Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 4.987
Filtrar
Más filtros

Publication year range
1.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34214472

RESUMEN

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Asunto(s)
Antígenos CD28/deficiencia , Patrón de Herencia/genética , Papillomaviridae/fisiología , Piel/virología , Linfocitos T/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/inmunología , Niño , Endopeptidasas/metabolismo , Femenino , Genes Recesivos , Células HEK293 , Homocigoto , Humanos , Inmunidad Humoral , Memoria Inmunológica , Células Jurkat , Queratinocitos/patología , Masculino , Ratones Endogámicos C57BL , Oncogenes , Papiloma/patología , Papiloma/virología , Linaje , Señales de Clasificación de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Cell ; 184(15): 3998-4015.e19, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34157302

RESUMEN

Foxp3+ T regulatory (Treg) cells promote immunological tumor tolerance, but how their immune-suppressive function is regulated in the tumor microenvironment (TME) remains unknown. Here, we used intravital microscopy to characterize the cellular interactions that provide tumor-infiltrating Treg cells with critical activation signals. We found that the polyclonal Treg cell repertoire is pre-enriched to recognize antigens presented by tumor-associated conventional dendritic cells (cDCs). Unstable cDC contacts sufficed to sustain Treg cell function, whereas T helper cells were activated during stable interactions. Contact instability resulted from CTLA-4-dependent downregulation of co-stimulatory B7-family proteins on cDCs, mediated by Treg cells themselves. CTLA-4-blockade triggered CD28-dependent Treg cell hyper-proliferation in the TME, and concomitant Treg cell inactivation was required to achieve tumor rejection. Therefore, Treg cells self-regulate through a CTLA-4- and CD28-dependent feedback loop that adjusts their population size to the amount of local co-stimulation. Its disruption through CTLA-4-blockade may off-set therapeutic benefits in cancer patients.


Asunto(s)
Antígeno CTLA-4/metabolismo , Retroalimentación Fisiológica , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD28/metabolismo , Proliferación Celular , Células Dendríticas/inmunología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Interleucina-2/metabolismo , Ligandos , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Microambiente Tumoral
3.
Immunity ; 56(6): 1187-1203.e12, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37160118

RESUMEN

B7 ligands (CD80 and CD86), expressed by professional antigen-presenting cells (APCs), activate the main co-stimulatory receptor CD28 on T cells in trans. However, in peripheral tissues, APCs expressing B7 ligands are relatively scarce. This raises the questions of whether and how CD28 co-stimulation occurs in peripheral tissues. Here, we report that CD8+ T cells displayed B7 ligands that interacted with CD28 in cis at membrane invaginations of the immunological synapse as a result of membrane remodeling driven by phosphoinositide-3-kinase (PI3K) and sorting-nexin-9 (SNX9). cis-B7:CD28 interactions triggered CD28 signaling through protein kinase C theta (PKCθ) and promoted CD8+ T cell survival, migration, and cytokine production. In mouse tumor models, loss of T cell-intrinsic cis-B7:CD28 interactions decreased intratumoral T cells and accelerated tumor growth. Thus, B7 ligands on CD8+ T cells can evoke cell-autonomous CD28 co-stimulation in cis in peripheral tissues, suggesting cis-signaling as a general mechanism for boosting T cell functionality.


Asunto(s)
Antígenos CD28 , Linfocitos T CD8-positivos , Ratones , Animales , Antígenos CD28/metabolismo , Antígenos CD/metabolismo , Ligandos , Membranas Sinápticas/metabolismo , Antígeno B7-2 , Glicoproteínas de Membrana/metabolismo , Antígeno B7-1/metabolismo , Moléculas de Adhesión Celular , Activación de Linfocitos
4.
Immunity ; 56(9): 2121-2136.e6, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37659412

RESUMEN

Genetic association studies have demonstrated the critical involvement of the microglial immune response in Alzheimer's disease (AD) pathogenesis. Phospholipase C-gamma-2 (PLCG2) is selectively expressed by microglia and functions in many immune receptor signaling pathways. In AD, PLCG2 is induced uniquely in plaque-associated microglia. A genetic variant of PLCG2, PLCG2P522R, is a mild hypermorph that attenuates AD risk. Here, we identified a loss-of-function PLCG2 variant, PLCG2M28L, that confers an increased AD risk. PLCG2P522R attenuated disease in an amyloidogenic murine AD model, whereas PLCG2M28L exacerbated the plaque burden associated with altered phagocytosis and Aß clearance. The variants bidirectionally modulated disease pathology by inducing distinct transcriptional programs that identified microglial subpopulations associated with protective or detrimental phenotypes. These findings identify PLCG2M28L as a potential AD risk variant and demonstrate that PLCG2 variants can differentially orchestrate microglial responses in AD pathogenesis that can be therapeutically targeted.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/genética , Estudios de Asociación Genética , Microglía , Fagocitosis/genética , Fenotipo , Placa Amiloide , Fosfolipasa C gamma/metabolismo
5.
Cell ; 171(2): 385-397.e11, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28919076

RESUMEN

T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.


Asunto(s)
Antígenos CD28/metabolismo , Activación de Linfocitos , Mitocondrias/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Carnitina O-Palmitoiltransferasa , Inhibidores Enzimáticos/farmacología , Compuestos Epoxi/farmacología , Humanos , Interleucina-15/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo , Estrés Fisiológico , Linfocitos T/metabolismo
6.
Annu Rev Biochem ; 85: 103-32, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27023846

RESUMEN

Mitochondrial ribosomes (mitoribosomes) perform protein synthesis inside mitochondria, the organelles responsible for energy conversion and adenosine triphosphate production in eukaryotic cells. Throughout evolution, mitoribosomes have become functionally specialized for synthesizing mitochondrial membrane proteins, and this has been accompanied by large changes to their structure and composition. We review recent high-resolution structural data that have provided unprecedented insight into the structure and function of mitoribosomes in mammals and fungi.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Ribosomas Mitocondriales/ultraestructura , Biosíntesis de Proteínas , Subunidades Ribosómicas/ultraestructura , Animales , Antibacterianos/farmacología , Evolución Biológica , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , ADN Mitocondrial/metabolismo , Mamíferos , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Ribosomas Mitocondriales/química , Ribosomas Mitocondriales/metabolismo , Modelos Moleculares , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Subunidades Ribosómicas/química , Subunidades Ribosómicas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
7.
Mol Cell ; 83(12): 1970-1982.e6, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37327775

RESUMEN

Pioneer transcription factors are essential for cell fate changes by targeting closed chromatin. OCT4 is a crucial pioneer factor that can induce cell reprogramming. However, the structural basis of how pioneer factors recognize the in vivo nucleosomal DNA targets is unknown. Here, we determine the high-resolution structures of the nucleosome containing human LIN28B DNA and its complexes with the OCT4 DNA binding region. Three OCT4s bind the pre-positioned nucleosome by recognizing non-canonical DNA sequences. Two use their POUS domains while the other uses the POUS-loop-POUHD region; POUHD serves as a wedge to unwrap ∼25 base pair DNA. Our analysis of previous genomic data and determination of the ESRRB-nucleosome-OCT4 structure confirmed the generality of these structural features. Moreover, biochemical studies suggest that multiple OCT4s cooperatively open the H1-condensed nucleosome array containing the LIN28B nucleosome. Thus, our study suggests a mechanism of how OCT4 can target the nucleosome and open closed chromatin.


Asunto(s)
Cromatina , Nucleosomas , Factor 3 de Transcripción de Unión a Octámeros , Proteínas de Unión al ARN , Humanos , Secuencia de Bases , Reprogramación Celular , Cromatina/genética , ADN/metabolismo , Nucleosomas/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
8.
Mol Cell ; 83(7): 1093-1108.e8, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36863348

RESUMEN

The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation.


Asunto(s)
Glucagón , Gluconeogénesis , Animales , Ratones , Gluconeogénesis/genética , Glucagón/metabolismo , Histonas/metabolismo , Fosforilación , Proteínas 14-3-3/metabolismo , Hígado/metabolismo , Ayuno/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
9.
Immunity ; 52(2): 313-327.e7, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32049052

RESUMEN

T cell responses upon infection display a remarkably reproducible pattern of expansion, contraction, and memory formation. If the robustness of this pattern builds entirely on signals derived from other cell types or if activated T cells themselves contribute to the orchestration of these population dynamics-akin to bacterial quorum regulation-is unclear. Here, we examined this question using time-lapse microscopy, genetic perturbation, bioinformatic predictions, and mathematical modeling. We found that ICAM-1-mediated cell clustering enabled CD8+ T cells to collectively regulate the balance between proliferation and apoptosis. Mechanistically, T cell expressed CD80 and CD86 interacted with the receptors CD28 and CTLA-4 on neighboring T cells; these interactions fed two nested antagonistic feedback circuits that regulated interleukin 2 production in a manner dependent on T cell density as confirmed by in vivo modulation of this network. Thus, CD8+ T cell-population-intrinsic mechanisms regulate cellular behavior, thereby promoting robustness of population dynamics.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/metabolismo , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular , Recuento de Células , Línea Celular , Supervivencia Celular , Rastreo Celular , Células Dendríticas/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-2/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Teóricos
10.
Immunity ; 51(6): 1059-1073.e9, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31757674

RESUMEN

Combined immunotherapy targeting the immune checkpoint receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1), or CTLA-4 and the PD-1 ligand (PD-L1) exhibits superior anti-tumor responses compared with single-agent therapy. Here, we examined the molecular basis for this synergy. Using reconstitution assays with fluorescence readouts, we found that PD-L1 and the CTLA-4 ligand CD80 heterodimerize in cis but not trans. Quantitative biochemistry and cell biology assays revealed that PD-L1:CD80 cis-heterodimerization inhibited both PD-L1:PD-1 and CD80:CTLA-4 interactions through distinct mechanisms but preserved the ability of CD80 to activate the T cell co-stimulatory receptor CD28. Furthermore, PD-L1 expression on antigen-presenting cells (APCs) prevented CTLA-4-mediated trans-endocytosis of CD80. Atezolizumab (anti-PD-L1), but not anti-PD-1, reduced cell surface expression of CD80 on APCs, and this effect was negated by co-blockade of CTLA-4 with ipilimumab (anti-CTLA-4). Thus, PD-L1 exerts an immunostimulatory effect by repressing the CTLA-4 axis; this has implications to the synergy of anti-PD-L1 and anti-CTLA-4 combination therapy.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD28/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunoterapia/métodos , Ipilimumab/farmacología , Células Jurkat , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
11.
Mol Cell ; 77(5): 970-984.e7, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31982308

RESUMEN

Cytosolic caspase-8 is a mediator of death receptor signaling. While caspase-8 expression is lost in some tumors, it is increased in others, indicating a conditional pro-survival function of caspase-8 in cancer. Here, we show that tumor cells employ DNA-damage-induced nuclear caspase-8 to override the p53-dependent G2/M cell-cycle checkpoint. Caspase-8 is upregulated and localized to the nucleus in multiple human cancers, correlating with treatment resistance and poor clinical outcome. Depletion of caspase-8 causes G2/M arrest, stabilization of p53, and induction of p53-dependent intrinsic apoptosis in tumor cells. In the nucleus, caspase-8 cleaves and inactivates the ubiquitin-specific peptidase 28 (USP28), preventing USP28 from de-ubiquitinating and stabilizing wild-type p53. This results in de facto p53 protein loss, switching cell fate from apoptosis toward mitosis. In summary, our work identifies a non-canonical role of caspase-8 exploited by cancer cells to override the p53-dependent G2/M cell-cycle checkpoint.


Asunto(s)
Caspasa 8/metabolismo , Núcleo Celular/enzimología , Proliferación Celular , Puntos de Control de la Fase G2 del Ciclo Celular , Neoplasias/enzimología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Antineoplásicos/farmacología , Apoptosis , Caspasa 8/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Masculino , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Células PC-3 , Estabilidad Proteica , Transducción de Señal , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ubiquitina Tiolesterasa/genética
12.
Mol Cell ; 78(6): 1133-1151.e14, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32402252

RESUMEN

Precise control of the RNA polymerase II (RNA Pol II) cycle, including pausing and pause release, maintains transcriptional homeostasis and organismal functions. Despite previous work to understand individual transcription steps, we reveal a mechanism that integrates RNA Pol II cycle transitions. Surprisingly, KAP1/TRIM28 uses a previously uncharacterized chromatin reader cassette to bind hypo-acetylated histone 4 tails at promoters, guaranteeing continuous progression of RNA Pol II entry to and exit from the pause state. Upon chromatin docking, KAP1 first associates with RNA Pol II and then recruits a pathway-specific transcription factor (SMAD2) in response to cognate ligands, enabling gene-selective CDK9-dependent pause release. This coupling mechanism is exploited by tumor cells to aberrantly sustain transcriptional programs commonly dysregulated in cancer patients. The discovery of a factor integrating transcription steps expands the functional repertoire by which chromatin readers operate and provides mechanistic understanding of transcription regulation, offering alternative therapeutic opportunities to target transcriptional dysregulation.


Asunto(s)
ARN Polimerasa II/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Acetilación , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Regulación de la Expresión Génica/genética , Histonas/metabolismo , Humanos , Oncogenes/genética , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/genética , ARN Polimerasa II/genética , Proteína Smad2/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína 28 que Contiene Motivos Tripartito/genética
13.
EMBO J ; 42(6): e111858, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36562188

RESUMEN

Phytosulfokine (PSK) is a plant pentapeptide hormone that fulfills a wide range of functions. Although PSK has frequently been reported to function in the inverse regulation of growth and defense in response to (hemi)biotrophic pathogens, the mechanisms involved remain largely unknown. Using the tomato (Solanum lycopersicum) and Pseudomonas syringae pv. tomato (Pst) DC3000 pathogen system, we present compelling evidence that the PSK receptor PSKR1 interacts with the calcium-dependent protein kinase CPK28, which in turn phosphorylates the key enzyme of nitrogen assimilation glutamine synthetase GS2 at two sites (Serine-334 and Serine-360). GS2 phosphorylation at S334 specifically regulates plant defense, whereas S360 regulates growth, uncoupling the PSK-induced effects on defense responses and growth regulation. The discovery of these sites will inform breeding strategies designed to optimize the growth-defense balance in a compatible manner.


Asunto(s)
Solanum lycopersicum , Fosforilación , Glutamato-Amoníaco Ligasa/metabolismo , Péptidos/metabolismo , Reguladores del Crecimiento de las Plantas
14.
Mol Cell ; 75(5): 1073-1085.e6, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31327635

RESUMEN

Mitochondrial AAA+ quality-control proteases regulate diverse aspects of mitochondrial biology through specialized protein degradation, but the underlying mechanisms of these enzymes remain poorly defined. The mitochondrial AAA+ protease AFG3L2 is of particular interest, as genetic mutations localized throughout AFG3L2 are linked to diverse neurodegenerative disorders. However, a lack of structural data has limited our understanding of how mutations impact enzymatic function. Here, we used cryoelectron microscopy (cryo-EM) to determine a substrate-bound structure of the catalytic core of human AFG3L2. This structure identifies multiple specialized structural features that integrate with conserved motifs required for ATP-dependent translocation to unfold and degrade targeted proteins. Many disease-relevant mutations localize to these unique structural features of AFG3L2 and distinctly influence its activity and stability. Our results provide a molecular basis for neurological phenotypes associated with different AFG3L2 mutations and establish a structural framework to understand how different members of the AAA+ superfamily achieve specialized biological functions.


Asunto(s)
Proteasas ATP-Dependientes/química , ATPasas Asociadas con Actividades Celulares Diversas/química , Proteínas Mitocondriales/química , Mutación , Proteasas ATP-Dependientes/genética , Proteasas ATP-Dependientes/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Microscopía por Crioelectrón , Células HEK293 , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Dominios Proteicos
15.
Mol Cell ; 74(3): 421-435.e10, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30926243

RESUMEN

Deubiquitinases have emerged as promising drug targets for cancer therapy. The two DUBs USP25 and USP28 share high similarity but vary in their cellular functions. USP28 is known for its tumor-promoting role, whereas USP25 is a regulator of the innate immune system and, recently, a role in tumorigenesis was proposed. We solved the structures of the catalytic domains of both proteins and established substantial differences in their activities. While USP28 is a constitutively active dimer, USP25 presents an auto-inhibited tetramer. Our data indicate that the activation of USP25 is not achieved through substrate or ubiquitin binding. USP25 cancer-associated mutations lead to activation in vitro and in vivo, thereby providing a functional link between auto-inhibition and the cancer-promoting role of the enzyme. Our work led to the identification of significant differences between USP25 and USP28 and provided the molecular basis for the development of new and highly specific anti-cancer drugs.


Asunto(s)
Carcinogénesis/genética , Neoplasias/genética , Ubiquitina Tiolesterasa/genética , Secuencia de Aminoácidos/genética , Dominio Catalítico/genética , Enzimas Desubicuitinizantes/química , Enzimas Desubicuitinizantes/genética , Humanos , Mutación/genética , Neoplasias/tratamiento farmacológico , Unión Proteica/genética , Conformación Proteica , Multimerización de Proteína/genética , Ubiquitina/genética , Ubiquitina Tiolesterasa/química
16.
Mol Cell ; 73(2): 304-313.e3, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30527666

RESUMEN

LIN28 RNA binding proteins are dynamically expressed throughout mammalian development and during disease. However, it remains unclear how changes in LIN28 expression define patterns of post-transcriptional gene regulation. Here we show that LIN28 expression level is a key variable that sets the magnitude of protein translation. By systematically varying LIN28B protein levels in human cells, we discovered a dose-dependent divergence in transcriptome-wide ribosome occupancy that enabled the formation of two discrete translational subpopulations composed of nearly all expressed genes. This bifurcation in gene expression was mediated by a redistribution in Argonaute association, from let-7 to non-let-7 microRNA families, resulting in a global shift in cellular miRNA activity. Post-transcriptional effects were scaled across the physiological LIN28 expression range. Together, these data highlight the central importance of RBP expression level and its ability to encode regulation.


Asunto(s)
Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Ribosomas/metabolismo , Transcriptoma , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sitios de Unión , Unión Competitiva , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Células 3T3 NIH , Unión Proteica , Proteínas de Unión al ARN/genética , Ribosomas/genética
17.
Mol Cell ; 76(4): 646-659.e6, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31543422

RESUMEN

Eukaryotic chromosomes contain compartments of various functions, which are marked by and enriched with specific histone modifications. However, the molecular mechanisms by which these histone marks function in chromosome compartmentalization are poorly understood. Constitutive heterochromatin is a largely silent chromosome compartment characterized in part by H3K9me2 and 3. Here, we show that heterochromatin protein 1 (HP1), an H3K9me2 and 3 "reader," interacts with SUV39H1, an H3K9me2 and 3 "writer," and with TRIM28, an abundant HP1 scaffolding protein, to form complexes with increased multivalent engagement of H3K9me2 and 3-modified chromatin. H3K9me2 and 3-marked nucleosomal arrays and associated complexes undergo phase separation to form macromolecule-enriched liquid droplets. The droplets are reminiscent of heterochromatin as they are highly dense chromatin-containing structures that are resistant to DNase and exclude the general transcription factor TFIIB. Our data suggest a general mechanism by which histone marks regulate chromosome compartmentalization by promoting phase separation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Heterocromatina/metabolismo , Histonas/metabolismo , Gotas Lipídicas/metabolismo , Nucleosomas/metabolismo , Procesamiento Proteico-Postraduccional , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Células HEK293 , Heterocromatina/genética , Humanos , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Complejos Multiproteicos , Nucleosomas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Tiempo , Proteína 28 que Contiene Motivos Tripartito/genética , Proteína 28 que Contiene Motivos Tripartito/metabolismo
18.
RNA ; 30(7): 770-778, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38570183

RESUMEN

30S subunits become inactive upon exposure to low Mg2+ concentration, because of a reversible conformational change that entails nucleotides (nt) in the neck helix (h28) and 3' tail of 16S rRNA. This active-to-inactive transition involves partial unwinding of h28 and repairing of nt 921-923 with nt 1532-1534, which requires flipping of the 3' tail by ∼180°. Growing evidence suggests that immature 30S particles adopt the inactive conformation in the cell, and transition to the active state occurs at a late stage of maturation. Here, we target nucleotides that form the alternative helix (hALT) of the inactive state. Using an orthogonal ribosome system, we find that disruption of hALT decreases translation activity in the cell modestly, by approximately twofold, without compromising ribosome fidelity. Ribosomes carrying substitutions at positions 1532-1533 support the growth of Escherichia coli strain Δ7 prrn (which carries a single rRNA operon), albeit at rates 10%-20% slower than wild-type ribosomes. These mutant Δ7 prrn strains accumulate free 30S particles and precursor 17S rRNA, indicative of biogenesis defects. Analysis of purified control and mutant subunits suggests that hALT stabilizes the inactive state by 1.2 kcal/mol with little-to-no impact on the active state or the transition state of conversion.


Asunto(s)
Escherichia coli , Conformación de Ácido Nucleico , ARN Ribosómico 16S , Subunidades Ribosómicas Pequeñas Bacterianas , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Subunidades Ribosómicas Pequeñas Bacterianas/metabolismo , Subunidades Ribosómicas Pequeñas Bacterianas/genética , Biosíntesis de Proteínas , Magnesio/metabolismo
19.
Immunity ; 47(5): 875-889.e10, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166588

RESUMEN

Migration of activated regulatory T (Treg) cells to inflamed tissue is crucial for their immune-modulatory function. While metabolic reprogramming during Treg cell differentiation has been extensively studied, the bioenergetics of Treg cell trafficking remains undefined. We have investigated the metabolic demands of migrating Treg cells in vitro and in vivo. We show that glycolysis was instrumental for their migration and was initiated by pro-migratory stimuli via a PI3K-mTORC2-mediated pathway culminating in induction of the enzyme glucokinase (GCK). Subsequently, GCK promoted cytoskeletal rearrangements by associating with actin. Treg cells lacking this pathway were functionally suppressive but failed to migrate to skin allografts and inhibit rejection. Similarly, human carriers of a loss-of-function GCK regulatory protein gene-leading to increased GCK activity-had reduced numbers of circulating Treg cells. These cells displayed enhanced migratory activity but similar suppressive function, while conventional T cells were unaffected. Thus, GCK-dependent glycolysis regulates Treg cell migration.


Asunto(s)
Glucoquinasa/fisiología , Glucólisis , Linfocitos T Reguladores/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD28/fisiología , Antígeno CTLA-4/fisiología , Células Cultivadas , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Ratones , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
20.
EMBO Rep ; 25(7): 2950-2973, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38816515

RESUMEN

The development of cancer therapeutics is often hindered by the fact that specific oncogenes cannot be directly pharmaceutically addressed. Targeting deubiquitylases that stabilize these oncogenes provides a promising alternative. USP28 and USP25 have been identified as such target deubiquitylases, and several small-molecule inhibitors indiscriminately inhibiting both enzymes have been developed. To obtain insights into their mode of inhibition, we structurally and functionally characterized USP28 in the presence of the three different inhibitors AZ1, Vismodegib and FT206. The compounds bind into a common pocket acting as a molecular sink. Our analysis provides an explanation why the two enzymes are inhibited with similar potency while other deubiquitylases are not affected. Furthermore, a key glutamate residue at position 366/373 in USP28/USP25 plays a central structural role for pocket stability and thereby for inhibition and activity. Obstructing the inhibitor-binding pocket by mutation of this glutamate may provide a tool to accelerate future drug development efforts for selective inhibitors of either USP28 or USP25 targeting distinct binding pockets.


Asunto(s)
Ubiquitina Tiolesterasa , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Humanos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Sitios de Unión , Piridinas/química , Piridinas/farmacología , Unión Proteica , Modelos Moleculares
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda