Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell ; 69(4): 594-609.e8, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452639

RESUMEN

Accumulating evidence indicates that the MDM2 oncoprotein promotes tumorigenesis beyond its canonical negative effects on the p53 tumor suppressor, but these p53-independent functions remain poorly understood. Here, we show that a fraction of endogenous MDM2 is actively imported in mitochondria to control respiration and mitochondrial dynamics independently of p53. Mitochondrial MDM2 represses the transcription of NADH-dehydrogenase 6 (MT-ND6) in vitro and in vivo, impinging on respiratory complex I activity and enhancing mitochondrial ROS production. Recruitment of MDM2 to mitochondria increases during oxidative stress and hypoxia. Accordingly, mice lacking MDM2 in skeletal muscles exhibit higher MT-ND6 levels, enhanced complex I activity, and increased muscular endurance in mild hypoxic conditions. Furthermore, increased mitochondrial MDM2 levels enhance the migratory and invasive properties of cancer cells. Collectively, these data uncover a previously unsuspected function of the MDM2 oncoprotein in mitochondria that play critical roles in skeletal muscle physiology and may contribute to tumor progression.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Transformación Celular Neoplásica/patología , Complejo I de Transporte de Electrón/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitocondrias/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Complejo I de Transporte de Electrón/genética , Genoma Mitocondrial , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Invasividad Neoplásica , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-mdm2/genética , Transducción de Señal , Transcripción Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Proc Natl Acad Sci U S A ; 120(25): e2219431120, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307458

RESUMEN

Gut microbiota imbalance (dysbiosis) is increasingly associated with pathological conditions, both within and outside the gastrointestinal tract. Intestinal Paneth cells are considered to be guardians of the gut microbiota, but the events linking Paneth cell dysfunction with dysbiosis remain unclear. We report a three-step mechanism for dysbiosis initiation. Initial alterations in Paneth cells, as frequently observed in obese and inflammatorybowel diseases patients, cause a mild remodeling of microbiota, with amplification of succinate-producing species. SucnR1-dependent activation of epithelial tuft cells triggers a type 2 immune response that, in turn, aggravates the Paneth cell defaults, promoting dysbiosis and chronic inflammation. We thus reveal a function of tuft cells in promoting dysbiosis following Paneth cell deficiency and an unappreciated essential role of Paneth cells in maintaining a balanced microbiota to prevent inappropriate activation of tuft cells and deleterious dysbiosis. This succinate-tuft cell inflammation circuit may also contribute to the chronic dysbiosis observed in patients.


Asunto(s)
Disbiosis , Membrana Mucosa , Humanos , Inflamación , Células de Paneth , Succinatos , Ácido Succínico
3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658388

RESUMEN

Ki-67 is a nuclear protein that is expressed in all proliferating vertebrate cells. Here, we demonstrate that, although Ki-67 is not required for cell proliferation, its genetic ablation inhibits each step of tumor initiation, growth, and metastasis. Mice lacking Ki-67 are resistant to chemical or genetic induction of intestinal tumorigenesis. In established cancer cells, Ki-67 knockout causes global transcriptome remodeling that alters the epithelial-mesenchymal balance and suppresses stem cell characteristics. When grafted into mice, tumor growth is slowed, and metastasis is abrogated, despite normal cell proliferation rates. Yet, Ki-67 loss also down-regulates major histocompatibility complex class I antigen presentation and, in the 4T1 syngeneic model of mammary carcinoma, leads to an immune-suppressive environment that prevents the early phase of tumor regression. Finally, genes involved in xenobiotic metabolism are down-regulated, and cells are sensitized to various drug classes. Our results suggest that Ki-67 enables transcriptional programs required for cellular adaptation to the environment. This facilitates multiple steps of carcinogenesis and drug resistance, yet may render cancer cells more susceptible to antitumor immune responses.


Asunto(s)
Carcinogénesis/metabolismo , Regulación Neoplásica de la Expresión Génica , Antígeno Ki-67/metabolismo , Neoplasias Mamarias Animales/metabolismo , Proteínas de Neoplasias/metabolismo , Transcripción Genética , Animales , Carcinogénesis/genética , Femenino , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Antígeno Ki-67/genética , Neoplasias Mamarias Animales/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética
4.
Mol Hum Reprod ; 27(5)2021 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-33851217

RESUMEN

Adenomyosis is characterised by epithelial gland and mesenchymal stroma invasion of the uterine myometrium. Adenomyosis is an oestrogen-dependent gynaecological disease in which a number of factors, such as inflammatory molecules, prostaglandins (PGs), angiogenic factors, cell proliferation and extracellular matrix remodelling proteins, also play a role as key disease mediators. In this study, we used mice lacking both lipocalin and hematopoietic-PG D synthase (L- and H-Pgds) genes in which PGD2 is not produced to elucidate PGD2 roles in the uterus. Gene expression studied by real-time PCR and hormone dosages performed by ELISA or liquid chromatography tandem mass spectroscopy in mouse uterus samples showed that components of the PGD2 signalling pathway, both PGDS and PGD2-receptors, are expressed in the mouse endometrium throughout the oestrus cycle with some differences among uterine compartments. We showed that PGE2 production and the steroidogenic pathway are dysregulated in the absence of PGD2. Histological analysis of L/H-Pgds-/- uteri, and immunohistochemistry and immunofluorescence analyses of proliferation (Ki67), endothelial cell (CD31), epithelial cell (pan-cytokeratin), myofibroblast (α-SMA) and mesenchymal cell (vimentin) markers, identify that 6-month-old L/H-Pgds-/- animals developed adenomyotic lesions, and that disease severity increased with age. In conclusion, this study suggests that the PGD2 pathway has major roles in the uterus by protecting the endometrium against adenomyosis development. Additional experiments, using for instance transcriptomic approaches, are necessary to fully determine the molecular mechanisms that lead to adenomyosis in L/H-Pgds-/- mice and to confirm whether this strain is an appropriate model for studying the human disease.


Asunto(s)
Adenomiosis/metabolismo , Prostaglandina D2/fisiología , Transducción de Señal , Útero/metabolismo , Animales , Dinoprostona/metabolismo , Femenino , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Ratones , Prostaglandina D2/genética , Prostaglandina D2/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Esteroides/biosíntesis , Útero/fisiología
5.
Eur J Nutr ; 60(4): 2013-2027, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32989473

RESUMEN

PURPOSE: Fatty acid esters of hydroxy fatty acids (FAHFAs) are a large family of endogenous bioactive lipids. To date, most of the studied FAHFAs are branched regioisomers of Palmitic Acid Hydroxyl Stearic Acid (PAHSA) that were reported to possess anti-diabetic and anti-inflammatory activity in humans and rodents. Recently, we have demonstrated that 9-PAHPA or 9-OAHPA intake increased basal metabolism and enhanced insulin sensitivity in healthy control diet-fed mice but induced liver damage in some mice. The present work aims to explore whether a long-term intake of 9-PAHPA or 9-OAHPA may have similar effects in obesogenic diet-fed mice. METHODS: C57Bl6 mice were fed with a control or high fat-high sugar (HFHS) diets for 12 weeks. The HFHS diet was supplemented or not with 9-PAHPA or 9-OAHPA. Whole-body metabolism was explored. Glucose and lipid metabolism as well as mitochondrial activity and oxidative stress status were analyzed. RESULTS: As expected, the intake of HFHS diet led to obesity and lower insulin sensitivity with minor effects on liver parameters. The long-term intake of 9-PAHPA or 9-OAHPA modulated favorably the basal metabolism and improved insulin sensitivity as measured by insulin tolerance test. On the contrary to what we have reported previously in healthy mice, no marked effect for these FAHFAs was observed on liver metabolism of obese diabetic mice. CONCLUSION: This study indicates that both 9-PAHPA and 9-OAHPA may have interesting insulin-sensitizing effects in obese mice with lower insulin sensitivity.


Asunto(s)
Diabetes Mellitus Experimental , Resistencia a la Insulina , Animales , Metabolismo Basal , Diabetes Mellitus Experimental/metabolismo , Insulina/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL
6.
Parasitology ; 147(3): 255-262, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31727197

RESUMEN

Although there is a plethora of cancer associated-factors that can ultimately culminate in death (cachexia, organ impairment, metastases, opportunistic infections, etc.), the focal element of every terminal malignancy is the failure of our natural defences to control unlimited cell proliferation. The reasons why our defences apparently lack efficiency is a complex question, potentially indicating that, under Darwinian terms, solutions other than preventing cancer progression are also important contributors. In analogy with host-parasite systems, we propose to call this latter option 'tolerance' to cancer. Here, we argue that the ubiquity of oncogenic processes among metazoans is at least partially attributable to both the limitations of resistance mechanisms and to the evolution of tolerance to cancer. Deciphering the ecological contexts of alternative responses to the cancer burden is not a semantic question, but rather a focal point in understanding the evolutionary ecology of host-tumour relationships, the evolution of our defences, as well as why and when certain cancers are likely to be detrimental for survival.


Asunto(s)
Antibiosis , Evolución Biológica , Interacciones Huésped-Parásitos/inmunología , Tolerancia Inmunológica , Neoplasias/inmunología , Animales
7.
Mol Cell ; 47(4): 523-34, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22771120

RESUMEN

We generated knockout mice for MCM8 and MCM9 and show that deficiency for these genes impairs homologous recombination (HR)-mediated DNA repair during gametogenesis and somatic cells cycles. MCM8(-/-) mice are sterile because spermatocytes are blocked in meiotic prophase I, and females have only arrested primary follicles and frequently develop ovarian tumors. MCM9(-/-) females also are sterile as ovaries are completely devoid of oocytes. In contrast, MCM9(-/-) testes produce spermatozoa, albeit in much reduced quantity. Mcm8(-/-) and Mcm9(-/-) embryonic fibroblasts show growth defects and chromosomal damage and cannot overcome a transient inhibition of replication fork progression. In these cells, chromatin recruitment of HR factors like Rad51 and RPA is impaired and HR strongly reduced. We further demonstrate that MCM8 and MCM9 form a complex and that they coregulate their stability. Our work uncovers essential functions of MCM8 and MCM9 in HR-mediated DSB repair during gametogenesis, replication fork maintenance, and DNA repair.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Proteínas de Unión al ADN/deficiencia , Gametogénesis/genética , Inestabilidad Genómica , Recombinación Homóloga/genética , Animales , Proteínas de Ciclo Celular/genética , Cromatina/genética , Reparación del ADN , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/metabolismo , Células Germinativas/metabolismo , Masculino , Profase Meiótica I/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Mantenimiento de Minicromosoma , Ovario/metabolismo , Espermatocitos/metabolismo
8.
Proc Natl Acad Sci U S A ; 113(39): 11004-9, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27621431

RESUMEN

The multifunctional protein E4 transcription factor 1 (E4F1) is an essential regulator of epidermal stem cell (ESC) maintenance. Here, we found that E4F1 transcriptionally regulates a metabolic program involved in pyruvate metabolism that is required to maintain skin homeostasis. E4F1 deficiency in basal keratinocytes resulted in deregulated expression of dihydrolipoamide acetyltransferase (Dlat), a gene encoding the E2 subunit of the mitochondrial pyruvate dehydrogenase (PDH) complex. Accordingly, E4f1 knock-out (KO) keratinocytes exhibited impaired PDH activity and a redirection of the glycolytic flux toward lactate production. The metabolic reprogramming of E4f1 KO keratinocytes associated with remodeling of their microenvironment and alterations of the basement membrane, led to ESC mislocalization and exhaustion of the ESC pool. ShRNA-mediated depletion of Dlat in primary keratinocytes recapitulated defects observed upon E4f1 inactivation, including increased lactate secretion, enhanced activity of extracellular matrix remodeling enzymes, and impaired clonogenic potential. Altogether, our data reveal a central role for Dlat in the metabolic program regulated by E4F1 in basal keratinocytes and illustrate the importance of PDH activity in skin homeostasis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/metabolismo , Homeostasis , Proteínas Mitocondriales/metabolismo , Piel/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Membrana Basal/metabolismo , Adhesión Celular , Células Cultivadas , Microambiente Celular , Proteínas de Unión al ADN/deficiencia , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/genética , Células Epidérmicas , Epidermis/metabolismo , Regulación de la Expresión Génica , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones Noqueados , Proteínas Mitocondriales/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Piruvatos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras , Células Madre/metabolismo , Factores de Transcripción/deficiencia , Ubiquitina-Proteína Ligasas
9.
Proc Natl Acad Sci U S A ; 113(39): 10998-1003, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27621446

RESUMEN

The mitochondrial pyruvate dehydrogenase (PDH) complex (PDC) acts as a central metabolic node that mediates pyruvate oxidation and fuels the tricarboxylic acid cycle to meet energy demand. Here, we reveal another level of regulation of the pyruvate oxidation pathway in mammals implicating the E4 transcription factor 1 (E4F1). E4F1 controls a set of four genes [dihydrolipoamide acetlytransferase (Dlat), dihydrolipoyl dehydrogenase (Dld), mitochondrial pyruvate carrier 1 (Mpc1), and solute carrier family 25 member 19 (Slc25a19)] involved in pyruvate oxidation and reported to be individually mutated in human metabolic syndromes. E4F1 dysfunction results in 80% decrease of PDH activity and alterations of pyruvate metabolism. Genetic inactivation of murine E4f1 in striated muscles results in viable animals that show low muscle PDH activity, severe endurance defects, and chronic lactic acidemia, recapitulating some clinical symptoms described in PDC-deficient patients. These phenotypes were attenuated by pharmacological stimulation of PDH or by a ketogenic diet, two treatments used for PDH deficiencies. Taken together, these data identify E4F1 as a master regulator of the PDC.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Proteínas de Unión al ADN/deficiencia , Dieta Cetogénica , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Músculo Estriado/metabolismo , Fenotipo , Ácido Pirúvico/metabolismo , Proteínas Represoras , Factores de Transcripción/deficiencia , Ubiquitina-Proteína Ligasas
10.
J Neurosci ; 36(37): 9558-71, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27629708

RESUMEN

UNLABELLED: Although we are beginning to understand the late stage of neurodegenerative diseases, the molecular defects associated with the initiation of impaired cognition are poorly characterized. Here, we demonstrate that in the adult brain, the coxsackievirus and adenovirus receptor (CAR) is located on neuron projections, at the presynapse in mature neurons, and on the soma of immature neurons in the hippocampus. In a proinflammatory or diseased environment, CAR is lost from immature neurons in the hippocampus. Strikingly, in hippocampi of patients at early stages of late-onset Alzheimer's disease (AD), CAR levels are significantly reduced. Similarly, in triple-transgenic AD mice, CAR levels in hippocampi are low and further reduced after systemic inflammation. Genetic deletion of CAR from the mouse brain triggers deficits in adult neurogenesis and synapse homeostasis that lead to impaired hippocampal plasticity and cognitive deficits. We propose that post-translational CAR loss of function contributes to cognitive defects in healthy and diseased-primed brains. SIGNIFICANCE STATEMENT: This study addressed the role of the coxsackievirus and adenovirus receptor (CAR), a single-pass cell adhesion molecule, in the adult brain. Our results demonstrate that CAR is expressed by mature neurons throughout the brain. In addition, we propose divergent roles for CAR in immature neurons, during neurogenesis, and at the mature synapse. Notably, CAR loss of function also affects hippocampal plasticity.


Asunto(s)
Enfermedad de Alzheimer/patología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/deficiencia , Hipocampo/patología , Neurogénesis/genética , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Animales , Células Cultivadas , Trastornos del Conocimiento/etiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo
11.
BMC Cancer ; 15: 792, 2015 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-26499116

RESUMEN

BACKGROUND: Carcinogenesis affects not only humans but almost all metazoan species. Understanding the rules driving the occurrence of cancers in the wild is currently expected to provide crucial insights into identifying how some species may have evolved efficient cancer resistance mechanisms. Recently the absence of correlation across species between cancer prevalence and body size (coined as Peto's paradox) has attracted a lot of attention. Indeed, the disparity between this null hypothesis, where every cell is assumed to have an identical probability to undergo malignant transformation, and empirical observations is particularly important to understand, due to the fact that it could facilitate the identification of animal species that are more resistant to carcinogenesis than expected. Moreover it would open up ways to identify the selective pressures that may be involved in cancer resistance. However, Peto's paradox relies on several questionable assumptions, complicating the interpretation of the divergence between expected and observed cancer incidences. DISCUSSIONS: Here we review and challenge the different hypotheses on which this paradox relies on with the aim of identifying how this null hypothesis could be better estimated in order to provide a standard protocol to study the deviation between theoretical/theoretically predicted and observed cancer incidence. We show that due to the disproportion and restricted nature of available data on animal cancers, applying Peto's hypotheses at species level could result in erroneous conclusions, and actually assume the existence of a paradox. Instead of using species level comparisons, we propose an organ level approach to be a more accurate test of Peto's assumptions. SUMMARY: The accuracy of Peto's paradox assumptions are rarely valid and/or quantifiable, suggesting the need to reconsider the use of Peto's paradox as a null hypothesis in identifying the influence of natural selection on cancer resistance mechanisms.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Inmunidad Innata/inmunología , Neoplasias/genética , Neoplasias/inmunología , Animales , Evolución Biológica , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Transformación Celular Neoplásica/patología , Humanos , Neoplasias/patología , Especificidad de la Especie
12.
Mol Ther ; 22(4): 762-73, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24343103

RESUMEN

Severe deficiency in lysosomal ß-glucuronidase (ß-glu) enzymatic activity results in mucopolysaccharidosis (MPS) VII, an orphan disease with symptoms often appearing in early childhood. Symptoms are variable, but many patients have multiple organ disorders including neurological defects. At the cellular level, deficiency in ß-glu activity leads to abnormal accumulation of glycosaminoglycans (GAGs), and secondary accumulation of GM2 and GM3 gangliosides, which have been linked to neuroinflammation. There have been encouraging gene transfer studies in the MPS VII mouse brain, but this is the first study attempting the correction of the >200-fold larger and challenging canine MPS VII brain. Here, the efficacy of a helper-dependent (HD) canine adenovirus (CAV-2) vector harboring a human GUSB expression cassette (HD-RIGIE) in the MPS VII dog brain was tested. Vector genomes, ß-glu activity, GAG content, lysosome morphology and neuropathology were analyzed and quantified. Our data demonstrated that CAV-2 vectors preferentially transduced neurons and axonal retrograde transport from the injection site to efferent regions was efficient. HD-RIGIE injections, associated with mild and transient immunosuppression, corrected neuropathology in injected and noninjected structures throughout the cerebrum. These data support the clinical evaluation of HD CAV-2 vectors to treat the neurological defects associated with MPS VII and possibly other neuropathic lysosomal storage diseases.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Mucopolisacaridosis VII/genética , beta-Glucosidasa/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Perros , Regulación Enzimológica de la Expresión Génica , Glicosaminoglicanos/metabolismo , Humanos , Ratones , Mucopolisacaridosis VII/terapia , Mucopolisacaridosis VII/veterinaria , beta-Glucosidasa/administración & dosificación , beta-Glucosidasa/biosíntesis
13.
Proc Natl Acad Sci U S A ; 108(32): 13141-6, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788492

RESUMEN

Serotonin (5-HT) has long been recognized as a neurotransmitter in the central nervous system, where it modulates a variety of behavioral functions. Availability of 5-HT depends on the expression of the enzyme tryptophan hydroxylase (TPH), and the recent discovery of a dual system for 5-HT synthesis in the brain (TPH2) and periphery (TPH1) has renewed interest in studying the potential functions played by 5-HT in nonnervous tissues. Moreover, characterization of the TPH1 knockout mouse model (TPH1(-/-)) led to the identification of unsuspected roles for peripheral 5-HT, revealing the importance of this monoamine in regulating key physiological functions outside the brain. Here, we present in vivo data showing that mice deficient in peripheral 5-HT display morphological and cellular features of ineffective erythropoiesis. The central event occurs in the bone marrow where the absence of 5-HT hampers progression of erythroid precursors expressing 5-HT(2A) and 5-HT(2B) receptors toward terminal differentiation. In addition, red blood cells from 5-HT-deficient mice are more sensitive to macrophage phagocytosis and have a shortened in vivo half-life. The combination of these two defects causes TPH1(-/-) animals to develop a phenotype of macrocytic anemia. Direct evidence for a 5-HT effect on erythroid precursors is provided by supplementation of the culture medium with 5-HT that increases the proliferative capacity of both 5-HT-deficient and normal cells. Our thorough analysis of TPH1(-/-) mice provides a unique model of morphological and functional aberrations of erythropoiesis and identifies 5-HT as a key factor for red blood cell production and survival.


Asunto(s)
Eritrocitos/patología , Eritropoyesis , Serotonina/deficiencia , Anemia Macrocítica/complicaciones , Anemia Macrocítica/enzimología , Anemia Macrocítica/patología , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos , Eritrocitos/efectos de los fármacos , Eritrocitos/enzimología , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Eritropoyesis/efectos de los fármacos , Hierro/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Receptores de Serotonina/metabolismo , Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Siderosis/complicaciones , Siderosis/patología , Bazo/efectos de los fármacos , Bazo/patología , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/metabolismo
14.
Mol Oncol ; 18(6): 1510-1530, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38459621

RESUMEN

The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.


Asunto(s)
Proliferación Celular , Neoplasias del Colon , Regulación Neoplásica de la Expresión Génica , Proteína de Interacción con Receptores Nucleares 1 , Factor de Transcripción HES-1 , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteína de Interacción con Receptores Nucleares 1/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , Transducción de Señal , Factor de Transcripción HES-1/metabolismo , Factor de Transcripción HES-1/genética
15.
Exp Mol Pathol ; 94(2): 314-21, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23295309

RESUMEN

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract and are often associated with KIT or PDGFRA gene mutations. GIST cells might arise from the interstitial cells of Cajal (ICCs) or from a mesenchymal precursor that is common to ICCs and smooth muscle cells (SMCs). Here, we analyzed the mRNA and protein expression of RNA-Binding Protein with Multiple Splicing-2 (RBPMS2), an early marker of gastrointestinal SMC precursors, in human GISTs (n=23) by in situ hybridization, quantitative RT-PCR analysis and immunohistochemistry. The mean RBPMS2 mRNA level in GISTs was 42-fold higher than in control gastrointestinal samples (p<0.001). RBPMS2 expression was not correlated with KIT and PDGFRA expression levels, but was higher in GISTs harboring KIT mutations than in tumors with wild type KIT and PDGFRA or in GISTs with PDGFRA mutations that were characterized by the lowest RBPMS2 levels. Moreover, RBPMS2 levels were 64-fold higher in GIST samples with high risk of aggressive behavior than in adult control gastrointestinal samples and 6.2-fold higher in high risk than in low risk GIST specimens. RBPMS2 protein level was high in 87% of the studied GISTs independently of their histological classification. Finally, by inhibiting the KIT signaling pathway in GIST882 cells, we show that RBPMS2 expression is independent of KIT activation. In conclusion, RBPMS2 is up-regulated in GISTs compared to normal adult gastrointestinal tissues, indicating that RBPMS2 might represent a new diagnostic marker for GISTs and a potential target for cancer therapy.


Asunto(s)
Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas de Unión al ARN/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Línea Celular Tumoral , Femenino , Tracto Gastrointestinal/metabolismo , Expresión Génica , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal
16.
Proc Natl Acad Sci U S A ; 107(43): 18640-5, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20921402

RESUMEN

The first step of Plasmodium development in vertebrates is the transformation of the sporozoite, the parasite stage injected by the mosquito in the skin, into merozoites, the stage that invades erythrocytes and initiates the disease. The current view is that, in mammals, this stage conversion occurs only inside hepatocytes. Here, we document the transformation of sporozoites of rodent-infecting Plasmodium into merozoites in the skin of mice. After mosquito bite, ∼50% of the parasites remain in the skin, and at 24 h ∼10% are developing in the epidermis and the dermis, as well as in the immunoprivileged hair follicles where they can survive for weeks. The parasite developmental pathway in skin cells, although frequently abortive, leads to the generation of merozoites that are infective to erythrocytes and are released via merosomes, as typically observed in the liver. Therefore, during malaria in rodents, the skin is not just the route to the liver but is also the final destination for many inoculated parasites, where they can differentiate into merozoites and possibly persist.


Asunto(s)
Plasmodium berghei/crecimiento & desarrollo , Plasmodium yoelii/crecimiento & desarrollo , Piel/parasitología , Animales , Anopheles/parasitología , Dermis/parasitología , Epidermis/parasitología , Proteínas Fluorescentes Verdes/genética , Folículo Piloso/parasitología , Interacciones Huésped-Parásitos , Malaria/parasitología , Malaria/transmisión , Merozoítos/crecimiento & desarrollo , Ratones , Ratones Pelados , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Plasmodium yoelii/genética , Plasmodium yoelii/patogenicidad , Esporozoítos/crecimiento & desarrollo
17.
Proc Natl Acad Sci U S A ; 107(33): 14775-80, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20679209

RESUMEN

Neuronal ceroid lipofuscinoses (NCLs) represent the most common group of inherited progressive encephalopathies in children. They are characterized by progressive loss of vision, mental and motor deterioration, epileptic seizures, and premature death. Rare adult forms of NCL with late onset are known as Kufs' disease. Loci underlying these adult forms remain unknown due to the small number of patients and genetic heterogeneity. Here we confirm that a late-onset form of NCL recessively segregates in US and French pedigrees of American Staffordshire Terrier (AST) dogs. Through combined association, linkage, and haplotype analyses, we mapped the disease locus to a single region of canine chromosome 9. We eventually identified a worldwide breed-specific variant in exon 2 of the Arylsulfatase G (ARSG) gene, which causes a p.R99H substitution in the vicinity of the catalytic domain of the enzyme. In transfected cells or leukocytes from affected dogs, the missense change leads to a 75% decrease in sulfatase activity, providing a functional confirmation that the variant might be the NCL-causing mutation. Our results uncover a protein involved in neuronal homeostasis, identify a family of candidate genes to be screened in patients with Kufs' disease, and suggest that a deficiency in sulfatase is part of the NCL pathogenesis.


Asunto(s)
Arilsulfatasas/genética , Enfermedades de los Perros/genética , Mutación Missense , Lipofuscinosis Ceroideas Neuronales/veterinaria , Transportadoras de Casetes de Unión a ATP/genética , Factores de Edad , Animales , Arilsulfatasas/deficiencia , Dominio Catalítico/genética , Línea Celular , Corteza Cerebelosa/metabolismo , Corteza Cerebelosa/patología , Corteza Cerebelosa/ultraestructura , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Enfermedades de los Perros/enzimología , Perros , Femenino , Perfilación de la Expresión Génica , Frecuencia de los Genes , Genotipo , Haplotipos , Humanos , Masculino , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Linaje , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Stem Cell Res ; 72: 103192, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37660555

RESUMEN

Type 2 Long QT Syndrome (LQT2) is a rare genetic heart rhythm disorder causing life-threatening arrhythmias. We derived induced pluripotent stem cell (iPSC) lines from two patients with LQT2, aged 18 and 6, both carrying a heterozygous missense mutation on the 3rd and 11th exons of KCNH2. The iPSC lines exhibited normal genomes, expressed pluripotent markers, and differentiated into trilineage embryonic layers. These patient-specific iPSC lines provide a valuable model to study the molecular and functional impact of the hERG channel gene mutation in LQT2 and to develop personalized therapeutic approaches for this syndrome.


Asunto(s)
Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Canal de Potasio ERG1/genética , Síndrome de QT Prolongado/metabolismo , Arritmias Cardíacas/metabolismo , Mutación
19.
Gastroenterology ; 141(4): 1509-19, 1519.e1-3, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21762662

RESUMEN

BACKGROUND & AIMS: Chronic, progressive hepatobiliary disease is the most severe complication of erythropoietic protoporphyria (EPP) and can require liver transplantation, although the mechanisms that lead to liver failure are unknown. We characterized protoporphyrin-IX (PPIX)-linked hepatobiliary disease in BALB/c and C57BL/6 (Fechm1Pas) mice with mutations in ferrochelatase as models for EPP. METHODS: Fechm1Pas and wild-type (control) mice were studied at 12-14 weeks of age. PPIX was quantified; its distribution in the liver, serum levels of lipoprotein-X, liver histology, contents of bile salt and cholesterol phospholipids, and expression of genes were compared in mice of the BALB/c and C57BL/6 backgrounds. The in vitro binding affinity of PPIX for bile components was determined. RESULTS: Compared with mice of the C57BL/6 background, BALB/c Fechm1Pas mice had a more severe pattern of cholestasis, fibrosis with portoportal bridging, bile acid regurgitation, sclerosing cholangitis, and hepatolithiasis. In C57BL/6 Fechm1Pas mice, PPIX was sequestrated mainly in the cytosol of hepatocytes and Kupffer cells, whereas, in BALB/c Fechm1Pas mice, PPIX was localized within enlarged bile canaliculi. Livers of C57BL/6 Fechm1Pas mice were protected through a combination of lower efflux of PPIX and reduced synthesis and export of bile acid. CONCLUSIONS: PPIX binds to bile components and disrupts the physiologic equilibrium of phospholipids, bile acids, and cholesterol in bile. This process might be involved in pathogenesis of sclerosing cholangitis from EPP; a better understanding might improve diagnosis and development of reagents to treat or prevent liver failure in patients with EPP.


Asunto(s)
Colangitis Esclerosante/prevención & control , Hepatocitos/metabolismo , Macrófagos del Hígado/metabolismo , Porfiria Eritropoyética/metabolismo , Protoporfirinas/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Colangitis Esclerosante/genética , Colangitis Esclerosante/metabolismo , Colangitis Esclerosante/patología , Colesterol/metabolismo , Modelos Animales de Enfermedad , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Regulación de la Expresión Génica , Genotipo , Hepatocitos/patología , Macrófagos del Hígado/patología , Lipoproteína X/sangre , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Fosfolípidos/metabolismo , Mutación Puntual , Porfiria Eritropoyética/complicaciones , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/patología , Índice de Severidad de la Enfermedad
20.
Development ; 136(21): 3647-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19820183

RESUMEN

The size of the mammalian body is determined by genetic and environmental factors differentially modulating pre- and postnatal growth. We now report a control of growth acting in the mouse from the first cleavages to the postnatal stages. It was evidenced by a hereditary epigenetic modification (paramutation) created by injection of a miR-124 microRNA into fertilized eggs. From the blastocyst to the adult, mouse pups born after microinjection of this miRNA showed a 30% increase in size. At the blastocyst stage, frequent duplication of the inner cell mass resulted in twin pregnancies. A role of sperm RNA as a transgenerational signal was confirmed by the giant phenotype of the progeny of transgenic males expressing miR-124 during spermiogenesis. In E2.5 to E8.5 embryos, increased levels of several transcripts with sequence homology to the microRNA were noted, including those of Sox9, a gene known for its crucial role in the progenitors of several adult tissues. A role in embryonic growth was confirmed by the large size of embryos expressing a Sox9 DNA transgene. Increased expression in the paramutants was not related to a change in miR-124 expression, but to the establishment of a distinct, heritable chromatin structure in the promoter region of Sox9. While the heritability of body size is not readily accounted for by Mendelian genetics, our results suggest the alternate model of RNA-mediated heritable epigenetic modifications.


Asunto(s)
Tamaño Corporal/genética , Epigénesis Genética , Ratones/embriología , MicroARNs/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Masculino , Ratones/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA