Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
EMBO Rep ; 24(5): e55326, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36929576

RESUMEN

The Sin3 transcriptional regulator homolog A (Sin3A) is the core member of a multiprotein chromatin-modifying complex. Its inactivation at the CD4/CD8 double-negative stage halts further thymocyte development. Among various functions, Sin3A regulates STAT3 transcriptional activity, central to the differentiation of Th17 cells active in inflammatory disorders and opportunistic infections. To further investigate the consequences of conditional Sin3A inactivation in more mature precursors and post-thymic T cell, we have generated CD4-Cre and CD4-CreERT2 Sin3AF/F mice. Sin3A inactivation in vivo hinders both thymocyte development and peripheral T-cell survival. In vitro, in Th17 skewing conditions, Sin3A-deficient cells proliferate and acquire memory markers and yet fail to properly upregulate Il17a, Il23r, and Il22. Instead, IL-2+ and FOXP3+ are mostly enriched for, and their inhibition partially rescues IL-17A+ T cells. Notably, Sin3A deletion also causes an enrichment of genes implicated in the mTORC1 signaling pathway, overt STAT3 activation, and aberrant cytoplasmic RORγt accumulation. Thus, together our data unveil a previously unappreciated role for Sin3A in shaping critical signaling events central to the acquisition of immunoregulatory T-cell phenotypes.


Asunto(s)
Linfocitos T CD4-Positivos , Interleucina-17 , Animales , Ratones , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Células Th17
2.
Rheumatology (Oxford) ; 62(10): 3469-3479, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-36802235

RESUMEN

OBJECTIVE: Trained immunity (TI) is a de facto memory program of innate immune cells, characterized by immunometabolic and epigenetic changes sustaining enhanced production of cytokines. TI evolved as a protective mechanism against infections; however, inappropriate activation can cause detrimental inflammation and might be implicated in the pathogenesis of chronic inflammatory diseases. In this study, we investigated the role of TI in the pathogenesis of giant cell arteritis (GCA), a large-vessel vasculitis characterized by aberrant macrophage activation and excess cytokine production. METHODS: Monocytes from GCA patients and from age- and sex-matched healthy donors were subjected to polyfunctional studies, including cytokine production assays at baseline and following stimulation, intracellular metabolomics, chromatin immunoprecipitation-qPCR, and combined ATAC/RNA sequencing. Immunometabolic activation (i.e. glycolysis) was assessed in inflamed vessels of GCA patients with FDG-PET and immunohistochemistry (IHC), and the role of this pathway in sustaining cytokine production was confirmed with selective pharmacologic inhibition in GCA monocytes. RESULTS: GCA monocytes exhibited hallmark molecular features of TI. Specifically, these included enhanced IL-6 production upon stimulation, typical immunometabolic changes (e.g. increased glycolysis and glutaminolysis) and epigenetic changes promoting enhanced transcription of genes governing pro-inflammatory activation. Immunometabolic changes of TI (i.e. glycolysis) were a feature of myelomonocytic cells in GCA lesions and were required for enhanced cytokine production. CONCLUSIONS: Myelomonocytic cells in GCA activate TI programs sustaining enhanced inflammatory activation with excess cytokine production.


Asunto(s)
Arteritis de Células Gigantes , Humanos , Arteritis de Células Gigantes/patología , Monocitos/metabolismo , Inmunidad Entrenada , Inflamación , Citocinas
3.
Nucleic Acids Res ; 48(16): 8993-9006, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32710624

RESUMEN

Eukaryotic DNA is organized in nucleosomes, which package DNA and regulate its accessibility to transcription, replication, recombination and repair. Here, we show that in living cells nucleosomes protect DNA from high-energy radiation and reactive oxygen species. We combined sequence-based methods (ATAC-seq and BLISS) to determine the position of both nucleosomes and double strand breaks (DSBs) in the genome of nucleosome-rich malignant mesothelioma cells, and of the same cells partially depleted of nucleosomes. The results were replicated in the human MCF-7 breast carcinoma cell line. We found that, for each genomic sequence, the probability of DSB formation is directly proportional to the fraction of time it is nucleosome-free; DSBs accumulate distal from the nucleosome dyad axis. Nucleosome free regions and promoters of actively transcribed genes are more sensitive to DSB formation, and consequently to mutation. We argue that this may be true for a variety of chemical and physical DNA damaging agents.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , ADN/efectos de la radiación , Nucleosomas/metabolismo , Animales , Línea Celular , Humanos , Células MCF-7 , Ratones
4.
Mol Med ; 27(1): 129, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663207

RESUMEN

BACKGROUND: Host inflammation contributes to determine whether SARS-CoV-2 infection causes mild or life-threatening disease. Tools are needed for early risk assessment. METHODS: We studied in 111 COVID-19 patients prospectively followed at a single reference Hospital fifty-three potential biomarkers including alarmins, cytokines, adipocytokines and growth factors, humoral innate immune and neuroendocrine molecules and regulators of iron metabolism. Biomarkers at hospital admission together with age, degree of hypoxia, neutrophil to lymphocyte ratio (NLR), lactate dehydrogenase (LDH), C-reactive protein (CRP) and creatinine were analysed within a data-driven approach to classify patients with respect to survival and ICU outcomes. Classification and regression tree (CART) models were used to identify prognostic biomarkers. RESULTS: Among the fifty-three potential biomarkers, the classification tree analysis selected CXCL10 at hospital admission, in combination with NLR and time from onset, as the best predictor of ICU transfer (AUC [95% CI] = 0.8374 [0.6233-0.8435]), while it was selected alone to predict death (AUC [95% CI] = 0.7334 [0.7547-0.9201]). CXCL10 concentration abated in COVID-19 survivors after healing and discharge from the hospital. CONCLUSIONS: CXCL10 results from a data-driven analysis, that accounts for presence of confounding factors, as the most robust predictive biomarker of patient outcome in COVID-19.


Asunto(s)
COVID-19/diagnóstico , Quimiocina CXCL10/sangre , Enfermedad de la Arteria Coronaria/diagnóstico , Diabetes Mellitus/diagnóstico , Hipertensión/diagnóstico , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , COVID-19/sangre , COVID-19/inmunología , COVID-19/mortalidad , Comorbilidad , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/inmunología , Enfermedad de la Arteria Coronaria/mortalidad , Creatina/sangre , Diabetes Mellitus/sangre , Diabetes Mellitus/inmunología , Diabetes Mellitus/mortalidad , Femenino , Hospitalización , Humanos , Hipertensión/sangre , Hipertensión/inmunología , Hipertensión/mortalidad , Inmunidad Humoral , Inmunidad Innata , Inflamación , Unidades de Cuidados Intensivos , L-Lactato Deshidrogenasa/sangre , Recuento de Leucocitos , Linfocitos/inmunología , Linfocitos/patología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/patología , Pronóstico , Estudios Prospectivos , Estudios Retrospectivos , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
5.
Blood ; 128(20): 2435-2449, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27574188

RESUMEN

Deep venous thrombosis (DVT) is one of the most common cardiovascular diseases, but its pathophysiology remains incompletely understood. Although sterile inflammation has recently been shown to boost coagulation during DVT, the underlying molecular mechanisms are not fully resolved, which could potentially identify new anti-inflammatory approaches to prophylaxis and therapy of DVT. Using a mouse model of venous thrombosis induced by flow reduction in the vena cava inferior, we identified blood-derived high-mobility group box 1 protein (HMGB1), a prototypical mediator of sterile inflammation, to be a master regulator of the prothrombotic cascade involving platelets and myeloid leukocytes fostering occlusive DVT formation. Transfer of platelets into Hmgb1-/- chimeras showed that this cell type is the major source of HMGB1, exposing reduced HMGB1 on their surface upon activation thereby enhancing the recruitment of monocytes. Activated leukocytes in turn support oxidation of HMGB1 unleashing its prothrombotic activity and promoting platelet aggregation. This potentiates the amount of HMGB1 and further nurtures the accumulation and activation of monocytes through receptor for advanced glycation end products (RAGE) and Toll-like receptor 2, leading to local delivery of monocyte-derived tissue factor and cytokines. Moreover, disulfide HMGB1 facilitates formation of prothrombotic neutrophil extracellular traps (NETs) mediated by RAGE, exposing additional HMGB1 on their extracellular DNA strands. Eventually, a vicious circle of coagulation and inflammation is set in motion leading to obstructive DVT formation. Therefore, platelet-derived disulfide HMGB1 is a central mediator of the sterile inflammatory process in venous thrombosis and could be an attractive target for an anti-inflammatory approach for DVT prophylaxis.


Asunto(s)
Plaquetas/metabolismo , Proteína HMGB1/fisiología , Trombosis de la Vena/genética , Animales , Plaquetas/patología , Disulfuros/química , Disulfuros/metabolismo , Proteína HMGB1/química , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
6.
Mol Med ; 21: 526-35, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26101955

RESUMEN

Salicylic acid (SA) and its derivatives have been used for millennia to reduce pain, fever and inflammation. In addition, prophylactic use of acetylsalicylic acid, commonly known as aspirin, reduces the risk of heart attack, stroke and certain cancers. Because aspirin is rapidly de-acetylated by esterases in human plasma, much of aspirin's bioactivity can be attributed to its primary metabolite, SA. Here we demonstrate that human high mobility group box 1 (HMGB1) is a novel SA-binding protein. SA-binding sites on HMGB1 were identified in the HMG-box domains by nuclear magnetic resonance (NMR) spectroscopic studies and confirmed by mutational analysis. Extracellular HMGB1 is a damage-associated molecular pattern molecule (DAMP), with multiple redox states. SA suppresses both the chemoattractant activity of fully reduced HMGB1 and the increased expression of proinflammatory cytokine genes and cyclooxygenase 2 (COX-2) induced by disulfide HMGB1. Natural and synthetic SA derivatives with greater potency for inhibition of HMGB1 were identified, providing proof-of-concept that new molecules with high efficacy against sterile inflammation are attainable. An HMGB1 protein mutated in one of the SA-binding sites identified by NMR chemical shift perturbation studies retained chemoattractant activity, but lost binding of and inhibition by SA and its derivatives, thereby firmly establishing that SA binding to HMGB1 directly suppresses its proinflammatory activities. Identification of HMGB1 as a pharmacological target of SA/aspirin provides new insights into the mechanisms of action of one of the world's longest and most used natural and synthetic drugs. It may also provide an explanation for the protective effects of low-dose aspirin usage.


Asunto(s)
Aspirina/farmacología , Proteína HMGB1/genética , Inflamación/genética , Ácido Salicílico/farmacología , Aspirina/química , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Proteína HMGB1/biosíntesis , Proteína HMGB1/química , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Mutación , Resonancia Magnética Nuclear Biomolecular , Ácido Salicílico/química
7.
PLoS Biol ; 9(6): e1001086, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21738444

RESUMEN

The basic unit of genome packaging is the nucleosome, and nucleosomes have long been proposed to restrict DNA accessibility both to damage and to transcription. Nucleosome number in cells was considered fixed, but recently aging yeast and mammalian cells were shown to contain fewer nucleosomes. We show here that mammalian cells lacking High Mobility Group Box 1 protein (HMGB1) contain a reduced amount of core, linker, and variant histones, and a correspondingly reduced number of nucleosomes, possibly because HMGB1 facilitates nucleosome assembly. Yeast nhp6 mutants lacking Nhp6a and -b proteins, which are related to HMGB1, also have a reduced amount of histones and fewer nucleosomes. Nucleosome limitation in both mammalian and yeast cells increases the sensitivity of DNA to damage, increases transcription globally, and affects the relative expression of about 10% of genes. In yeast nhp6 cells the loss of more than one nucleosome in four does not affect the location of nucleosomes and their spacing, but nucleosomal occupancy. The decrease in nucleosomal occupancy is non-uniform and can be modelled assuming that different nucleosomal sites compete for available histones. Sites with a high propensity to occupation are almost always packaged into nucleosomes both in wild type and nucleosome-depleted cells; nucleosomes on sites with low propensity to occupation are disproportionately lost in nucleosome-depleted cells. We suggest that variation in nucleosome number, by affecting nucleosomal occupancy both genomewide and gene-specifically, constitutes a novel layer of epigenetic regulation.


Asunto(s)
Genoma , Proteína HMGB1/metabolismo , Histonas/metabolismo , Nucleosomas/metabolismo , Transcripción Genética , Animales , ADN/genética , ADN/metabolismo , Daño del ADN , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/fisiología , Proteína HMGB1/genética , Células HeLa , Histonas/genética , Humanos , Ratones , Modelos Teóricos , ARN/genética , ARN/metabolismo , Levaduras/genética , Levaduras/metabolismo
8.
J Theor Biol ; 347: 44-53, 2014 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-24447586

RESUMEN

The mathematical modeling of the NF-κB oscillations has attracted considerable attention in recent times, but there is a lack of simple models in the literature that can capture the main features of the dynamics of this important transcription factor. For this reason we propose a simple model that summarizes the key steps of the NF-κB pathway. We show that the resulting 5-dimensional dynamical system can reproduce different phenomena observed in experiments. Our model can display smooth and spiky oscillations in the amount of nuclear NF-κB and can reproduce the variety of dynamics observed when different stimulations such as TNF-α and LPS are used. Furthermore we show that the model can be easily extended to reproduce the expression of early, intermediate and late genes upon stimulation. As a final example we show that our simple model can mimic the different transcriptional outputs observed when cells are treated with two different drugs leading to nuclear localization of NF-κB: Leptomycin B and Cycloheximide.


Asunto(s)
Modelos Teóricos , FN-kappa B/metabolismo , Transcripción Genética
9.
Neurobiol Dis ; 58: 102-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23523633

RESUMEN

Toll-like receptor 4 (TLR4) activation in neuron and astrocytes by High Mobility Group Box 1 (HMGB1) protein is a key mechanism of seizure generation. HMGB1 also activates the Receptor for Advanced Glycation Endproducts (RAGE), but it was unknown whether RAGE activation contributes to seizures or to HMGB1 proictogenic effects. We found that acute EEG seizures induced by 7ng intrahippocampal kainic acid (KA) were significantly reduced in Rage-/- mice relative to wild type (Wt) mice. The proictogenic effect of HMGB1 was decreased in Rage-/- mice, but less so, than in Tlr4-/- mice. In a mouse mesial temporal lobe epilepsy (mTLE) model, status epilepticus induced by 200ng intrahippocampal KA and the onset of the spontaneous epileptic activity were similar in Rage-/-, Tlr4-/- and Wt mice. However, the number of hippocampal paroxysmal episodes and their duration were both decreased in epileptic Rage-/- and Tlr4-/- mice vs Wt mice. All strains of epileptic mice displayed similar cognitive deficits in the novel object recognition test vs the corresponding control mice. CA1 neuronal cell loss was increased in epileptic Rage-/- vs epileptic Wt mice, while granule cell dispersion and doublecortin (DCX)-positive neurons were similarly affected. Notably, DCX neurons were preserved in epileptic Tlr4-/- mice. We did not find compensatory changes in HMGB1-related inflammatory signaling nor in glutamate receptor subunits in Rage-/- and Tlr4-/- naïve mice, except for ~20% NR2B subunit reduction in Rage-/- mice. RAGE was induced in neurons, astrocytes and microvessels in human and experimental mTLE hippocampi. We conclude that RAGE contributes to hyperexcitability underlying acute and chronic seizures, as well as to the proictogenic effects of HMGB1. RAGE and TLR4 play different roles in the neuropathologic sequelae developing after status epilepticus. These findings reveal new molecular mechanisms underlying seizures, cell loss and neurogenesis which involve inflammatory pathways upregulated in human epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Regulación de la Expresión Génica/genética , Receptores Inmunológicos/metabolismo , Convulsiones/metabolismo , Regulación hacia Arriba/fisiología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Estimulación Eléctrica/efectos adversos , Electroencefalografía , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/patología , Agonistas de Aminoácidos Excitadores/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Proteína HMGB1/administración & dosificación , Proteína HMGB1/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Ácido Kaínico/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/deficiencia , Convulsiones/inducido químicamente , Convulsiones/etiología , Receptor Toll-Like 4/deficiencia , Regulación hacia Arriba/genética
10.
iScience ; 26(12): 108573, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38144455

RESUMEN

Transcription factor dynamics is fundamental to determine the activation of accurate transcriptional programs and yet is heterogeneous at a single-cell level, even within homogeneous populations. We asked how such heterogeneity emerges for the nuclear factor κB (NF-κB). We found that clonal populations of immortalized fibroblasts derived from a single mouse embryo display robustly distinct NF-κB dynamics upon tumor necrosis factor ɑ (TNF-ɑ) stimulation including persistent, oscillatory, and weak activation, giving rise to differences in the transcription of its targets. By combining transcriptomics and simulations we show how less than two-fold differences in the expression levels of genes coding for key proteins of the signaling cascade and feedback system are predictive of the differences of the NF-κB dynamic response of the clones to TNF-ɑ and IL-1ß. We propose that small transcriptional differences in the regulatory circuit of a transcription factor can lead to distinct signaling dynamics in cells within homogeneous cell populations and among different cell types.

11.
Eur J Immunol ; 41(7): 2086-96, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21480212

RESUMEN

The mammalian target of rapamycin (mTOR) controls T-cell differentiation in response to polarizing cytokines. We previously found that mTOR blockade by rapamycin (RAPA) delays the G1-S cell cycle transition and lymphocyte proliferation. Here, we report that both mTOR complex 1 and mTOR complex 2 are readily activated following TCR/CD28 engagement and are critical for early expression of Ifng, Il4 and Foxp3, and for effector T cell differentiation in the absence of polarizing cytokines. While inhibition of mTOR complex 1 and cell division were evident at low doses of RAPA, inhibition of mTOR complex 2, Ifng, Il4 and Foxp3 expression, and T-cell polarization required higher doses and more prolonged treatments. We found that while T-bet and GATA3 were readily induced following TCR/CD28 engagement, administration of RAPA delayed their expression, and interfered with the loss of DNA methylation within Ifng and Il4 promoter regions. In contrast, RAPA prevented activation-dependent DNA methylation of the Foxp3 promoter favoring Foxp3 expression. As a result, RAPA-cultured cells lacked immediate effector functions and instead were enriched for IL-2+ cells. We propose that mTOR-signaling, by timing the expression of critical transcription factors and DNA methylation of proximal promoter regions, regulates transcriptional competence at immunologically relevant sites and hence lymphocyte differentiation.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/genética , Interferón gamma/genética , Interleucina-4/genética , Sirolimus/farmacología , Transcripción Genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Metilación de ADN , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/biosíntesis , Interferón gamma/metabolismo , Interleucina-2/biosíntesis , Interleucina-4/metabolismo , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Complejos Multiproteicos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteínas/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo
12.
Nat Commun ; 12(1): 5205, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471128

RESUMEN

Molecular mechanisms associated with human germ cell aplasia in infertile men remain undefined. Here we perform single-cell transcriptome profiling to highlight differentially expressed genes and pathways in each somatic cell type in testes of men with idiopathic germ cell aplasia. We identify immaturity of Leydig cells, chronic tissue inflammation, fibrosis, and senescence phenotype of the somatic cells, as well markers of chronic inflammation in the blood. We find that deregulated expression of parentally imprinted genes in myoid and immature Leydig cells, with relevant changes in the ratio of Lamin A/C transcripts and an active DNA damage response in Leydig and peritubular myoid cells are also indicative of senescence of the testicular niche. This study offers molecular insights into the pathogenesis of idiopathic germ cell aplasia.


Asunto(s)
Envejecimiento/fisiología , Daño del ADN , Inflamación , Testículo/metabolismo , Envejecimiento/genética , Comunicación Celular , Quimiocinas , Perfilación de la Expresión Génica , Células Germinativas , Humanos , Inflamación/patología , Células Intersticiales del Testículo , Masculino , Fenotipo , Alineación de Secuencia , Espermatogénesis/genética , Espermatogénesis/fisiología , Espermatogonias/metabolismo , Transcriptoma
13.
iScience ; 23(9): 101529, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-33083759

RESUMEN

Nuclear factor (NF)-κB controls the transcriptional response to inflammatory signals by translocating into the nucleus, but we lack a single-cell characterization of the resulting transcription dynamics. Here we show that upon tumor necrosis factor (TNF)-α transcription of NF-κB target genes is heterogeneous in individual cells but results in an average nascent transcription profile that is prompt (i.e., occurs almost immediately) and sharp (i.e., increases and decreases rapidly) compared with NF-κB nuclear localization. Using an NF-κB-controlled MS2 reporter we show that the single-cell nascent transcription is more heterogeneous than NF-κB translocation dynamics, with a fraction of synchronized "first responders" that shape the average transcriptional profile and are more prone to respond to multiple TNF-α stimulations. A mathematical model combining NF-κB-mediated gene activation and a gene refractory state is able to reproduce these features. Our work shows how the expression of target genes induced by transcriptional activators can be heterogeneous across single cells and yet time resolved on average.

14.
Curr Opin Genet Dev ; 15(5): 496-506, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16102963

RESUMEN

Core histones package the genome into nucleosomes and control its accessibility to transcription factors. High mobility group proteins (HMGs) are, after histones, the second most abundant chromatin proteins and exert global genomic functions in establishing active or inactive chromatin domains. It is becoming increasingly clear that they also specifically control the expression of a limited number of genes. Moreover, they contribute to the fine tuning of transcription in response to rapid environmental changes. They do so by interacting with nucleosomes, transcription factors, nucleosome-remodelling machines, and with histone H1.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Animales , Mamíferos
15.
J Vis Exp ; (146)2019 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-31033961

RESUMEN

Nuclear positioning within cells is important for multiple cellular processes in development and regeneration. The most intriguing example of nuclear positioning occurs during skeletal muscle differentiation. Muscle fibers (myofibers) are multinucleated cells formed by the fusion of muscle precursor cells (myoblasts) derived from muscle stem cells (satellite cells) that undergo proliferation and differentiation. Correct nuclear positioning within myofibers is required for the proper muscle regeneration and function. The common procedure to assess myoblast differentiation and myofiber formation relies on fixed cells analyzed by immunofluorescence, which impedes the study of nuclear movement and cell behavior over time. Here, we describe a method for the analysis of myoblast differentiation and myofiber formation by live cell imaging. We provide a software for automated nuclear tracking to obtain a high-throughput quantitative characterization of nuclear dynamics and myoblast behavior (i.e., the trajectory) during differentiation and fusion.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , Imagen Molecular , Mioblastos/citología , Animales , Fusión Celular , Supervivencia Celular , Ratones , Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología
16.
Curr Opin Genet Dev ; 13(2): 170-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12672494

RESUMEN

High mobility group (HMG) proteins are chromatin proteins endowed with 'architectural' capabilities. HMGA proteins are moderately sequence-specific, and help build enhanceosomes by interacting with partner proteins and binding stably to the minor groove of DNA; their acetylation/deacetylation signal enhanceosome assembly or disassembly. HMGBs are much more dynamic proteins: they have no sequence specificity, and help transcription factors and other nuclear proteins bind to their cognate sites by bending the DNA molecule. However, HMGBs are rarely retained within the complex. Similarly, HMGBs interact with nucleosomes and promote their sliding, but remain bound only for fractions of a second. We argue that HMGBs fluidize chromatin - an action that appears opposite to that of histone H1.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas HMGB/fisiología , Animales , ADN/metabolismo , Elementos de Facilitación Genéticos/fisiología , Humanos , Nucleosomas/metabolismo
17.
Chem Biol ; 14(4): 431-41, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17462578

RESUMEN

High-mobility group box 1 protein (HMGB1) is a nuclear component, but extracellularly it serves as a signaling molecule involved in acute and chronic inflammation, for example in sepsis and arthritis. The identification of HMGB1 inhibitors is therefore of significant experimental and clinical interest. We show that glycyrrhizin, a natural anti-inflammatory and antiviral triterpene in clinical use, inhibits HMGB1 chemoattractant and mitogenic activities, and has a weak inhibitory effect on its intranuclear DNA-binding function. NMR and fluorescence studies indicate that glycyrrhizin binds directly to HMGB1 (K(d) approximately 150 microM), interacting with two shallow concave surfaces formed by the two arms of both HMG boxes. Our results explain in part the anti-inflammatory properties of glycyrrhizin, and might direct the design of new derivatives with improved HMGB1-binding properties.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Citocinas/antagonistas & inhibidores , Ácido Glicirrínico/metabolismo , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Células 3T3 , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Sitios de Unión , Citocinas/metabolismo , ADN/metabolismo , Fluorescencia , Ácido Glicirrínico/química , Ácido Glicirrínico/farmacología , Proteína HMGB1/química , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Mitógenos/antagonistas & inhibidores , Mitógenos/farmacología , Modelos Moleculares , Unión Proteica
18.
Biomedicines ; 6(2)2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29673148

RESUMEN

In this review, we aim at describing the results obtained in the past years on dynamics features defining NF-κB regulatory functions, as we believe that these developments might have a transformative effect on the way in which NF-κB involvement in cancer is studied. We will also describe technical aspects of the studies performed in this context, including the use of different cellular models, culture conditions, microscopy approaches and quantification of the imaging data, balancing their strengths and limitations and pointing out to common features and to some open questions. Our emphasis in the methodology will allow a critical overview of literature and will show how these cutting-edge approaches can contribute to shed light on the involvement of NF-κB deregulation in tumour onset and progression. We hypothesize that this “dynamic point of view” can be fruitfully applied to untangle the complex relationship between NF-κB and cancer and to find new targets to restrain cancer growth.

19.
Front Immunol ; 9: 1463, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29997623

RESUMEN

Histones are the protein component of nucleosomes, which are the basic packing unit of chromatin. However, histones are also found in the blood, both as components of nucleosomes leaked out from dead cells, or expelled from neutrophils in the active process of NET formation. Circulating histones contribute to inflammation, and to lethality in sepsis, a hyperinflammatory condition, by interacting with specific receptors, notably toll-like receptor 4 (TLR4). Here, we show that histones are also actively released by LPS-activated macrophages in association with extracellular vesicles. Vesicle-associated histones can be recovered from the plasma of mice with sepsis. Actively released histones are on the outer surface of vesicles and can interact with TLR4. Thus, activated macrophages release histones without dying, at the same time, making their DNA more accessible and communicating to other cells that infection is present.

20.
Nat Commun ; 9(1): 1590, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29686321

RESUMEN

Chromatin is the template for the basic processes of replication and transcription, making the maintenance of chromosomal integrity critical for cell viability. To elucidate how dividing cells respond to alterations in chromatin structure, here we analyse the replication programme of primary cells with altered chromatin configuration caused by the genetic ablation of the HMGB1 gene, or three histone H1 genes. We find that loss of chromatin compaction in H1-depleted cells triggers the accumulation of stalled forks and DNA damage as a consequence of transcription-replication conflicts. In contrast, reductions in nucleosome occupancy due to the lack of HMGB1 cause faster fork progression without impacting the initiation landscape or fork stability. Thus, perturbations in chromatin integrity elicit a range of responses in the dynamics of DNA replication and transcription, with different consequences on replicative stress. These findings have broad implications for our understanding of how defects in chromatin structure contribute to genomic instability.


Asunto(s)
Cromatina/química , Replicación del ADN/fisiología , Conformación Molecular , Transcripción Genética/fisiología , Animales , Cromatina/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Fibroblastos , Inestabilidad Genómica/fisiología , Células HCT116 , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Células Madre Embrionarias de Ratones , Nucleosomas/metabolismo , Cultivo Primario de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA