Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 190
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurol Sci ; 43(9): 5729-5734, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35676560

RESUMEN

INTRODUCTION: Gender medicine is a new medical approach aimed at the study of the differences between women and men in terms of prevention, diagnosis, and the outcome of all diseases. Migraines are among these. They represent the most common neurological illness; they are most prevalent in adults between 20 and 50 years of age and are three to four times more frequent in woman than in men. Affecting people in working age, migraines are a problem that strongly impacts the psychophysical health and productivity of workers, regardless of the specific job task they have. METHODS: A narrative review was performed, searching for the most relevant articles describing gender differences in people suffering from migraines, and particularly in workers. RESULTS: Migraine global prevalence is 20.7% in women and 9.7% in men whereas prevalence in Italy is 32.9% for women and only 13.0% for men. This difference is partly explained by hormonal differences, as well as by differences in brain structure, genetic polymorphisms and neuronal pathways. Sex differences may also play a role in the progression from episodic to chronic migraine. In workers, migraines are mostly associated with strenuous physical work in men, whilst migraines triggered by night shifts, lack of sleep, or irregular sleep patterns are more common in women. CONCLUSIONS: To this day, the reasons of sex/gender disparity for migraine are still obscure. However, migraines, chronic migraine in particular, have a negative impact on the lives of all individuals affected by this disease, but particularly in women in which family cares and working activity are often superimposed. Migraine prevention strategies should be planned in workers through the occupational health physician.


Asunto(s)
Trastornos Migrañosos , Adulto , Femenino , Humanos , Italia , Masculino , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/epidemiología , Prevalencia , Factores Sexuales , Sueño
2.
J Cell Physiol ; 234(11): 20453-20468, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30982981

RESUMEN

Cardiac dysfunction is often observed in patients with cancer also representing a serious problem limiting chemotherapeutic intervention and even patient survival. In view of the recently established role of the immune system in the control of cancer growth, the present work has been undertaken to investigate the effects of a panel of the most important inflammatory cytokines on the integrity and function of mitochondria, as well as of the cytoskeleton, two key elements in the functioning of cardiomyocytes. Either mitochondria features or actomyosin cytoskeleton organization of in vitro-cultured cardiomyocytes treated with different inflammatory cytokines were analyzed. In addition, to investigate the interplay between tumor growth and cardiac function in an in vivo system, immunocompetent female mice were inoculated with cancer cells and treated with the chemotherapeutic drug doxorubicin at a dosing schedule able to suppress tumor growth without inducing cardiac alterations. Analyses carried out in cardiomyocytes treated with the inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interferon γ (IFN-γ), interleukin 6 (IL-6), IL-8, and IL-1ß revealed severe phenotypic changes, for example, of contractile cytoskeletal elements, mitochondrial membrane potential, mitochondrial reactive oxygen species production and mitochondria network organization. Accordingly, in immunocompetent mice, the tumor growth was accompanied by increased levels of the inflammatory cytokines TNF-α, IFN-γ, IL-6, and IL-8, either in serum or in the heart tissue, together with a significant reduction of ventricular systolic function. The alterations of mitochondria and of microfilament system of cardiomyocytes, due to the systemic inflammation associated with cancer growth, could be responsible for remote cardiac injury and impairment of systolic function observed in vivo.


Asunto(s)
Citocinas/farmacología , Citoesqueleto/efectos de los fármacos , Inflamación/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Células Cultivadas , Femenino , Interferón gamma/farmacología , Interleucina-6/farmacología , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
3.
Cell Physiol Biochem ; 53(1): 186-199, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31278696

RESUMEN

BACKGROUND/AIMS: Estrogen could play a key role in the mechanisms underlying sex-related disparity in the incidence of thrombotic events. We investigated whether estrogen receptors (ERs) were expressed in human red blood cells (RBCs), and if they affected cell signaling of erythrocyte constitutive isoform of endothelial NO-synthase (eNOS) and nitric oxide (NO) release. METHODS: RBCs from 29 non-smoker volunteers (15 males and 14 females) aged between 20 and 40 years were analyzed by cytometry and western blot. In particular, content and distribution of ER-α and ER-ß, tyrosine kinases and eNOS phosphorylation and NO release were analyzed. RESULTS: We demonstrated that: i) both ER-α and ER-ß were expressed by RBCs; ii) they were both functionally active; and iii) ERs distribution and function were different in males and females. In particular, ERs modulated eNOS phosphorylation and NO release in RBCs from both sexes, but they induced the phosphorylation of specific tyrosine residues of kinases linked to eNOS activation and NO release in the RBCs from females only. CONCLUSION: Collectively, these data suggest that ERs could play a critical role in RBC intracellular signaling. The possible implication of this signaling in sex-linked risk disparity in human cardiovascular diseases, e.g. in thrombotic events, may not be ruled out.


Asunto(s)
Receptores de Estrógenos/metabolismo , Transducción de Señal , Adulto , Dronabinol/farmacología , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Adulto Joven
4.
J Cell Mol Med ; 22(7): 3308-3314, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29664231

RESUMEN

Systemic sclerosis (SSc) is a rare disorder of the connective tissue characterized by fibrosis of the skin, skeletal muscles and visceral organs. Additional manifestations include activation of the immune system and vascular injury. SSc causes disability and death as the result of end-stage organ failure. Two clinical subsets of the SSc are accepted: limited cutaneous SSc (lc-SSc) and diffuse cutaneous SSc (dc-SSc). At present, the aetiology and pathogenesis of SSc remain obscure, and consequently, disease outcome is unpredictable. Numerous studies suggest that reactive oxidizing species (ROS) play an important role in the pathogenesis of scleroderma. Over the years, several reports have supported this hypothesis for both lc-SSc and dc-SSc, although the specific role of oxidative stress in the pathogenesis of vascular injury and fibrosis remains to be clarified. The aim of the present review was to report and comment the recent findings regarding the involvement and role of oxidative stress in SSc pathogenesis. Biomarkers proving the link between ROS and the main pathological features of SSc have been summarized.


Asunto(s)
Antioxidantes/farmacología , Estrés Oxidativo/fisiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Esclerodermia Sistémica/tratamiento farmacológico
5.
Adv Exp Med Biol ; 1065: 241-256, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30051389

RESUMEN

The focus of this chapter is the gender differences in mitochondria in cardiovascular disease. There is broad evidence suggesting that some of the gender differences in cardiovascular outcome may be partially related to differences in mitochondrial biology (Ventura-Clapier R, Moulin M, Piquereau J, Lemaire C, Mericskay M, Veksler V, Garnier A, Clin Sci (Lond) 131(9):803-822, 2017)). Mitochondrial disorders are causally affected by mutations in either nuclear or mitochondrial genes involved in the synthesis of respiratory chain subunits or in their posttranslational control. This can be due to mutations of the mtDNA which are transmitted by the mother or mutations in the nuclear DNA. Because natural selection on mitochondria operates only in females, mutations may have had more deleterious effects in males than in females (Ventura-Clapier R, Moulin M, Piquereau J, Lemaire C, Mericskay M, Veksler V, Garnier A, Clin Sci (Lond) 131(9):803-822, 2017; Camara AK, Lesnefsky EJ, Stowe DF. Antioxid Redox Signal 13(3):279-347, 2010). As mitochondrial mutations can affect all tissues, they are responsible for a large panel of pathologies including neuromuscular disorders, encephalopathies, metabolic disorders, cardiomyopathies, neuropathies, renal dysfunction, etc. Many of these pathologies present sex/gender specificity. Thus, alleviating or preventing mitochondrial dysfunction will contribute to mitigating the severity or progression of the development of diseases. Here, we present evidence for the involvement of mitochondria in the sex specificity of cardiovascular disorders.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Disparidades en el Estado de Salud , Corazón/fisiopatología , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Animales , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/patología , Femenino , Humanos , Masculino , Mitocondrias Cardíacas/patología , Miocardio/patología , Factores de Riesgo , Caracteres Sexuales , Factores Sexuales , Transducción de Señal
6.
J Cell Mol Med ; 21(4): 632-639, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28063205

RESUMEN

Kawasaki disease is a multisystemic vasculitis that can result in coronary artery lesions. It predominantly affects young children and is characterized by prolonged fever, diffuse mucosal inflammation, indurative oedema of the hands and feet, a polymorphous skin rash and non-suppurative lymphadenopathy. Coronary artery involvement is the most important complication of Kawasaki disease and may cause significant coronary stenosis resulting in ischemic heart disease. The introduction of intravenous immunoglobulin decreases the incidence of coronary artery lesions to less than 5%. The etiopathogenesis of this disease remains unclear. Several lines of evidence suggest that an interplay between a microbial infection and a genetic predisposition could take place in the development of the disease. In this review, we summarize the state of the art of pathogenetic mechanisms of Kawasaki disease underscoring the relevance of haematological features as a novel field of investigation.


Asunto(s)
Síndrome Mucocutáneo Linfonodular/sangre , Síndrome Mucocutáneo Linfonodular/etiología , Biomarcadores/sangre , Humanos , Inmunidad , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/patología , Pronóstico
7.
Apoptosis ; 22(5): 681-695, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28299505

RESUMEN

Once activated, some surface receptors promote membrane movements that open new portals of endocytosis, in part to facilitate the internalization of their activated complexes. The prototypic death receptor Fas (CD95/Apo1) promotes a wave of enhanced endocytosis that induces a transient intermixing of endosomes with mitochondria in cells that require mitochondria to amplify death signaling. This initiates a global alteration in membrane traffic that originates from changes in key membrane lipids occurring in the endoplasmic reticulum (ER). We have focused the current study on specific lipid changes occurring early after Fas ligation. We analyzed the interaction between endosomes and mitochondria in Jurkat T cells by nanospray-Time-of-flight (ToF) Mass Spectrometry. Immediately after Fas ligation, we found a transient wave of lipid changes that drives a subpopulation of early endosomes to merge with mitochondria. The earliest event appears to be a decrease of phosphatidylcholine (PC), linked to a metabolic switch enhancing phosphatidylinositol (PI) and phosphoinositides, which are crucial for the formation of vacuolar membranes and endocytosis. Lipid changes occur independently of caspase activation and appear to be exacerbated by caspase inhibition. Conversely, inhibition or compensation of PC deficiency attenuates endocytosis, endosome-mitochondria mixing and the induction of cell death. Deficiency of receptor interacting protein, RIP, also limits the specific changes in membrane lipids that are induced by Fas activation, with parallel reduction of endocytosis. Thus, Fas activation rapidly changes the interconversion of PC and PI, which then drives enhanced endocytosis, thus likely propagating death signaling from the cell surface to mitochondria and other organelles.


Asunto(s)
Caspasas/metabolismo , Endocitosis/genética , Lípidos de la Membrana/metabolismo , Receptor fas/genética , Humanos , Células Jurkat , Espectrometría de Masas , Lípidos de la Membrana/genética , Mitocondrias/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilinositoles/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptor fas/metabolismo
9.
Clin Sci (Lond) ; 131(6): 449-469, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28265036

RESUMEN

The biology of sex differences deals with the study of the disparities between females and males and the related biological mechanisms. Gender medicine focuses on the impact of gender and sex on human physiology, pathophysiology and clinical features of diseases that are common to women and men. The term gender refers to a complex interrelation and integration of sex-as a biological and functional determinant-and psychological and cultural behaviours (due to ethnical, social or religious background). The attention to the impact of gender differences on the pathophysiology and, therefore, on the clinical management of the most common diseases, such as cardiovascular diseases (CVD), neurodegenerative disorders, immune and autoimmune diseases as well as several tumours, is in fact often neglected. Hence, studies covering different fields of investigation and including sex differences in the pathogenesis, in diagnostic and prognostic criteria as well as in response to therapy appear mandatory. However, prerequisites for this development are preclinical studies, including in vitro and in vivo approaches. They represent the first step in the development of a drug or in the comprehension of the pathogenetic mechanisms of diseases, in turn a necessary step for the development of new or more appropriate therapeutic strategies. However, sex differences are still poorly considered and the great majority of preclinical studies do not take into account the relevance of such disparities. In this review, we describe the state of the art of these studies and provide some paradigmatic examples of key fields of investigation, such as oncology, neurology and CVD, where preclinical models should be improved.


Asunto(s)
Modelos Animales de Enfermedad , Caracteres Sexuales , Enfermedad de Alzheimer/epidemiología , Animales , Línea Celular , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Leucemia/epidemiología , Linfoma/epidemiología , Masculino , Melanoma/epidemiología , Distribución por Sexo , Accidente Cerebrovascular/epidemiología
10.
J Cell Physiol ; 231(2): 295-302, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26206426

RESUMEN

As a general rule, smooth muscle cells (SMC) are able to switch from a contractile phenotype to a less mature synthetic phenotype. This switch is accompanied by a loss of differentiation with decreased expression of contractile markers, increased proliferation as well as the synthesis and the release of several signaling molecules such as pro-inflammatory cytokines, chemotaxis-associated molecules, and growth factors. This SMC phenotypic plasticity has extensively been investigated in vascular diseases, but interest is also emerging in the field of gastroenterology. It has in fact been postulated that altered microenvironmental conditions, including the composition of microbiota, could trigger the remodeling of the enteric SMC, with phenotype changes and consequent alterations of contraction and impairment of gut motility. Several molecular actors participate in this phenotype remodeling. These include extracellular molecules such as cytokines and extracellular matrix proteins, as well as intracellular proteins, for example, transcription factors. Epigenetic control mechanisms and miRNA have also been suggested to participate. In this review key roles and actors of smooth muscle phenotypic switch, mainly in GI tissue, are described and discussed in the light of literature data available so far. J. Cell. Physiol. 231: 295-302, 2016. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Microambiente Celular , Microbioma Gastrointestinal , Motilidad Gastrointestinal , Tracto Gastrointestinal/patología , Homeostasis , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Contracción Muscular , Miocitos del Músculo Liso/patología , Fenotipo , Transducción de Señal
11.
Apoptosis ; 20(5): 645-57, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25697338

RESUMEN

Apoptosis and autophagy are two evolutionary conserved processes that exert a critical role in the maintenance of tissue homeostasis. While apoptosis is a tightly regulated cell program implicated in the removal of damaged or unwanted cells, autophagy is a cellular catabolic pathway that is involved in the lysosomal degradation and recycling of proteins and organelles, and is thereby considered an important cytoprotection mechanism. Sphingolipids (SLs), which are ubiquitous membrane lipids in eukaryotes, participate in the generation of various membrane structures, including lipid rafts and caveolae, and contribute to a number of cellular functions such as cell proliferation, apoptosis and, as suggested more recently, autophagy. For instance, SLs are hypothesized to be involved in several intracellular processes, including organelle membrane scrambling, whilst at the plasma membrane lipid rafts, acting as catalytic domains, strongly contribute to the ignition of critical signaling pathways determining cell fate. In particular, by targeting several shared regulators, ceramide, sphingosine-1-phosphate, dihydroceramide, sphingomyelin and gangliosides seem able to differentially regulate the autophagic pathway and/or contribute to the autophagosome formation. This review illustrates recent studies on this matter, particularly lipid rafts, briefly underscoring the possible implication of SLs and their alterations in the autophagy disturbances and in the pathogenesis of some human diseases.


Asunto(s)
Autofagia , Fagosomas/fisiología , Esfingolípidos/fisiología , Animales , Apoptosis , Humanos , Morfogénesis , Biogénesis de Organelos , Transducción de Señal
12.
Apoptosis ; 20(12): 1577-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423274

RESUMEN

Prostate cancer is among the most commonly diagnosed male diseases and a leading cause of cancer mortality in men. There is emerging evidence that autophagy plays an important role in malignant cell survival and offers protection from the anti-cancer drugs in prostate cancer cells. AMBRA1 and the autophagic protein sequestosome-1 (SQSTM1; p62) expression were evaluated by immunohistochemistry and western blot on tissue samples from both benign and malignant prostatic lesions. The data reported in this pilot study demonstrated an increased expression of AMBRA1 and SQSTM1, which were also associated with an accumulation of LC3II in prostate cancer but not in benign lesion. In the present study we found that: (i) at variance with benign lesion, prostate cancer cells underwent SQSTM1 accumulation, i.e., clearly displayed a defective autophagic process but, also, (ii) prostate cancer accumulated AMBRA1 and (iii) this increase positively correlated with the Gleason score. These results underscore a possible implication of autophagy in prostate cancer phenotype and of AMBRA1 as possible cancer progression biomarker in this malignancy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Próstata/metabolismo , Anciano , Antineoplásicos/uso terapéutico , Autofagia/fisiología , Biomarcadores de Tumor/metabolismo , Supervivencia Celular/fisiología , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Estudios Retrospectivos , Proteína Sequestosoma-1
13.
J Autoimmun ; 58: 78-89, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25623267

RESUMEN

T lymphocytes from patients with Systemic Lupus Erythematosus (SLE) display multiple abnormalities, including increased cell activation, abnormal cell death by apoptosis and impairment of autophagy pathway. In the present study we report the presence of specific antibodies to D4GDI, a small GTPase family inhibitor, in a significant percentage (46%) of SLE patient sera. We also found a significant association between the presence of these autoantibodies and hematologic manifestations occurring in these patients. Investigating the possible implication of anti-D4GDI autoantibodies in SLE pathogenesis or progression, we found that these antibodies were capable of binding D4GDI expressed at the lymphocyte surface and triggering a series of subcellular events, including Rho GTPase activation. These antibodies were also able to induce autophagy in T cells from both healthy donors and SLE patients, but only those negative to these antibodies. We can conclude that anti-D4GDI autoantibodies could be capable of triggering important responses in T cells such as cytoskeleton remodeling and autophagy pathway and that, in SLE patients, the chronic exposure to these specific autoantibodies could lead to the selection of autophagy-resistant T cell clones contributing to the pathogenesis of the disease.


Asunto(s)
Autoanticuerpos/sangre , Lupus Eritematoso Sistémico/inmunología , Linfocitos T/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Inhibidor beta de Disociación del Nucleótido Guanina rho/inmunología , Adulto , Anciano , Autofagia/genética , Citoesqueleto/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Células Jurkat , Masculino , Persona de Mediana Edad , Unión Proteica/genética , ARN Interferente Pequeño/genética , Linfocitos T/inmunología , Adulto Joven , Inhibidor beta de Disociación del Nucleótido Guanina rho/genética
14.
FASEB J ; 28(2): 524-35, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24196588

RESUMEN

Autophagy represents a key mechanism of cytoprotection that can be activated by a variety of extracellular and intracellular stresses and allows the cell to sequester cytoplasmic components and damaged organelles, delivering them to lysosomes for degradation and recycling. However, the autophagy process has also been associated with the death of the cell. It has been demonstrated to be constitutive in some instances and inducible in others, and the idea that it could represent a pathogenetic determinant as well as a possible prognostic tool and a therapeutic target in a plethora of human diseases has recently been considered. Among these, cancer represents a major one. In this review, we recapitulate the critical implications of autophagy in the pathogenesis, progression, and treatment of lymphoproliferative disorders. Leukemias and lymphomas, in fact, represent paradigmatic human diseases in which advances have recently been made in this respect.


Asunto(s)
Autofagia/fisiología , Trastornos Linfoproliferativos/fisiopatología , Animales , Enfermedad de Hodgkin/fisiopatología , Humanos , Linfocitos/fisiología , Linfoma no Hodgkin/fisiopatología
15.
FASEB J ; 28(8): 3745-57, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24806198

RESUMEN

The mineralocorticoid receptor (MR) controls adipocyte function, but its role in the conversion of white adipose tissue (WAT) into thermogenic fat has not been elucidated. We investigated responses to the MR antagonists spironolactone (spiro; 20 mg/kg/d) and drospirenone (DRSP; 6 mg/kg/d) in C57BL/6 mice fed a high-fat (HF) diet for 90 d. DRSP and spiro curbed HF diet-induced impairment in glucose tolerance, and prevented body weight gain and white fat expansion. Notably, either MR antagonist induced up-regulation of brown adipocyte-specific transcripts and markedly increased protein levels of uncoupling protein 1 (UCP1) in visceral and inguinal fat depots when compared with the HF diet group. Positron emission tomography and magnetic resonance spectroscopy confirmed acquisition of brown fat features in WAT. Interestingly, MR antagonists markedly reduced the autophagic rate both in murine preadipocytes in vitro (10(-5) M) and in WAT depots in vivo, with a concomitant increase in UCP1 protein expression. Moreover, the autophagy repressor bafilomycin A1 (10(-8) M) mimicked the effect of MR antagonists, increasing UCP1 protein expression in primary preadipocytes. Hence, we showed that adipocyte MR regulates brown remodeling of WAT through a modulation of autophagy. These results provide a rationale for the use of MR antagonists to prevent the adverse metabolic consequences of adipocyte dysfunction.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Androstenos/farmacología , Autofagia/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/prevención & control , Antagonistas de Receptores de Mineralocorticoides/farmacología , Receptores de Mineralocorticoides/fisiología , Espironolactona/farmacología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Aldosterona/farmacología , Androstenos/uso terapéutico , Animales , Composición Corporal/efectos de los fármacos , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Intolerancia a la Glucosa/etiología , Conducto Inguinal , Grasa Intraabdominal/efectos de los fármacos , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Macrólidos/farmacología , Ratones , Ratones Endogámicos C57BL , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Obesidad/prevención & control , Receptores de Mineralocorticoides/efectos de los fármacos , Espironolactona/uso terapéutico , Proteína Desacopladora 1 , Regulación hacia Arriba/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
16.
J Cell Physiol ; 229(12): 1990-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24777636

RESUMEN

Statins, widely used for treatment of hypercholesterolemia, have been demonstrated to exert pleiotropic beneficial effects independently of their cholesterol-lowering action, such as anti-inflammatory activity. A gender disparity has been observed in their cholesterol lowering activity as well as in response to these "off label" effects. Monocytes play a central role in atherosclerotic disease and, more in general, in inflammatory responses, through their chemotactic function and cytokine production. On these bases, in the present work, we examined the effect of statins on homeostasis and migration properties of freshly isolated monocytes from male and female healthy donors. Two prototypic natural and synthetic statins with different polarity, that is, type 1 and type 2 statins, have been considered: simvastatin and atorvastatin. Freshly isolated monocytes from peripheral blood of male and female healthy donors were treated with these drugs in the absence or presence of lipopolysaccharide (LPS) stimulation. Results obtained indicated that the polar statin efficiently inhibited chemotaxis of monocytes more than the apolar statin and that this effect was more significantly induced in cells from females than in cells from males. Dissecting the mechanisms involved, we found that these results could mainly be due to differential effects on: (i) the release of key cytokines, for example, MCP-1 and TNF-α; (ii) the maintenance of the redox homeostasis; (iii) a target activity on microfilament network integrity and function. All in all these results could suggest a reappraisal of "off-label" effects of statins taking into account either their chemical structure, that is, molecular polarity, or the gender issue.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Monocitos/efectos de los fármacos , Caracteres Sexuales , Aterosclerosis/patología , Atorvastatina , Movimiento Celular/efectos de los fármacos , Femenino , Voluntarios Sanos , Ácidos Heptanoicos/administración & dosificación , Humanos , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Masculino , Pirroles/administración & dosificación , Simvastatina/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
17.
Ann Rheum Dis ; 73(10): 1888-97, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897774

RESUMEN

OBJECTIVE: Accumulation of mitochondria underlies T-cell dysfunction in systemic lupus erythematosus (SLE). Mitochondrial turnover involves endosomal traffic regulated by HRES-1/Rab4, a small GTPase that is overexpressed in lupus T cells. Therefore, we investigated whether (1) HRES-1/Rab4 impacts mitochondrial homeostasis and (2) Rab geranylgeranyl transferase inhibitor 3-PEHPC blocks mitochondrial accumulation in T cells, autoimmunity and disease development in lupus-prone mice. METHODS: Mitochondria were evaluated in peripheral blood lymphocytes (PBL) of 38 SLE patients and 21 healthy controls and mouse models by flow cytometry, microscopy and western blot. MRL/lpr mice were treated with 125 µg/kg 3-PEHPC or 1 mg/kg rapamycin for 10 weeks, from 4 weeks of age. Disease was monitored by antinuclear antibody (ANA) production, proteinuria, and renal histology. RESULTS: Overexpression of HRES-1/Rab4 increased the mitochondrial mass of PBL (1.4-fold; p=0.019) and Jurkat cells (2-fold; p=0.000016) and depleted the mitophagy initiator protein Drp1 both in human (-49%; p=0.01) and mouse lymphocytes (-41%; p=0.03). Drp1 protein levels were profoundly diminished in PBL of SLE patients (-86±3%; p=0.012). T cells of 4-week-old MRL/lpr mice exhibited 4.7-fold over-expression of Rab4A (p=0.0002), the murine homologue of HRES-1/Rab4, and depletion of Drp1 that preceded the accumulation of mitochondria, ANA production and nephritis. 3-PEHPC increased Drp1 (p=0.03) and reduced mitochondrial mass in T cells (p=0.02) and diminished ANA production (p=0.021), proteinuria (p=0.00004), and nephritis scores of lupus-prone mice (p<0.001). CONCLUSIONS: These data reveal a pathogenic role for HRES-1/Rab4-mediated Drp1 depletion and identify endocytic control of mitophagy as a treatment target in SLE.


Asunto(s)
GTP Fosfohidrolasas/sangre , Lupus Eritematoso Sistémico/sangre , Proteínas Asociadas a Microtúbulos/sangre , Mitocondrias/metabolismo , Proteínas Mitocondriales/sangre , Proteínas de Unión al GTP rab4/fisiología , Animales , Autofagia/fisiología , Estudios de Casos y Controles , Células Cultivadas , Difosfonatos/uso terapéutico , Dinaminas/sangre , Dinaminas/fisiología , Femenino , GTP Fosfohidrolasas/fisiología , Homeostasis/fisiología , Humanos , Células Jurkat , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lisosomas/metabolismo , Ratones Endogámicos MRL lpr , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Mitocondriales/fisiología , Mitofagia/inmunología , Terapia Molecular Dirigida/métodos , Piridinas/uso terapéutico , Linfocitos T/metabolismo
18.
J Pathol ; 229(2): 208-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23018629

RESUMEN

Fibrotic disorders are multistage progressive processes that often arise from different causes and are commonly associated with chronic inflammation. Excessive deposition of extracellular matrix is the hallmark of many fibrotic diseases. This may be due to an excess of fibroblast recruitment and activation, as well as to their differentiation in myofibroblasts. These events may be triggered by cytokines, chemokines and growth factors released by lymphocytes or macrophages. The excessive production of extracellular matrix is apparently due to alterations of metabolic pathways in activated fibroblasts. It has been suggested that a defective autophagy, an important subcellular pathway with multiple homeostatic roles, also recognized as a key component of both innate and acquired immunity, could play a role. In this review we illustrate recent insights in the field, suggesting the possible implication of the immune system in orchestrating the fibrotic response via the modulation of autophagic pathways.


Asunto(s)
Autofagia , Fibroblastos/patología , Animales , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibrosis , Homeostasis , Humanos , Transducción de Señal
19.
Carcinogenesis ; 34(10): 2424-33, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23729654

RESUMEN

The oncoprotein E7 from human papillomavirus-16 (HPV-16 E7) plays a pivotal role in HPV postinfective carcinogenesis, and its physical interaction with host cell targets is essential to its activity. We identified a novel cellular partner for the viral oncoprotein: the actin-binding protein gelsolin (GSN), a key regulator of actin filament assembly and disassembly. In fact, biochemical analyses, generation of a 3D molecular interaction model and the use of specific HPV-16 E7 mutants provided clear cut evidence supporting the crucial role of HPV-16 E7 in affecting GSN integrity and function in human immortalized keratinocytes. Accordingly, functional analyses clearly suggested that stable HPV-16 E7 expression induced an imbalance between polymeric and monomeric actin in favor of the former. These events also lead to changes of cell cycle (increased S phase), to the inhibition of apoptosis and to the increase of cell survival. These results provide support to the hypotheses generated from the 3D molecular interaction model and encourage the design of small molecules hindering HPV-induced host cell reprogramming by specifically targeting HPV-16 E7-expressing cells.


Asunto(s)
Apoptosis , Gelsolina/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Actinas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Caspasa 3/metabolismo , Línea Celular Tumoral , Gelsolina/química , Humanos , Simulación del Acoplamiento Molecular , Mutación , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/genética , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas
20.
FASEB J ; 26(4): 1400-12, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22247332

RESUMEN

Autophagy is a lysosome-mediated catabolic process that allows cells to degrade unwanted cytoplasmic constituents and to recycle nutrients. Autophagy is also involved in innate and adaptive immune responses, playing a key role in interactions against microbes, in antigen processing for major histocompatibility complex (MHC) presentation, and in lymphocyte development, survival, and proliferation. Over recent years, perturbations in autophagy have been implicated in a number of diseases, including autoimmunity. Systemic lupus erythematosus (SLE) is a multifactorial disease characterized by autoimmune responses against self-antigens generated by dying cells. Genome-wide association studies have linked several single-nucleotide polymorphisms (SNPs) in the autophagy-related gene Atg5 to SLE susceptibility. Loss of Atg5-dependent effects, including clearance of dying cells and cell antigen presentation, might contribute to the autoimmunity and inflammation associated with SLE. Moreover, activation of the mammalian target of rapamycin (mTOR), a key player in the autophagy regulation, has recently been demonstrated in SLE, confirming an altered autophagy pathway in this disease. In the present review, we summarize the autophagy mechanisms, their molecular regulation, and their relevance in immunity and autoimmunity. The potential of targeting autophagy pathway in SLE, by developing innovative therapeutic approaches, has finally been discussed.


Asunto(s)
Inmunidad Adaptativa , Autoinmunidad/inmunología , Autofagia/inmunología , Inmunidad Innata , Lupus Eritematoso Sistémico/inmunología , Humanos , Lupus Eritematoso Sistémico/fisiopatología , Linfocitos/inmunología , Fagocitos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA