protected from hyperinsulinemia compared with HFD PBS-treated mice. Indirect calorimetry experiments showed that apelin-treated mice had a better use of lipids. The complete FAO, the oxidative capacity, and mitochondrial biogenesis were increased in soleus of apelin-treated mice. The action of apelin was AMP-activated protein kinase (AMPK) dependent since all the effects studied were abrogated in HFD apelin-treated mice with muscle-specific inactive AMPK. Finally, the apelin-stimulated improvement of oxidative capacity led to decreased levels of acylcarnitines and enhanced insulin-stimulated glucose uptake in soleus. Thus, by promoting complete lipid use in muscle of insulin-resistant mice through mitochondrial biogenesis and tighter matching between FAO and the tricarboxylic acid cycle, apelin treatment could contribute to insulin sensitivity improvement.
- Published
- 2012
- Full Text
- View/download PDF
39. Circadian feeding drive of metabolic activity in adipose tissue and not hyperphagia triggers overweight in mice: is there a role of the pentose-phosphate pathway?
- Author
-
Stucchi P, Gil-Ortega M, Merino B, Guzmán-Ruiz R, Cano V, Valladolid-Acebes I, Somoza B, Le Gonidec S, Argente J, Valet P, Chowen JA, Fernández-Alfonso M, and Ruiz-Gayo M
- Subjects
- Adiposity, Animals, Corticosterone blood, Corticosterone metabolism, Dietary Fats administration & dosage, Fatty Acids, Nonesterified blood, Fatty Acids, Nonesterified metabolism, Insulin blood, Insulin metabolism, Leptin blood, Leptin metabolism, Male, Mice, Mice, Inbred C57BL, NADP metabolism, Weight Gain physiology, Adipose Tissue physiology, Circadian Rhythm physiology, Energy Metabolism physiology, Hyperphagia metabolism, Overweight etiology, Pentose Phosphate Pathway physiology
- Abstract
High-fat (HF) diets trigger an increase in adipose tissue and body weight (BW) and disordered eating behavior. Our study deals with the hypothesis that circadian distribution of energy intake is more relevant for BW dynamics than diet composition. Four-week-old mice were exposed for 8 wk to a HF diet and compared with animals receiving control chow. HF mice progressively increased BW, decreased the amount of nocturnal (1800-0900 h) calories (energy or food intake) (30%) and increased diurnal (0900-1800 h) caloric intake (energy or food intake), although total daily intake was identical between groups. Animals were killed at 3-h intervals and plasma insulin, leptin, corticosterone, glucose, and fatty acid levels quantified. Adipose tissue was weighed, and enzymatic activities integral to the pentose phosphate pathway (PPP) assayed in lumbar adipose tissue. Phosphorylated AMP-dependent protein kinase and fatty acid synthase were quantified by Western blotting. In HF mice, there was a shift in the circadian oscillations of plasma parameters together with an inhibition of PPP activity and a decrease in phosphorylated AMP-dependent protein kinase and fatty acid synthase. In a second experiment, HF mice were forced to adhere to a circadian pattern of food intake similar to that in control animals. In this case, BW, adipose tissue, morning plasma parameters and PPP activity appeared to be normal. These data indicate that disordered feeding behavior can trigger BW gain independently of food composition and daily energy intake. Because PPP is the main source of reduced nicotinamide adenine dinucleotide phosphate, we suggest that PPP inhibition might be an early marker of adipose dysfunction in diet-induced obesity.
- Published
- 2012
- Full Text
- View/download PDF
40. Leptin resistance develops spontaneously in mice during adult life in a tissue-specific manner. Consequences for hepatic steatosis.
- Author
-
Stucchi P, Guzmán-Ruiz R, Gil-Ortega M, Merino B, Somoza B, Cano V, de Castro J, Sevillano J, Ramos MP, Fernández-Alfonso MS, and Ruiz-Gayo M
- Subjects
- Animals, Blotting, Western, Body Weight drug effects, Lipids, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Myocardium metabolism, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Fatty Liver metabolism, Leptin pharmacology, Receptors, Leptin metabolism
- Abstract
Leptin is an adipocyte-derived hormone which stimulates β-oxidation in peripheral tissues and prevents steatosis. Because leptin production naturally increases during adult life, we have hypothesized that leptin receptors might undergo a physiological and gradual desensitization during ageing. Therefore we have characterized in three- five- and ten-month old mice i) the weight of different white adipose pads, heart and liver, ii) lipid content in these tissues/organs, and iii) responsiveness to acute leptin, measured in terms of phosphorylation of signal transducer and activator of transcription 3 (STAT3) and protein kinase B (Akt). In this study we have detected that leptin-mediated STAT3 phosphorylation appears to be preserved in cardiac tissue even in 10-month old animals but not in adipose tissue and liver of five- and ten-month old mice, respectively. Nevertheless, leptin increased pAkt content in the liver of these mice. In a parallel study we have analyzed the functionality of leptin signalling pathways in 10-month old obese mice and we have observed that the STAT3 pathway appears to be only operative in the heart whereas the Akt pathway remains functional both in heart and liver. Nevertheless, hepatic lipids increased almost 300% compared to age-matched lean controls. Our data demonstrate that during adult life there is a lost of leptin receptor functionality which is tissue-dependent and mainly affects the STAT3 pathway. Otherwise we demonstrate that the antisteatotic effect of leptin is independent of the Akt signalling pathway., (Copyright © 2011 Elsevier Masson SAS. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
41. Adaptative nitric oxide overproduction in perivascular adipose tissue during early diet-induced obesity.
- Author
-
Gil-Ortega M, Stucchi P, Guzmán-Ruiz R, Cano V, Arribas S, González MC, Ruiz-Gayo M, Fernández-Alfonso MS, and Somoza B
- Subjects
- Acetylcholine pharmacology, Adiponectin blood, Adipose Tissue drug effects, Animals, Dietary Fats pharmacology, Leptin blood, Male, Mesenteric Arteries drug effects, Mesenteric Arteries metabolism, Mice, Mice, Inbred C57BL, Nitroprusside metabolism, Obesity chemically induced, Radioimmunoassay, Adipose Tissue metabolism, Nitric Oxide metabolism, Obesity metabolism
- Abstract
Perivascular adipose tissue (PVAT) plays a paracrine role in regulating vascular tone. We hypothesize that PVAT undergoes adaptative mechanisms during initial steps of diet-induced obesity (DIO) which contribute to preserve vascular function. Four-week-old male C57BL/6J mice were assigned either to a control [low-fat (LF); 10% kcal from fat] or to a high-fat diet (HF; 45% kcal from fat). After 8 wk of dietary treatment vascular function was analyzed in the whole perfused mesenteric bed (MB) and in isolated mesenteric arteries cleaned of PVAT. Relaxant responses to acetylcholine (10(-9)-10(-4) m) and sodium nitroprusside (10(-12)-10(-5) m) were significantly ameliorated in the whole MB from HF animals. However, there was no difference between HF and LF groups in isolated mesenteric arteries devoid of PVAT. The enhancement of relaxant responses detected in HF mice was not attributable to an increased release of nitric oxide (NO) from the endothelium nor to an increased sensitivity and/or activity of muscular guanilylcyclase. Mesenteric PVAT of HF animals showed an increased bioavailability of NO, detected by 4,5-diaminofluorescein diacetate (DAF2-DA) staining, which positively correlated with plasma leptin levels. DAF-2DA staining was absent in PVAT from ob/ob mice but was detected in these animals after 4-wk leptin replacement. The main finding in this study is that adaptative NO overproduction occurs in PVAT during early DIO which might be aimed at preserving vascular function.
- Published
- 2010
- Full Text
- View/download PDF
42. Sensitivity of cardiac carnitine palmitoyltransferase to malonyl-CoA is regulated by leptin: similarities with a model of endogenous hyperleptinemia.
- Author
-
Guzmán-Ruiz R, Somoza B, Gil-Ortega M, Merino B, Cano V, Attané C, Castan-Laurell I, Valet P, Fernández-Alfonso MS, and Ruiz-Gayo M
- Subjects
- Animals, Disease Models, Animal, Heart Diseases etiology, Heart Diseases prevention & control, Leptin administration & dosage, Lipid Metabolism, Male, Mice, Mice, Inbred C57BL, Myocardium metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Ribonucleosides, STAT3 Transcription Factor metabolism, Triglycerides metabolism, Carnitine O-Palmitoyltransferase metabolism, Dietary Fats adverse effects, Heart Diseases metabolism, Leptin blood, Malonyl Coenzyme A metabolism
- Abstract
Acute leptin increase as well as endogenous hyperleptinemia evoked by high-fat diets (HF) activate fatty acid metabolism in nonadipose tissues. This supports the notion that hyperleptinemia is pivotal to prevent/delay steatosis during periods of positive energy balance. We have previously shown that long-term HF spares ectopic accumulation of lipids specifically in the miocardium. Because carnitine palmitoyltransferase I (CPT-I) allows mitochondrial uptake/oxidation of fatty acids, we have hypothesized that leptin drives cardiac CPT-I activity. In the current study, hyperleptinemia was induced in C57BL/6J mice either by exogenous leptin administration or by means of HF, and the ability of malonyl-coenzyme A (malonyl-CoA) (the main endogenous inhibitor of CPT-I) to inhibit cardiac CPT was analyzed. IC(50) values of malonyl-CoA were 8.1 +/- 1.5 micromol/liter in controls vs. 69.3 +/- 5.2 micromol/liter (P < 0.01) in leptin-treated mice. This effect was also observed in cardiac explants incubated with leptin and was blocked by triciribine, a compound shown to inhibit protein kinase B (Akt) phosphorylation (pAkt). In accordance, acute leptin evoked an increase of cardiac pAkt levels, which correlated with CPT sensitivity to malonyl-CoA. Otherwise, the inhibitory effect of malonyl-CoA was hindered in HF hyperleptinemic mice, and in this case, pAkt levels also correlated with CPT sensitivity to malonyl-CoA. Our data show that leptin reduces the sensitivity of cardiac CPT-I to malonyl-CoA and suggest the involvement of an Akt-related signaling pathway in this effect. This mechanism appears to be sensitive to both acute and chronic hyperleptinemia. We conclude that this action of leptin is pivotal to drive cardiac metabolism under situations associated to hyperleptinemia.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.