Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 37(8): e23061, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37389926

RESUMEN

Endometriosis is a chronic gynecological disease affecting ~10% women in the reproductive age characterized by the growth of endometrial glands and stroma outside the uterine cavity. The inflammatory process has a key role in the initiation and progression of the disorder. Currently, there are no available early diagnostic tests and therapy relies exclusively on symptomatic drugs, so that elucidation of the complex molecular mechanisms involved in the pathogenesis of endometriosis is an unmet need. The signaling of the bioactive sphingolipid sphingosine 1-phosphate (S1P) is deeply dysregulated in endometriosis. S1P modulates a variety of fundamental cellular processes, including inflammation, neo-angiogenesis, and immune responses acting mainly as ligand of a family of G-protein-coupled receptors named S1P receptors (S1PR), S1P1-5 . Here, we demonstrated that the mitogen-activated protein kinase ERK5, that is expressed in endometriotic lesions as determined by quantitative PCR, is activated by S1P in human endometrial stromal cells. S1P-induced ERK5 activation was shown to be triggered by S1P1/3 receptors via a SFK/MEK5-dependent axis. S1P-induced ERK5 activation was, in turn, responsible for the increase of reactive oxygen species and proinflammatory cytokine expression in human endometrial stromal cells. The present findings indicate that the S1P signaling, via ERK5 activation, supports a proinflammatory response in the endometrium and establish the rationale for the exploitation of innovative therapeutic targets for endometriosis.


Asunto(s)
Endometriosis , Humanos , Femenino , Masculino , Especies Reactivas de Oxígeno , Esfingosina , Esfingolípidos
2.
Reprod Biomed Online ; 45(1): 15-18, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35562234

RESUMEN

RESEARCH QUESTION: Is sphingosine 1-phosphate (S1P) pathway involved in the process of fibrosis in adenomyosis? DESIGN: RNA was extracted from paraffin-embedded slices collected from the ectopic endometrium of patients with nodular adenomyosis (n = 27) and eutopic endometrium of healthy controls women (n = 29). Expression of genes involved in the metabolism and signalling of S1P, and actin-alpha-2 smooth muscle, encoded by ACTA2 gene, a gene involved in fibrogenesis, was evaluated by real-time polymerase chain reaction analysis. RESULTS: In adenomyotic samples, the expression of sphingosine kinase 1 (SPHK1), the enzyme responsible for the synthesis of S1P, and of S1P phosphatase 2 (SGPP2), the enzyme responsible for the conversion of S1P back to sphingosine, was lower (P = 0.0006; P = 0.0015), whereas that of calcium and integrin-binding protein 1, responsible for membrane translocation of SPHK1, was higher (P = 0.0001) compared with healthy controls. In S1P signalling, a higher expression of S1P receptor S1P3 (P = 0.001), and a lower expression of S1P2 (P = 0.0019) mRNA levels, were found compared with healthy endometrium. In adenomyotic nodules, a higher expression of ACTA2 mRNA levels were observed (P = 0.0001), which correlated with S1P3 levels (P = 0.0138). CONCLUSION: Present data show a profound dysregulation of the S1P signalling axis in adenomyosis. This study also highlights that the bioactive sphingolipid might be involved in the fibrotic tract of the disease, correlated with the expression of ACTA2, suggesting its role as novel potential biomarker of adenomyosis.


Asunto(s)
Adenomiosis , Esfingosina , Adenomiosis/genética , Adenomiosis/metabolismo , Femenino , Fibrosis , Humanos , Lisofosfolípidos/genética , Lisofosfolípidos/metabolismo , ARN Mensajero , Esfingosina/análogos & derivados , Esfingosina/genética , Esfingosina/metabolismo
3.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36362089

RESUMEN

Ovarian cancer recurrence is frequent and associated with chemoresistance, leading to extremely poor prognosis. Herein, we explored the potential anti-cancer effect of a series of highly charged Ru(II)-polypyridyl complexes as photosensitizers in photodynamic therapy (PDT), which were able to efficiently sensitize the formation of singlet oxygen upon irradiation (Ru12+ and Ru22+) and to produce reactive oxygen species (ROS) in their corresponding dinuclear metal complexes with the Fenton active Cu(II) ion/s ([CuRu1]4+ and [Cu2Ru2]6+). Their cytotoxic and anti-tumor effects were evaluated on human ovarian cancer A2780 cells both in the absence or presence of photoirradiation, respectively. All the compounds tested were well tolerated under dark conditions, whereas they switched to exert anti-tumor activity following photoirradiation. The specific effect was mediated by the onset of programed cell death, but only in the case of Ru12+ and Ru22+ was preceded by the loss of mitochondrial membrane potential soon after photoactivation and ROS production, thus supporting the occurrence of apoptosis via type II photochemical reactions. Thus, Ru(II)-polypyridyl-based photosensitizers represent challenging tools to be further investigated in the identification of new therapeutic approaches to overcome the innate chemoresistance to platinum derivatives of some ovarian epithelial cancers and to find innovative drugs for recurrent ovarian cancer.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias Ováricas , Fotoquimioterapia , Rutenio , Humanos , Femenino , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Rutenio/farmacología , Rutenio/química , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Línea Celular Tumoral , Especies Reactivas de Oxígeno , Células HeLa , Neoplasias Ováricas/tratamiento farmacológico , Recurrencia Local de Neoplasia , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Antineoplásicos/farmacología , Antineoplásicos/química
4.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36232401

RESUMEN

The sphingosine 1-phosphate (S1P) and endocannabinoid (ECS) systems comprehend bioactive lipids widely involved in the regulation of similar biological processes. Interactions between S1P and ECS have not been so far investigated in skeletal muscle, where both systems are active. Here, we used murine C2C12 myoblasts to investigate the effects of S1P on ECS elements by qRT-PCR, Western blotting and UHPLC-MS. In addition, the modulation of the mitochondrial membrane potential (ΔΨm), by JC-1 and Mitotracker Red CMX-Ros fluorescent dyes, as well as levels of protein controlling mitochondrial function, along with the oxygen consumption were assessed, by Western blotting and respirometry, respectively, after cell treatment with methanandamide (mAEA) and in the presence of S1P or antagonists to endocannabinoid-binding receptors. S1P induced a significant increase in TRPV1 expression both at mRNA and protein level, while it reduced the protein content of CB2. A dose-dependent effect of mAEA on ΔΨm, mediated by TRPV1, was evidenced; in particular, low doses were responsible for increased ΔΨm, whereas a high dose negatively modulated ΔΨm and cell survival. Moreover, mAEA-induced hyperpolarization was counteracted by S1P. These findings open new dimension to S1P and endocannabinoids cross-talk in skeletal muscle, identifying TRPV1 as a pivotal target.


Asunto(s)
Endocannabinoides , Colorantes Fluorescentes , Animales , Ácidos Araquidónicos , Línea Celular , Endocannabinoides/metabolismo , Endocannabinoides/farmacología , Colorantes Fluorescentes/metabolismo , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Ratones , Mitocondrias/metabolismo , Mioblastos/metabolismo , Alcamidas Poliinsaturadas , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
5.
Int J Mol Sci ; 22(3)2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33535610

RESUMEN

Phosphatidic acid (PA) is a bioactive phospholipid capable of regulating key biological functions, including neutrophil respiratory burst, chemotaxis, or cell growth and differentiation. However, the mechanisms whereby PA exerts these actions are not completely understood. In this work, we show that PA stimulates myoblast proliferation, as determined by measuring the incorporation of [3H]thymidine into DNA and by staining the cells with crystal violet. PA induced the rapid phosphorylation of Akt and ERK1/2, and pretreatment of the cells with specific small interferin RNA (siRNA) to silence the genes encoding these kinases, or with selective pharmacologic inhibitors, blocked PA-stimulated myoblast proliferation. The mitogenic effects of PA were abolished by the preincubation of the myoblasts with pertussis toxin, a Gi protein inhibitor, suggesting the implication of Gi protein-coupled receptors in this action. Although some of the effects of PA have been associated with its possible conversion to lysoPA (LPA), treatment of the myoblasts with PA for up to 60 min did not produce any significant amount of LPA in these cells. Of interest, pharmacological blockade of the LPA receptors 1 and 2, or specific siRNA to silence the genes encoding these receptors, abolished PA-stimulated myoblast proliferation. Moreover, PA was able to compete with LPA for binding to LPA receptors, suggesting that PA can act as a ligand of LPA receptors. It can be concluded that PA stimulates myoblast proliferation through interaction with LPA1 and LPA2 receptors and the subsequent activation of the PI3K/Akt and MEK/ERK1-2 pathways, independently of LPA formation.


Asunto(s)
Mioblastos/metabolismo , Ácidos Fosfatidicos/química , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Quimiotaxis/efectos de los fármacos , ADN/metabolismo , Lisofosfolípidos/química , Lisofosfolípidos/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525436

RESUMEN

Skeletal muscle atrophy is characterized by a decrease in muscle mass causing reduced agility, increased fatigability and higher risk of bone fractures. Inflammatory cytokines, such as tumor necrosis factor-alpha (TNFα), are strong inducers of skeletal muscle atrophy. The bioactive sphingolipid sphingosine 1-phoshate (S1P) plays an important role in skeletal muscle biology. S1P, generated by the phosphorylation of sphingosine catalyzed by sphingosine kinase (SK1/2), exerts most of its actions through its specific receptors, S1P1-5. Here, we provide experimental evidence that TNFα induces atrophy and autophagy in skeletal muscle C2C12 myotubes, modulating the expression of specific markers and both active and passive membrane electrophysiological properties. NMR-metabolomics provided a clear picture of the deep remodelling of skeletal muscle fibre metabolism induced by TNFα challenge. The cytokine is responsible for the modulation of S1P signalling axis, upregulating mRNA levels of S1P2 and S1P3 and downregulating those of SK2. TNFα increases the phosphorylated form of SK1, readout of its activation. Interestingly, pharmacological inhibition of SK1 and specific antagonism of S1P3 prevented the increase in autophagy markers and the changes in the electrophysiological properties of C2C12 myotubes without affecting metabolic remodelling induced by the cytokine, highlighting the involvement of S1P signalling axis on TNFα-induced atrophy in skeletal muscle.


Asunto(s)
Lisofosfolípidos/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptores de Esfingosina-1-Fosfato/genética , Esfingosina/análogos & derivados , Factor de Necrosis Tumoral alfa/farmacología , Animales , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Humanos , Metabolómica/métodos , Ratones , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Mioblastos/metabolismo , Mioblastos/patología , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
7.
Biochim Biophys Acta Mol Cell Res ; 1864(5): 814-824, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28188805

RESUMEN

Hearing loss affects millions of people in the world. In mammals the auditory system comprises diverse cell types which are terminally differentiated and with no regenerative potential. There is a tremendous research interest aimed at identifying cell therapy based solutions or pharmacological approaches that could be applied therapeutically alongside auditory devices to prevent hair cell and neuron loss. Sphingosine 1-phosphate (S1P) is a pleiotropic bioactive sphingolipid that plays key role in the regulation of many physiological and pathological functions. S1P is intracellularly produced by sphingosine kinase (SK) 1 and SK2 and exerts many of its action consequently to its ligation to S1P specific receptors (S1PR), S1P1-5. In this study, murine auditory neuroblasts named US/VOT-N33 have been used as progenitors of neurons of the spiral ganglion. We demonstrated that the fibroblast growth factor 2 (FGF2)-induced proliferative action was dependent on SK1, SK2 as well as S1P1 and S1P2. Moreover, the pro-survival effect of FGF2 from apoptotic cell death induced by staurosporine treatment was dependent on SK but not on S1PR. Additionally, ERK1/2 and Akt signaling pathways were found to mediate the mitogenic and survival action of FGF2, respectively. Taken together, these findings demonstrate a crucial role for S1P signaling axis in the proliferation and the survival of otic vesicle neuroprogenitors, highlighting the identification of possible novel therapeutical approaches to prevent neuronal degeneration during hearing loss.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Lisofosfolípidos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Esfingosina/análogos & derivados , Ganglio Espiral de la Cóclea/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cóclea/citología , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Ratones , Células-Madre Neurales/fisiología , Transducción de Señal/efectos de los fármacos , Esfingosina/metabolismo , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/metabolismo
8.
Int J Mol Sci ; 19(1)2018 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-29300303

RESUMEN

Sphingolipids are not only crucial for membrane architecture but act as critical regulators of cell functions. The bioactive sphingolipid ceramide 1-phosphate (C1P), generated by the action of ceramide kinase, has been reported to stimulate cell proliferation, cell migration and to regulate inflammatory responses via activation of different signaling pathways. We have previously shown that skeletal muscle is a tissue target for C1P since the phosphosphingolipid plays a positive role in myoblast proliferation implying a role in muscle regeneration. Skeletal muscle displays strong capacity of regeneration thanks to the presence of quiescent adult stem cells called satellite cells that upon trauma enter into the cell cycle and start proliferating. However, at present, the exact molecular mechanism by which C1P triggers its mitogenic effect in myoblasts is lacking. Here, we report for the first time that C1P stimulates C2C12 myoblast proliferation via lysophosphatidic acid (LPA) signaling axis. Indeed, C1P subsequently to phospholipase A2 activation leads to LPA1 and LPA3 engagement, which in turn drive Akt (protein kinase B) and ERK1/2 (extracellular signal-regulated kinases 1/2) activation, thus stimulating DNA synthesis. The present findings shed new light on the key role of bioactive sphingolipids in skeletal muscle and provide further support to the notion that these pleiotropic molecules might be useful therapeutic targets for skeletal muscle regeneration.


Asunto(s)
Ceramidas/farmacología , Lisofosfolípidos/metabolismo , Mioblastos/citología , Transducción de Señal , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Ratones , Mitógenos/farmacología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Fosfolipasas A2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Am J Physiol Cell Physiol ; 313(1): C54-C67, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28446426

RESUMEN

We investigated the effects of S1P3 deficiency on the age-related atrophy, decline in force, and regenerative capacity of soleus muscle from 23-mo-old male (old) mice. Compared with muscle from 5-mo-old (adult) mice, soleus mass and muscle fiber cross-sectional area (CSA) in old wild-type mice were reduced by ~26% and 24%, respectively. By contrast, the mass and fiber CSA of soleus muscle in old S1P3-null mice were comparable to those of adult muscle. Moreover, in soleus muscle of wild-type mice, twitch and tetanic tensions diminished from adulthood to old age. A slowing of contractile properties was also observed in soleus from old wild-type mice. In S1P3-null mice, neither force nor the contractile properties of soleus changed during aging. We also evaluated the regenerative capacity of soleus in old S1P3-null mice by stimulating muscle regeneration through myotoxic injury. After 10 days of regeneration, the mean fiber CSA of soleus in old wild-type mice was significantly smaller (-28%) compared with that of regenerated muscle in adult mice. On the contrary, the mean fiber CSA of regenerated soleus in old S1P3-null mice was similar to that of muscle in adult mice. We conclude that in the absence of S1P3, soleus muscle is protected from the decrease in muscle mass and force, and the attenuation of regenerative capacity, all of which are typical characteristics of aging.


Asunto(s)
Envejecimiento/genética , Músculo Esquelético/metabolismo , Receptores de Lisoesfingolípidos/genética , Sarcopenia/genética , Envejecimiento/metabolismo , Animales , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Contracción Muscular/fisiología , Fibras Musculares de Contracción Lenta/metabolismo , Fibras Musculares de Contracción Lenta/patología , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Receptores de Lisoesfingolípidos/deficiencia , Regeneración/fisiología , Sarcopenia/metabolismo , Sarcopenia/fisiopatología , Receptores de Esfingosina-1-Fosfato
10.
Biochim Biophys Acta ; 1860(3): 527-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26723170

RESUMEN

BACKGROUND: In-cell NMR is a powerful technique to investigate proteins in living human cells at atomic resolution. Ideally, when studying functional processes involving protein-protein interactions by NMR, only one partner should be isotopically labeled. Here we show that constitutive and transient protein expression can be combined with protein silencing to obtain selective protein labeling in human cells. METHODS: We established a human cell line stably overexpressing the copper binding protein HAH1. A second protein (human superoxide dismutase 1, SOD1) was overexpressed by transient transfection and isotopically labeled. A silencing vector containing shRNA sequences against the HAH1 gene was used to decrease the rate of HAH1 synthesis during the expression of SOD1. The levels of HAH1 mRNA and protein were measured as a function of time following transfection by RT-PCR and Western Blot, and the final cell samples were analyzed by in-cell NMR. RESULTS: SOD1 was ectopically expressed and labeled in a time window during which HAH1 biosynthesis was strongly decreased by shRNA, thus preventing its labeling. In-cell NMR spectra confirmed that, while both proteins were present, only SOD1 was selectively labeled and could be detected by (1)H-(15)N heteronuclear NMR. CONCLUSIONS AND GENERAL SIGNIFICANCE: We showed that controlling protein expression by specifically silencing a stably expressed protein is a useful strategy to obtain selective isotope labeling of only one protein. This approach relies on established techniques thus permitting the investigation of protein-protein interactions by NMR in human cells.


Asunto(s)
Espectroscopía de Resonancia Magnética , Metalochaperonas/genética , Proteínas Transportadoras de Cobre , Células HEK293 , Humanos , Chaperonas Moleculares , Isótopos de Nitrógeno , ARN Interferente Pequeño/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
11.
Biochim Biophys Acta ; 1851(2): 194-202, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25457224

RESUMEN

The matricellular protein connective tissue growth factor (CTGF/CCN2) is recognized as key player in the onset of fibrosis in various tissues, including skeletal muscle. In many circumstances, CTGF has been shown to be induced by transforming growth factor beta (TGFß) and accounting, at least in part, for its biological action. In this study it was verified that in cultured myoblasts CTGF/CCN2 causes their transdifferentiation into myofibroblasts by up-regulating the expression of fibrosis marker proteins α-smooth muscle actin and transgelin. Interestingly, it was also found that the profibrotic effect exerted by CTGF/CCN2 was mediated by the sphingosine kinase (SK)-1/S1P3 signaling axis specifically induced by the treatment with the profibrotic cue. Following CTGF/CCN2-induced up-regulation, S1P3 became the S1P receptor subtype expressed at the highest degree, at least at mRNA level, and was thus capable of readdressing the sphingosine 1-phosphate signaling towards fibrosis rather than myogenic differentiation. Another interesting finding is that CTGF/CCN2 silencing prevented the TGFß-dependent up-regulation of SK1/S1P3 signaling axis and strongly reduced the profibrotic effect exerted by TGFß, pointing at a crucial role of endogenous CTGF/CCN2 generated following TGFß challenge in the transmission of at least part of its profibrotic effect. These results provide new insights into the molecular mechanism by which CTGF/CCN2 drives its biological action and strengthen the concept that SK1/S1P3 axis plays a critical role in the onset of fibrotic cell phenotype.


Asunto(s)
Transdiferenciación Celular , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Mioblastos Esqueléticos/enzimología , Miofibroblastos/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Relación Dosis-Respuesta a Droga , Fibrosis , Ratones , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Proteínas Recombinantes/farmacología , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
12.
Hemoglobin ; 39(5): 366-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26193974

RESUMEN

A 4-year-old male child of Caucasian ethnicity was investigated for moderate hemolytic and non immune-mediated anemia. The presence of splenomegaly and the elevation of Hb A(2) and Hb F and the exclusion of a defect of protein of red blood cell (RBC) membranes defined a clinical picture of ß-thalassemia intermedia (ß-TI). The molecular analysis showed a heterozygous IVS-II-1 (HBB: c.315G > A) mutation on the ß-globin gene, in the absence of extra α-globin genes or unstable hemoglobin (Hb) chains.


Asunto(s)
Mutación , Globinas beta/genética , Talasemia beta/diagnóstico , Talasemia beta/genética , Preescolar , Índices de Eritrocitos , Humanos , Intrones , Masculino , Fenotipo
13.
Biochim Biophys Acta ; 1831(1): 176-84, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22877992

RESUMEN

Lysophospholipids are bioactive molecules that are implicated in the control of fundamental biological processes such as proliferation, differentiation, survival and motility in different cell types. Here we review the role of sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) in the regulation of skeletal muscle biology. Indeed, a wealth of experimental data indicate that these molecules are crucial players in the skeletal muscle regeneration process, acting by controllers of activation, proliferation and differentiation not only of muscle-resident satellite cells but also of mesenchymal progenitors that originate outside the skeletal muscle. Moreover, S1P and LPA are clearly involved in the regulation of skeletal muscle metabolism, muscle adaptation to different physiological needs and resistance to muscle fatigue. Notably, studies accomplished so far, have highlighted the complexity of S1P and LPA signaling in skeletal muscle cells that appears to be further complicated by their close dependence on functional cross-talks with growth factors, hormones and cytokines. Our increasing understanding of bioactive lipid signaling can individuate novel molecular targets aimed at enhancing skeletal muscle regeneration and reducing the fibrotic process that impairs full functional recovery of the tissue during aging, after a trauma or skeletal muscle diseases. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Asunto(s)
Lisofosfolípidos/metabolismo , Células Musculares/citología , Células Musculares/metabolismo , Músculo Esquelético/citología , Esfingosina/análogos & derivados , Animales , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Esfingosina/metabolismo
14.
FASEB J ; 27(11): 4532-46, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23913862

RESUMEN

In view of its multiple detrimental effects, transforming growth factor ß1 (TGFß1) is recognized as critical negative regulator of skeletal muscle repair. Apoptosis of skeletal muscle precursor cells driven by TGFß1 contributes to the negative role exerted by the cytokine in tissue repair, although the underlying molecular mechanisms are still elusive. Herein we report the identification of a new signaling pathway, relying on Rho kinase-2 stimulation, subsequent to SMAD-dependent S1P4 up-regulation and transactivation via sphingosine kinase (SK)-2, that accounts for TGFß1-induced apoptosis in cultured myoblasts. S1P4-specific gene silencing reduced by almost 50% activation of caspase-3 and poly-ADP ribosyl transferase cleavage elicited by TGFß1. Moreover, the selective S1P4 antagonist CYM50358 also reduced the TGFß1 proapoptotic effects. By employing pharmacological and molecular biological approaches, the involvement of SK2 and ROCK2 in the transmission of the TGFß1 apoptotic action was also demonstrated. These results reinforce the notion that the SK/S1P axis plays a fundamental role in TGFß1 mode of action in skeletal muscle cells and, by disclosing a novel mechanism by which TGFß1 exerts its harmful action, pinpoint new molecular targets that in principle could be beneficial in the treatment of several skeletal muscle disorders or aging-dependent muscle atrophy.


Asunto(s)
Apoptosis , Mioblastos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología , Quinasas Asociadas a rho/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Ratones , Mioblastos/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Esfingosina-1-Fosfato
15.
Fertil Steril ; 121(4): 631-641, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38072366

RESUMEN

OBJECTIVE: To study the molecular mechanisms responsible for fibrosis in endometriosis by investigating whether the protein expression levels of sphingosine-1-phosphate receptor 3 (S1PR3), one of the five specific receptors of the bioactive sphingolipid sphingosine-1-phosphate (S1P), correlate with fibrosis extent in endometriotic lesions and which are the cellular mechanisms involved in this process. DESIGN: Case-control laboratory study and cultured endometriotic cells. SETTING: University research institute and university hospital. PATIENT(S): A total of 33 women, with and without endometriosis, were included in the study. INTERVENTIONS(S): Endometriotic lesions were obtained from women with endometriosis (ovarian endometrioma, n = 8; deep infiltrating endometriosis, n = 15; [urological n = 5, gastrointestinal n = 6, and posterior n = 4]) and control endometrium from healthy women, n = 10, by means of laparoscopic and hysteroscopic surgery. The expression of S1PR3 was evaluated using immunohistochemistry and the extent of fibrosis was assessed using Masson's trichrome staining. Human-cultured epithelial endometriotic 12Z cells were used to evaluate the mechanisms involved in the profibrotic effect of S1PR3 activation. MAIN OUTCOME MEASURE(S): The expression of S1PR3 in endometriotic lesions is positively correlated with endometriosis-associated fibrosis. In addition, S1P induced epithelial-mesenchymal transition (EMT) and fibrosis in epithelial endometriotic cells. Using RNA interference and pharmacological approaches, the profibrotic effect of S1P was shown to rely on S1PR3, thus unveiling the molecular mechanism implicated in the profibrotic action of the bioactive sphingolipid. RESULT(S): The protein expression levels of S1PR3 were significantly augmented in the glandular sections of endometrioma and deep infiltrating endometriosis of different localizations with respect to the control endometrium and positively correlated with the extent of fibrosis. Sphingosine-1-phosphate was shown to have a crucial role in the onset of fibrosis in epithelial endometriotic cells, stimulating the expression of EMT and fibrotic markers. Genetic approaches have highlighted that S1PR3 mediates the fibrotic effect of S1P. Downstream of S1PR3, ezrin and extracellular-signal-regulated kinases 1 and 2 signaling were found to be critically implicated in the EMT and fibrosis elicited by S1P. CONCLUSION(S): Sphingosine-1-phosphate receptor 3 may represent a possible innovative pharmacological target for endometriosis.


Asunto(s)
Endometriosis , Lisofosfolípidos , Esfingosina/análogos & derivados , Humanos , Femenino , Receptores de Esfingosina-1-Fosfato , Endometriosis/complicaciones , Endometriosis/genética , Endometriosis/metabolismo , Fibrosis , Esfingolípidos
16.
FEBS J ; 291(8): 1744-1758, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38287231

RESUMEN

Endometriosis is a chronic gynecological syndrome characterized by endometrial cell invasion of the extra-uterine milieu, pelvic pain and infertility. Treatment relies on either symptomatic drugs or hormonal therapies, even though the mechanism involved in the onset of endometriosis is yet to be elucidated. The signaling of sphingolipid sphingosine 1-phosphate (S1P) is profoundly dysregulated in endometriosis. Indeed, sphingosine kinase (SK)1, one of the two isoenzymes responsible for S1P biosynthesis, and S1P1, S1P3 and S1P5, three of its five specific receptors, are more highly expressed in endometriotic lesions compared to healthy endometrium. Recently, missense coding variants of the gene encoding the receptor 1 for neuropeptide S (NPS) have been robustly associated with endometriosis in humans. This study aimed to characterize the biological effect of NPS in endometriotic epithelial cells and the possible involvement of the S1P signaling axis in its action. NPS was found to potently induce cell invasion and actin cytoskeletal remodeling. Of note, the NPS-induced invasive phenotype was dependent on SK1 and SK2 as well as on S1P1 and S1P3, given that the biological action of the neuropeptide was fully prevented when one of the two biosynthetic enzymes or one of the two selective receptors was inhibited or silenced. Furthermore, the RhoA/Rho kinase pathway, downstream to S1P receptor signaling, was found to be critically implicated in invasion and cytoskeletal remodeling elicited by NPS. These findings provide new information to the understanding of the molecular mechanisms implicated in endometriosis pathogenesis, establishing the rationale for non-hormonal therapeutic targets for its treatment.


Asunto(s)
Endometriosis , Receptores de Lisoesfingolípidos , Esfingosina , Femenino , Humanos , Endometriosis/genética , Lisofosfolípidos/metabolismo , Fenotipo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Esfingosina/análogos & derivados
17.
Biochim Biophys Acta ; 1823(2): 439-50, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22178384

RESUMEN

Satellite cells are resident stem cells of skeletal muscle; they are normally quiescent but upon post-trauma activation start to proliferate and fuse with damaged fibers contributing to muscle regeneration. In this study the effect of the bioactive sphingolipid sphingosine 1-phosphate (S1P) on the proliferative and migratory response of murine satellite cells has been examined. S1P was found to stimulate labeled thymidine incorporation in a phosphatidylinositol 3-kinase-dependent manner. Moreover, by employing selective S1P receptor agonists and antagonists and silencing individual S1P receptors, the mitogenic action of S1P in satellite cells was shown to depend on S1P2 and S1P3. Notably, by using different experimental approaches S1P was found to positively influence satellite cell migration, necessary for their recruitment at the site of muscle damage. Interestingly, the specific silencing of individual S1P receptor subtypes demonstrated the pivotal role of S1P1 and S1P4 in mediating the S1P migratory effect. This latter result demonstrates for the first time that S1P4 receptor has a role in skeletal muscle cells, supporting the notion that this receptor subtype plays a biological action broader than that so far identified in lymphoid tissue. On the contrary, S1P2 was found to negatively regulate cell migration. Collectively, these results are in favour of an important function of S1P in satellite cell biology that could in principle be exploited as novel pharmacological target for improving skeletal muscle regeneration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Lisofosfolípidos/farmacología , Isoformas de Proteínas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/fisiología , Esfingosina/análogos & derivados , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/genética , Células Satélite del Músculo Esquelético/citología , Transducción de Señal/fisiología , Esfingosina/metabolismo , Esfingosina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Handb Exp Pharmacol ; (216): 457-67, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23563671

RESUMEN

Studies performed in the last fifteen years have clearly established that the bioactive sphingolipid sphingosine 1-phosphate (S1P) affects various different biological properties of myogenic precursor cells as well as physiological features of adult skeletal muscle. Noticeably, in myogenic precursor cells multiple growth factors and cytokines cross-communicate with S1P axis and the engagement of distinct S1P receptor subtypes appears to be crucially implicated in transmitting specific biological effects. This paper summarizes current research findings and discloses the potential for new therapeutics designed to alter S1P signaling with the aim of improving skeletal muscle repair.


Asunto(s)
Lisofosfolípidos/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Citocinas/metabolismo , Hormonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Mioblastos Esqueléticos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/metabolismo
19.
Reprod Sci ; 30(5): 1453-1461, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36289173

RESUMEN

Endometriosis is a chronic inflammatory condition affecting women of reproductive age. A relevant feature of endometriosis is the presence of fibrotic tissue inside and around the lesions, thus contributing to the classic endometriosis-related symptoms, pain, and infertility. The molecular mechanisms responsible for the development of fibrosis in endometriosis are not yet defined. The present review aimed to examine the biological mechanisms and signalling pathways involved in fibrogenesis of endometriotic lesions, highlighting the difference between deep infiltrating and ovarian endometriosis. The main cell types involved in the development of fibrosis are platelets, myofibroblasts, macrophages, and sensory nerve fibers. Members of the transforming growth factor (TGF) -ß family, as well as the receptor Notch, or the bioactive sphingolipid sphingosine 1-phosphate (S1P), play a role in the development of tissue fibrosis, resulting in their metabolism and/or their signalling pathways altered in endometriotic lesions. It is relevant the knowledge of the molecular mechanisms that guide and support fibrosis in endometriosis, to identify new drug targets and provide new therapeutic approaches to patients.


Asunto(s)
Endometriosis , Humanos , Femenino , Endometriosis/metabolismo , Transducción de Señal/fisiología , Miofibroblastos/metabolismo , Plaquetas , Factor de Crecimiento Transformador beta , Fibrosis
20.
FEBS J ; 290(1): 112-133, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35851748

RESUMEN

Soluble oligomers arising from the aggregation of the amyloid beta peptide (Aß) have been identified as the main pathogenic agents in Alzheimer's disease (AD). Prefibrillar oligomers of the 42-residue form of Aß (Aß42 O) show membrane-binding capacity and trigger the disruption of Ca2+ homeostasis, a causative event in neuron degeneration. Since bioactive lipids have been recently proposed as potent protective agents against Aß toxicity, we investigated the involvement of sphingosine 1-phosphate (S1P) signalling pathway in Ca2+ homeostasis in living neurons exposed to Aß42 O. We show that both exogenous and endogenous S1P rescued neuronal Ca2+ dyshomeostasis induced by toxic Aß42 O in primary rat cortical neurons and human neuroblastoma SH-SY5Y cells. Further analysis revealed a strong neuroprotective effect of S1P1 and S1P4 receptors, and to a lower extent of S1P3 and S1P5 receptors, which activate the Gi -dependent signalling pathways, thus resulting in the endocytic internalization of the extrasynaptic GluN2B-containing N-methyl-D-aspartate receptors (NMDARs). Notably, the S1P beneficial effect can be sustained over time by sphingosine kinase-1 overexpression, thus counteracting the down-regulation of the S1P signalling induced by Aß42 O. Our findings disclose underlying mechanisms of S1P neuronal protection against harmful Aß42 O, suggesting that S1P and its signalling axis can be considered promising targets for therapeutic approaches for AD.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Ratas , Humanos , Animales , Receptores de N-Metil-D-Aspartato/genética , Péptidos beta-Amiloides/metabolismo , Neuroblastoma/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA