Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.431
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38986619

RESUMEN

Posterior fossa group A (PFA) ependymoma is a lethal brain cancer diagnosed in infants and young children. The lack of driver events in the PFA linear genome led us to search its 3D genome for characteristic features. Here, we reconstructed 3D genomes from diverse childhood tumor types and uncovered a global topology in PFA that is highly reminiscent of stem and progenitor cells in a variety of human tissues. A remarkable feature exclusively present in PFA are type B ultra long-range interactions in PFAs (TULIPs), regions separated by great distances along the linear genome that interact with each other in the 3D nuclear space with surprising strength. TULIPs occur in all PFA samples and recur at predictable genomic coordinates, and their formation is induced by expression of EZHIP. The universality of TULIPs across PFA samples suggests a conservation of molecular principles that could be exploited therapeutically.

2.
Cell ; 173(3): 693-705.e22, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677513

RESUMEN

Liquid-liquid phase separation (LLPS) is believed to underlie formation of biomolecular condensates, cellular compartments that concentrate macromolecules without surrounding membranes. Physical mechanisms that control condensate formation/dissolution are poorly understood. The RNA-binding protein fused in sarcoma (FUS) undergoes LLPS in vitro and associates with condensates in cells. We show that the importin karyopherin-ß2/transportin-1 inhibits LLPS of FUS. This activity depends on tight binding of karyopherin-ß2 to the C-terminal proline-tyrosine nuclear localization signal (PY-NLS) of FUS. Nuclear magnetic resonance (NMR) analyses reveal weak interactions of karyopherin-ß2 with sequence elements and structural domains distributed throughout the entirety of FUS. Biochemical analyses demonstrate that most of these same regions also contribute to LLPS of FUS. The data lead to a model where high-affinity binding of karyopherin-ß2 to the FUS PY-NLS tethers the proteins together, allowing multiple, distributed weak intermolecular contacts to disrupt FUS self-association, blocking LLPS. Karyopherin-ß2 may act analogously to control condensates in diverse cellular contexts.


Asunto(s)
Transporte Activo de Núcleo Celular , Señales de Localización Nuclear , Proteína FUS de Unión a ARN/química , beta Carioferinas/química , Sitios de Unión , Degeneración Lobar Frontotemporal/metabolismo , Humanos , Carioferinas/metabolismo , Luz , Extracción Líquido-Líquido , Sustancias Macromoleculares , Espectroscopía de Resonancia Magnética , Mutación , Nefelometría y Turbidimetría , Unión Proteica , Dominios Proteicos , ARN/química , Dispersión de Radiación , Temperatura
3.
Nat Immunol ; 21(10): 1194-1204, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32895539

RESUMEN

Early atherosclerosis depends upon responses by immune cells resident in the intimal aortic wall. Specifically, the healthy intima is thought to be populated by vascular dendritic cells (DCs) that, during hypercholesterolemia, initiate atherosclerosis by being the first to accumulate cholesterol. Whether these cells remain key players in later stages of disease is unknown. Using murine lineage-tracing models and gene expression profiling, we reveal that myeloid cells present in the intima of the aortic arch are not DCs but instead specialized aortic intima resident macrophages (MacAIR) that depend upon colony-stimulating factor 1 and are sustained by local proliferation. Although MacAIR comprise the earliest foam cells in plaques, their proliferation during plaque progression is limited. After months of hypercholesterolemia, their presence in plaques is overtaken by recruited monocytes, which induce MacAIR-defining genes. These data redefine the lineage of intimal phagocytes and suggest that proliferation is insufficient to sustain generations of macrophages during plaque progression.


Asunto(s)
Aorta/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Placa Aterosclerótica/inmunología , Túnica Íntima/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Colesterol/metabolismo , Progresión de la Enfermedad , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Parabiosis , Fagocitosis
4.
Cell ; 171(2): 321-330.e14, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28965763

RESUMEN

As organisms age, cells accumulate genetic and epigenetic errors that eventually lead to impaired organ function or catastrophic transformation such as cancer. Because aging reflects a stochastic process of increasing disorder, cells in an organ will be individually affected in different ways, thus rendering bulk analyses of postmitotic adult cells difficult to interpret. Here, we directly measure the effects of aging in human tissue by performing single-cell transcriptome analysis of 2,544 human pancreas cells from eight donors spanning six decades of life. We find that islet endocrine cells from older donors display increased levels of transcriptional noise and potential fate drift. By determining the mutational history of individual cells, we uncover a novel mutational signature in healthy aging endocrine cells. Our results demonstrate the feasibility of using single-cell RNA sequencing (RNA-seq) data from primary cells to derive insights into genetic and transcriptional processes that operate on aging human tissue.


Asunto(s)
Envejecimiento/patología , Senescencia Celular , Mutación , Páncreas/patología , Análisis de la Célula Individual , Adulto , Niño , Preescolar , Humanos , Lactante , Persona de Mediana Edad , Páncreas/citología , Páncreas/fisiología , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ARN , Transcripción Genética
5.
Immunity ; 55(11): 2027-2043.e9, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36243007

RESUMEN

T helper 17 (Th17) cells regulate mucosal barrier defenses but also promote multiple autoinflammatory diseases. Although many molecular determinants of Th17 cell differentiation have been elucidated, the transcriptional programs that sustain Th17 cells in vivo remain obscure. The transcription factor RORγt is critical for Th17 cell differentiation; however, it is not clear whether the closely related RORα, which is co-expressed in Th17 cells, has a distinct role. Here, we demonstrated that although dispensable for Th17 cell differentiation, RORα was necessary for optimal Th17 responses in peripheral tissues. The absence of RORα in T cells led to reductions in both RORγt expression and effector function among Th17 cells. Cooperative binding of RORα and RORγt to a previously unidentified Rorc cis-regulatory element was essential for Th17 lineage maintenance in vivo. These data point to a non-redundant role of RORα in Th17 lineage maintenance via reinforcement of the RORγt transcriptional program.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Regulación de la Expresión Génica , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células Th17/metabolismo , Factores de Transcripción/metabolismo
6.
Cell ; 167(5): 1215-1228.e25, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27839866

RESUMEN

The last steps in mRNA export and remodeling are performed by the Nup82 complex, a large conserved assembly at the cytoplasmic face of the nuclear pore complex (NPC). By integrating diverse structural data, we have determined the molecular architecture of the native Nup82 complex at subnanometer precision. The complex consists of two compositionally identical multiprotein subunits that adopt different configurations. The Nup82 complex fits into the NPC through the outer ring Nup84 complex. Our map shows that this entire 14-MDa Nup82-Nup84 complex assembly positions the cytoplasmic mRNA export factor docking sites and messenger ribonucleoprotein (mRNP) remodeling machinery right over the NPC's central channel rather than on distal cytoplasmic filaments, as previously supposed. We suggest that this configuration efficiently captures and remodels exporting mRNP particles immediately upon reaching the cytoplasmic side of the NPC.


Asunto(s)
Proteínas de Complejo Poro Nuclear/química , Poro Nuclear/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Levaduras/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Fúngicas , Proteínas de Complejo Poro Nuclear/ultraestructura , ARN Mensajero , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/ultraestructura
7.
Mol Cell ; 81(10): 2064-2075.e8, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33756105

RESUMEN

Dysregulated mTORC1 signaling alters a wide range of cellular processes, contributing to metabolic disorders and cancer. Defining the molecular details of downstream effectors is thus critical for uncovering selective therapeutic targets. We report that mTORC1 and its downstream kinase S6K enhance eIF4A/4B-mediated translation of Wilms' tumor 1-associated protein (WTAP), an adaptor for the N6-methyladenosine (m6A) RNA methyltransferase complex. This regulation is mediated by 5' UTR of WTAP mRNA that is targeted by eIF4A/4B. Single-nucleotide-resolution m6A mapping revealed that MAX dimerization protein 2 (MXD2) mRNA contains m6A, and increased m6A modification enhances its degradation. WTAP induces cMyc-MAX association by suppressing MXD2 expression, which promotes cMyc transcriptional activity and proliferation of mTORC1-activated cancer cells. These results elucidate a mechanism whereby mTORC1 stimulates oncogenic signaling via m6A RNA modification and illuminates the WTAP-MXD2-cMyc axis as a potential therapeutic target for mTORC1-driven cancers.


Asunto(s)
Adenosina/análogos & derivados , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Estabilidad del ARN , Adenosina/metabolismo , Animales , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Factores Eucarióticos de Iniciación/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Modelos Biológicos , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal
8.
Genes Dev ; 35(17-18): 1243-1255, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34385262

RESUMEN

Multiple G protein-coupled receptors (GPCRs) are expressed in pancreatic islet cells, but the majority have unknown functions. We observed specific GPCRs localized to primary cilia, a prominent signaling organelle, in pancreatic α and ß cells. Loss of cilia disrupts ß-cell endocrine function, but the molecular drivers are unknown. Using functional expression, we identified multiple GPCRs localized to cilia in mouse and human islet α and ß cells, including FFAR4, PTGER4, ADRB2, KISS1R, and P2RY14. Free fatty acid receptor 4 (FFAR4) and prostaglandin E receptor 4 (PTGER4) agonists stimulate ciliary cAMP signaling and promote glucagon and insulin secretion by α- and ß-cell lines and by mouse and human islets. Transport of GPCRs to primary cilia requires TULP3, whose knockdown in primary human and mouse islets relocalized ciliary FFAR4 and PTGER4 and impaired regulated glucagon or insulin secretion, without affecting ciliary structure. Our findings provide index evidence that regulated hormone secretion by islet α and ß cells is controlled by ciliary GPCRs providing new targets for diabetes.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Glucagón/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Receptores Acoplados a Proteínas G/genética
9.
Genes Dev ; 35(3-4): 234-249, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33446570

RESUMEN

The physiological functions of many vital tissues and organs continue to mature after birth, but the genetic mechanisms governing this postnatal maturation remain an unsolved mystery. Human pancreatic ß cells produce and secrete insulin in response to physiological cues like glucose, and these hallmark functions improve in the years after birth. This coincides with expression of the transcription factors SIX2 and SIX3, whose functions in native human ß cells remain unknown. Here, we show that shRNA-mediated SIX2 or SIX3 suppression in human pancreatic adult islets impairs insulin secretion. However, transcriptome studies revealed that SIX2 and SIX3 regulate distinct targets. Loss of SIX2 markedly impaired expression of genes governing ß-cell insulin processing and output, glucose sensing, and electrophysiology, while SIX3 loss led to inappropriate expression of genes normally expressed in fetal ß cells, adult α cells, and other non-ß cells. Chromatin accessibility studies identified genes directly regulated by SIX2. Moreover, ß cells from diabetic humans with impaired insulin secretion also had reduced SIX2 transcript levels. Revealing how SIX2 and SIX3 govern functional maturation and maintain developmental fate in native human ß cells should advance ß-cell replacement and other therapeutic strategies for diabetes.


Asunto(s)
Diferenciación Celular/genética , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Proteínas del Tejido Nervioso/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Secreción de Insulina/genética , ARN Interferente Pequeño/metabolismo , Transcriptoma , Proteína Homeobox SIX3
10.
Nature ; 609(7929): 1021-1028, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36131014

RESUMEN

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Asunto(s)
Diferenciación Celular , Neoplasias Cerebelosas , Meduloblastoma , Metencéfalo , Diferenciación Celular/genética , Linaje de la Célula , Neoplasias Cerebelosas/clasificación , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Cerebelo/embriología , Cerebelo/patología , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Proteínas Hedgehog/metabolismo , Histona Demetilasas , Humanos , Antígeno Ki-67/metabolismo , Meduloblastoma/clasificación , Meduloblastoma/genética , Meduloblastoma/patología , Metencéfalo/embriología , Metencéfalo/patología , Proteínas Musculares , Mutación , Factores de Transcripción Otx/deficiencia , Factores de Transcripción Otx/genética , Proteínas Represoras , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción
11.
EMBO J ; 42(24): e114462, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-37934086

RESUMEN

Mammalian cells repress expression of repetitive genomic sequences by forming heterochromatin. However, the consequences of ectopic repeat expression remain unclear. Here we demonstrate that inhibitors of EZH2, the catalytic subunit of the Polycomb repressive complex 2 (PRC2), stimulate repeat misexpression and cell death in resting splenic B cells. B cells are uniquely sensitive to these agents because they exhibit high levels of histone H3 lysine 27 trimethylation (H3K27me3) and correspondingly low DNA methylation at repeat elements. We generated a pattern recognition receptor loss-of-function mouse model, called RIC, with mutations in Rigi (encoding for RIG-I), Ifih1 (MDA5), and Cgas. In both wildtype and RIC mutant B cells, EZH2 inhibition caused loss of H3K27me3 at repetitive elements and upregulated their expression. However, NF-κB-dependent expression of inflammatory chemokines and subsequent cell death was suppressed by the RIC mutations. We further show that inhibition of EZH2 in cancer cells requires the same pattern recognition receptors to activate an interferon response. Together, the results reveal chemokine expression induced by EZH2 inhibitors in B cells as a novel inflammatory response to genomic repeat expression. Given the overlap of genes induced by EZH2 inhibitors and Epstein-Barr virus infection, this response can be described as a form of viral mimicry.


Asunto(s)
Linfocitos B , Proteína Potenciadora del Homólogo Zeste 2 , Infecciones por Virus de Epstein-Barr , Animales , Ratones , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Histonas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos
12.
Immunity ; 49(2): 275-287.e5, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30054206

RESUMEN

Airway epithelial cells (AECs) secrete innate immune cytokines that regulate adaptive immune effector cells. In allergen-sensitized humans and mice, the airway and alveolar microenvironment is enriched with colony stimulating factor-1 (CSF1) in response to allergen exposure. In this study we found that AEC-derived CSF1 had a critical role in the production of allergen reactive-IgE production. Furthermore, spatiotemporally secreted CSF1 regulated the recruitment of alveolar dendritic cells (DCs) and enhanced the migration of conventional DC2s (cDC2s) to the draining lymph node in an interferon regulatory factor 4 (IRF4)-dependent manner. CSF1 selectively upregulated the expression of the chemokine receptor CCR7 on the CSF1R+ cDC2, but not the cDC1, population in response to allergen stimuli. Our data describe the functional specification of CSF1-dependent DC subsets that link the innate and adaptive immune responses in T helper 2 (Th2) cell-mediated allergic lung inflammation.


Asunto(s)
Alérgenos/inmunología , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Receptores CCR7/biosíntesis , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Animales , Línea Celular , Movimiento Celular/inmunología , Células Dendríticas/clasificación , Células Epiteliales/citología , Células Epiteliales/inmunología , Humanos , Inmunidad Innata/inmunología , Inmunoglobulina E/inmunología , Factores Reguladores del Interferón/inmunología , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células RAW 264.7 , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Th2/inmunología , Regulación hacia Arriba/inmunología
13.
Nature ; 592(7854): 428-432, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33790465

RESUMEN

Chronic, sustained exposure to stressors can profoundly affect tissue homeostasis, although the mechanisms by which these changes occur are largely unknown. Here we report that the stress hormone corticosterone-which is derived from the adrenal gland and is the rodent equivalent of cortisol in humans-regulates hair follicle stem cell (HFSC) quiescence and hair growth in mice. In the absence of systemic corticosterone, HFSCs enter substantially more rounds of the regeneration cycle throughout life. Conversely, under chronic stress, increased levels of corticosterone prolong HFSC quiescence and maintain hair follicles in an extended resting phase. Mechanistically, corticosterone acts on the dermal papillae to suppress the expression of Gas6, a gene that encodes the secreted factor growth arrest specific 6. Restoring Gas6 expression overcomes the stress-induced inhibition of HFSC activation and hair growth. Our work identifies corticosterone as a systemic inhibitor of HFSC activity through its effect on the niche, and demonstrates that the removal of such inhibition drives HFSCs into frequent regeneration cycles, with no observable defects in the long-term.


Asunto(s)
Corticosterona/farmacología , Folículo Piloso/citología , Células Madre/citología , Células Madre/efectos de los fármacos , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/cirugía , Adrenalectomía , Animales , División Celular/efectos de los fármacos , Femenino , Folículo Piloso/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Transcriptoma , Regulación hacia Arriba
14.
Nature ; 594(7863): 418-423, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33953400

RESUMEN

Although RAF monomer inhibitors (type I.5, BRAF(V600)) are clinically approved for the treatment of BRAFV600-mutant melanoma, they are ineffective in non-BRAFV600 mutant cells1-3. Belvarafenib is a potent and selective RAF dimer (type II) inhibitor that exhibits clinical activity in patients with BRAFV600E- and NRAS-mutant melanomas. Here we report the first-in-human phase I study investigating the maximum tolerated dose, and assessing the safety and preliminary efficacy of belvarafenib in BRAFV600E- and RAS-mutated advanced solid tumours (NCT02405065, NCT03118817). By generating belvarafenib-resistant NRAS-mutant melanoma cells and analysing circulating tumour DNA from patients treated with belvarafenib, we identified new recurrent mutations in ARAF within the kinase domain. ARAF mutants conferred resistance to belvarafenib in both a dimer- and a kinase activity-dependent manner. Belvarafenib induced ARAF mutant dimers, and dimers containing mutant ARAF were active in the presence of inhibitor. ARAF mutations may serve as a general resistance mechanism for RAF dimer inhibitors as the mutants exhibit reduced sensitivity to a panel of type II RAF inhibitors. The combination of RAF plus MEK inhibition may be used to delay ARAF-driven resistance and suggests a rational combination for clinical use. Together, our findings reveal specific and compensatory functions for the ARAF isoform and implicate ARAF mutations as a driver of resistance to RAF dimer inhibitors.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Mutación , Proteínas Proto-Oncogénicas A-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas A-raf/genética , Quinasas raf/antagonistas & inhibidores , Animales , Línea Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Melanoma/patología , Ratones , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas A-raf/química , Quinasas raf/química
15.
Mol Cell ; 73(1): 1-2, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30609388

RESUMEN

PD-L1 plays a central role in immune recognition of cancer cells. In this issue of Molecular Cell, Jin et al. (2019) report that a phosphorylated retinoblastoma protein contacts the DNA-binding domain of p65 NF-κB, thereby blocking transcription of PD-L1.


Asunto(s)
Antígeno B7-H1 , Factor de Transcripción ReIA/genética , Regulación de la Expresión Génica , FN-kappa B/genética , Proteína de Retinoblastoma , Transducción de Señal
16.
Proc Natl Acad Sci U S A ; 121(31): e2400687121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39042677

RESUMEN

The seemingly straightforward task of tying one's shoes requires a sophisticated interplay of joints, muscles, and neural pathways, posing a formidable challenge for researchers studying the intricacies of coordination. A widely accepted framework for measuring coordinated behavior is the Haken-Kelso-Bunz (HKB) model. However, a significant limitation of this model is its lack of accounting for the diverse variability structures inherent in the coordinated systems it frequently models. Variability is a pervasive phenomenon across various biological and physical systems, and it changes in healthy adults, older adults, and pathological populations. Here, we show, both empirically and with simulations, that manipulating the variability in coordinated movements significantly impacts the ability to change coordination patterns-a fundamental feature of the HKB model. Our results demonstrate that synchronized bimanual coordination, mirroring a state of healthy variability, instigates earlier transitions of coordinated movements compared to other variability conditions. This suggests a heightened adaptability when movements possess a healthy variability. We anticipate our study to show the necessity of adapting the HKB model to encompass variability, particularly in predictive applications such as neuroimaging, cognition, skill development, biomechanics, and beyond.


Asunto(s)
Movimiento , Desempeño Psicomotor , Humanos , Masculino , Femenino , Desempeño Psicomotor/fisiología , Adulto , Movimiento/fisiología , Fenómenos Biomecánicos , Adulto Joven , Mano/fisiología
17.
Genes Dev ; 33(11-12): 620-625, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30923167

RESUMEN

DOT1L is a histone H3 Lys79 methyltransferase whose activity is stimulated by histone H2B Lys120 ubiquitination, suggesting cross-talk between histone H3 methylation and H2B ubiquitination. Here, we present cryo-EM structures of DOT1L complexes with unmodified or H2B ubiquitinated nucleosomes, showing that DOT1L recognizes H2B ubiquitin and the H2A/H2B acidic patch through a C-terminal hydrophobic helix and an arginine anchor in DOT1L, respectively. Furthermore, the structures combined with single-molecule FRET experiments show that H2B ubiquitination enhances a noncatalytic function of the DOT1L-destabilizing nucleosome. These results establish the molecular basis of the cross-talk between H2B ubiquitination and H3 Lys79 methylation as well as nucleosome destabilization by DOT1L.


Asunto(s)
Histonas/química , Histonas/metabolismo , Metiltransferasas/química , Metiltransferasas/metabolismo , Nucleosomas/química , Nucleosomas/metabolismo , Arginina/metabolismo , Dominio Catalítico , Microscopía por Crioelectrón , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metilación , Modelos Moleculares , Estabilidad Proteica , Estructura Secundaria de Proteína , Ubiquitina/metabolismo , Ubiquitinación
18.
PLoS Pathog ; 20(7): e1012295, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39052544

RESUMEN

The emergence of drug-resistant Mycobacterium tuberculosis (M.tb) has led to the development of novel anti-tuberculosis (anti-TB) drugs. Common methods for testing the efficacy of new drugs, including two-dimensional cell culture models or animal models, have several limitations. Therefore, an appropriate model representative of the human organism is required. Here, we developed an M.tb infection model using human lung organoids (hLOs) and demonstrated that M.tb H37Rv can infect lung epithelial cells and human macrophages (hMφs) in hLOs. This novel M.tb infection model can be cultured long-term and split several times while maintaining a similar number of M.tb H37Rv inside the hLOs. Anti-TB drugs reduced the intracellular survival of M.tb in hLOs. Notably, M.tb growth in hLOs was effectively suppressed at each passage by rifampicin and bedaquiline. Furthermore, a reduction in inflammatory cytokine production and intracellular survival of M.tb were observed upon knockdown of MFN2 and HERPUD1 (host-directed therapeutic targets for TB) in our M.tb H37Rv-infected hLO model. Thus, the incorporation of hMφs and M.tb into hLOs provides a powerful strategy for generating an M.tb infection model. This model can effectively reflect host-pathogen interactions and be utilized to test the efficacy of anti-TB drugs and host-directed therapies.


Asunto(s)
Antituberculosos , Pulmón , Mycobacterium tuberculosis , Organoides , Humanos , Organoides/microbiología , Mycobacterium tuberculosis/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología , Macrófagos/microbiología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Células Epiteliales/microbiología
19.
PLoS Biol ; 21(6): e3002156, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37315086

RESUMEN

Bak is a critical executor of apoptosis belonging to the Bcl-2 protein family. Bak contains a hydrophobic groove where the BH3 domain of proapoptotic Bcl-2 family members can be accommodated, which initiates its activation. Once activated, Bak undergoes a conformational change to oligomerize, which leads to mitochondrial destabilization and the release of cytochrome c into the cytosol and eventual apoptotic cell death. In this study, we investigated the molecular aspects and functional consequences of the interaction between Bak and peroxisomal testis-specific 1 (Pxt1), a noncanonical BH3-only protein exclusively expressed in the testis. Together with various biochemical approaches, this interaction was verified and analyzed at the atomic level by determining the crystal structure of the Bak-Pxt1 BH3 complex. In-depth biochemical and cellular analyses demonstrated that Pxt1 functions as a Bak-activating proapoptotic factor, and its BH3 domain, which mediates direct intermolecular interaction with Bak, plays a critical role in triggering apoptosis. Therefore, this study provides a molecular basis for the Pxt1-mediated novel pathway for the activation of apoptosis and expands our understanding of the cell death signaling coordinated by diverse BH3 domain-containing proteins.


Asunto(s)
Proteínas Proto-Oncogénicas c-bcl-2 , Humanos , Masculino , Apoptosis/fisiología , Proteína X Asociada a bcl-2 , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteínas Portadoras/metabolismo , Mitocondrias/metabolismo
20.
EMBO Rep ; 25(7): 3008-3039, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38831125

RESUMEN

The circular RNA (circRNA) Cdr1as is conserved across mammals and highly expressed in neurons, where it directly interacts with microRNA miR-7. However, the biological function of this interaction is unknown. Here, using primary cortical murine neurons, we demonstrate that stimulating neurons by sustained depolarization rapidly induces two-fold transcriptional upregulation of Cdr1as and strong post-transcriptional stabilization of miR-7. Cdr1as loss causes doubling of glutamate release from stimulated synapses and increased frequency and duration of local neuronal bursts. Moreover, the periodicity of neuronal networks increases, and synchronicity is impaired. Strikingly, these effects are reverted by sustained expression of miR-7, which also clears Cdr1as molecules from neuronal projections. Consistently, without Cdr1as, transcriptomic changes caused by miR-7 overexpression are stronger (including miR-7-targets downregulation) and enriched in secretion/synaptic plasticity pathways. Altogether, our results suggest that in cortical neurons Cdr1as buffers miR-7 activity to control glutamatergic excitatory transmission and neuronal connectivity important for long-lasting synaptic adaptations.


Asunto(s)
Ácido Glutámico , MicroARNs , Neuronas , Transmisión Sináptica , MicroARNs/genética , MicroARNs/metabolismo , Animales , Neuronas/metabolismo , Ratones , Ácido Glutámico/metabolismo , Transmisión Sináptica/genética , Plasticidad Neuronal/genética , ARN Circular/genética , ARN Circular/metabolismo , Sinapsis/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación de la Expresión Génica , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA