Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cell ; 82(20): 3826-3839.e9, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36113481

RESUMEN

Ribosomal RNAs (rRNAs) are the most abundant cellular RNAs, and their synthesis from rDNA repeats by RNA polymerase I accounts for the bulk of all transcription. Despite substantial variation in rRNA transcription rates across cell types, little is known about cell-type-specific factors that bind rDNA and regulate rRNA transcription to meet tissue-specific needs. Using hematopoiesis as a model system, we mapped about 2,200 ChIP-seq datasets for 250 transcription factors (TFs) and chromatin proteins to human and mouse rDNA and identified robust binding of multiple TF families to canonical TF motifs on rDNA. Using a 47S-FISH-Flow assay developed for nascent rRNA quantification, we demonstrated that targeted degradation of C/EBP alpha (CEBPA), a critical hematopoietic TF with conserved rDNA binding, caused rapid reduction in rRNA transcription due to reduced RNA Pol I occupancy. Our work identifies numerous potential rRNA regulators and provides a template for dissection of TF roles in rRNA transcription.


Asunto(s)
ARN Polimerasa I , Factores de Transcripción , Humanos , Ratones , Animales , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , ARN Ribosómico/genética , Transcripción Genética , ADN Ribosómico/genética , ARN , Cromatina
2.
Genes Dev ; 36(15-16): 871-873, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36207141

RESUMEN

Bmi1 is essential for normal and leukemic hematopoiesis, but its target genes in hematopoietic stem cells (HSCs) are incompletely understood. In this issue of Genes & Development, Burgess et al. (pp. 887-900) demonstrate a novel role of Bmi1 in regulating ribosome biogenesis and protein synthesis. Bmi1-deficient HSCs exhibited reduced transplantability, with the up-regulation of ARX and genes involved in ribosome biogenesis. However, depletion of ARX or its known targets, p16 Ink4a /p19 Arf , only partially rescues Bmi1 loss-induced hematopoietic defects. They further demonstrate an increased protein synthesis rate and resultant proteostatic stress in Bmi1 -/- HSCs, indicating a novel mechanism by which Bmi1 controls HSC maintenance.


Asunto(s)
Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
3.
Genes Dev ; 31(10): 1007-1023, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28611190

RESUMEN

Janus kinase 2 (JAK2) is a central kinase in hematopoietic stem/progenitor cells (HSPCs), and its uncontrolled activation is a prominent oncogenic driver of hematopoietic neoplasms. However, molecular mechanisms underlying the regulation of JAK2 have remained elusive. Here we report that the Casitas B-cell lymphoma (CBL) family E3 ubiquitin ligases down-regulate JAK2 stability and signaling via the adaptor protein LNK/SH2B3. We demonstrated that depletion of CBL/CBL-B or LNK abrogated JAK2 ubiquitination, extended JAK2 half-life, and enhanced JAK2 signaling and cell growth in human cell lines as well as primary murine HSPCs. Built on these findings, we showed that JAK inhibitor (JAKi) significantly reduced aberrant HSPCs and mitigated leukemia development in a mouse model of aggressive myeloid leukemia driven by loss of Cbl and Cbl-b Importantly, primary human CBL mutated (CBLmut ) leukemias exhibited increased JAK2 protein levels and signaling and were hypersensitive to JAKi. Loss-of-function mutations in CBL E3 ubiquitin ligases are found in a wide range of myeloid malignancies, which are diseases without effective treatment options. Hence, our studies reveal a novel signaling axis that regulates JAK2 in normal and malignant HSPCs and suggest new therapeutic strategies for treating CBLmut myeloid malignancies.


Asunto(s)
Janus Quinasa 2/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/fisiopatología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Citocinas/metabolismo , Estabilidad de Enzimas , Células Madre Hematopoyéticas/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Janus Quinasa 2/genética , Leucemia Mieloide Aguda/genética , Proteínas de la Membrana , Ratones , Mutación , Proteolisis , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal/genética , Ubiquitinación
4.
Cell Mol Life Sci ; 80(8): 223, 2023 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-37480504

RESUMEN

Kindlin-2 is critical for development and homeostasis of key organs, including skeleton, liver, islet, etc., yet its role in modulating angiogenesis is unknown. Here, we report that sufficient KINDLIN-2 is extremely important for NOTCH-mediated physiological angiogenesis. The expression of KINDLIN-2 in HUVECs is significantly modulated by angiogenic factors such as vascular endothelial growth factor A or tumor necrosis factor α. A strong co-localization of CD31 and Kindlin-2 in tissue sections is demonstrated by immunofluorescence staining. Endothelial-cell-specific Kindlin-2 deletion embryos die on E10.5 due to hemorrhage caused by the impaired physiological angiogenesis. Experiments in vitro show that vascular endothelial growth factor A-induced multiple functions of endothelial cells, including migration, matrix proteolysis, morphogenesis and sprouting, are all strengthened by KINDLIN-2 overexpression and severely impaired in the absence of KINDLIN-2. Mechanistically, we demonstrate that KINDLIN-2 inhibits the release of Notch intracellular domain through binding to and maintaining the integrity of NOTCH1. The impaired angiogenesis and avascular retinas caused by KINDLIN-2 deficiency can be rescued by DAPT, an inhibitor of γ-secretase which releases the intracellular domain from NOTCH1. Moreover, we demonstrate that high glucose stimulated hyperactive angiogenesis by increasing KINDLIN-2 expression could be prevented by KINDLIN-2 knockdown, indicating Kindlin-2 as a potential therapeutic target in treatment of diabetic retinopathy. Our study for the first time demonstrates the significance of Kindlin-2 in determining Notch-mediated angiogenesis during development and highlights Kindlin-2 as the potential therapeutic target in angiogenic diseases, such as diabetic retinopathy.


Asunto(s)
Retinopatía Diabética , Humanos , Fenómenos Fisiológicos Cardiovasculares , Células Endoteliales , Morfogénesis , Factor A de Crecimiento Endotelial Vascular/genética
5.
Blood ; 138(22): 2244-2255, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34111291

RESUMEN

Internal tandem duplication within FLT3 (FLT3-ITD) is one of the most frequent mutations in acute myeloid leukemia (AML) and correlates with a poor prognosis. Whereas the FLT3 receptor tyrosine kinase is activated at the plasma membrane to transduce PI3K/AKT and RAS/MAPK signaling, FLT3-ITD resides in the endoplasmic reticulum and triggers constitutive STAT5 phosphorylation. Mechanisms underlying this aberrant FLT3-ITD subcellular localization or its impact on leukemogenesis remain poorly established. In this study, we discovered that FLT3-ITD is S-palmitoylated by the palmitoyl acyltransferase ZDHHC6. Disruption of palmitoylation redirected FLT3-ITD to the plasma membrane and rewired its downstream signaling by activating AKT and extracellular signal-regulated kinase pathways in addition to STAT5. Consequently, abrogation of palmitoylation increased FLT3-ITD-mediated progression of leukemia in xenotransplant-recipient mouse models. We further demonstrate that FLT3 proteins were palmitoylated in primary human AML cells. ZDHHC6-mediated palmitoylation restrained FLT3-ITD surface expression, signaling, and colonogenic growth of primary FLT3-ITD+ AML. More important, pharmacological inhibition of FLT3-ITD depalmitoylation synergized with the US Food and Drug Administration-approved FLT3 kinase inhibitor gilteritinib in abrogating the growth of primary FLT3-ITD+ AML cells. These findings provide novel insights into lipid-dependent compartmentalization of FLT3-ITD signaling in AML and suggest targeting depalmitoylation as a new therapeutic strategy to treat FLT3-ITD+ leukemias.


Asunto(s)
Leucemia/patología , Lipoilación , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Duplicación de Gen , Humanos , Leucemia/genética , Leucemia/metabolismo , Ratones SCID , Tirosina Quinasa 3 Similar a fms/genética
6.
Blood ; 133(14): 1560-1571, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30755420

RESUMEN

Hematopoietic stem cell (HSC) homeostasis is controlled by cytokine receptor-mediated Janus kinase 2 (JAK2) signaling. We previously found that JAK2 is promptly ubiquitinated upon cytokine stimulation. Whether a competing JAK2 deubiquitination activity exists is unknown. LNK is an essential adaptor protein that constrains HSC expansion through dampening thrombopoietin (TPO)-induced JAK2 signaling. We show here that a LNK-associated lysine-63 (K63)-deubiquitinating enzyme complex, Brcc36 isopeptidase complex (BRISC), attenuates HSC expansion through control of JAK2 signaling. We pinpoint a direct interaction between the LNK SH2 domain and a phosphorylated tyrosine residue in KIAA0157 (Abraxas2), a unique and defining BRISC component. Kiaa0157 deficiency in mice led to an expansion of phenotypic and functional HSCs. Endogenous JAK2 and phospho-JAK2 were rapidly K63-ubiquitinated upon TPO stimulation, and this action was augmented in cells depleted of the BRISC core components KIAA0157, MERIT40, or BRCC36. This increase in JAK2 ubiquitination after BRISC knockdown was associated with increased TPO-mediated JAK2 activation and protein levels, and increased MPL receptor presence at the cell surface. In addition, BRISC depletion promoted membrane proximal association between the MPL receptor and pJAK2/JAK2, thus enhancing activated JAK2/MPL at the cell membrane. These findings define a novel pathway by which K63-ubiquitination promotes JAK2 stability and activation in a proteasome-independent manner. Moreover, mutations in BRCC36 are found in clonal hematopoiesis in humans. This research may shed light on the mechanistic understanding of a potential role of BRCC36 in human HSCs.


Asunto(s)
Proliferación Celular , Enzimas Desubicuitinizantes/fisiología , Células Madre Hematopoyéticas/citología , Janus Quinasa 2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Enzimas Desubicuitinizantes/genética , Humanos , Ratones , Proteínas Asociadas a Matriz Nuclear/metabolismo , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Trombopoyetina/farmacología , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación , Dominios Homologos src
7.
Blood ; 142(21): 1766-1768, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37995103
9.
J Neurosci ; 35(6): 2559-71, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25673849

RESUMEN

Thyroid receptor-interacting protein 6 (Trip6), a multifunctional protein belonging to the zyxin family of LIM proteins, is involved in various physiological and pathological processes, including cell migration and tumorigenesis. However, the role of Trip6 in neurons remains unknown. Here, we show that Trip6 is expressed in mouse hippocampal neurons and promotes dendritic morphogenesis. Through interaction with the glutamate receptor-interacting protein 1 (GRIP1) and myosin VI, Trip6 is crucial for the total dendritic length and the number of primary dendrites in cultured hippocampal neurons. Trip6 depletion reduces F-actin content and impairs dendritic morphology, and this phenocopies GRIP1 or myosin VI knockdown. Furthermore, phosphorylation of GRIP1(956T) by AKT1 inhibits the interaction between GRIP1 and myosin VI, but facilitates GRIP1 binding to 14-3-3 protein, which is required for regulating F-actin organization and dendritic morphogenesis. Thus, the Trip6-GRIP1-myosin VI interaction and its regulation on F-actin network play a significant role in dendritic morphogenesis.


Asunto(s)
Actinas/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Portadoras/fisiología , Dendritas/ultraestructura , Proteínas con Dominio LIM/fisiología , Cadenas Pesadas de Miosina/fisiología , Proteínas del Tejido Nervioso/fisiología , Factores de Transcripción/fisiología , Proteínas 14-3-3/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Células HEK293 , Hipocampo/citología , Hipocampo/ultraestructura , Humanos , Morfogénesis , Neuronas/ultraestructura , Fosforilación , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas c-akt/metabolismo
10.
J Biol Chem ; 289(49): 33971-83, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25336651

RESUMEN

P311, a conserved 8-kDa intracellular protein expressed in brain, smooth muscle, regenerating tissues, and malignant glioblastomas, represents the first documented stimulator of TGF-ß1-3 translation in vitro and in vivo. Here we initiated efforts to define the mechanism underlying P311 function. PONDR® (Predictor Of Naturally Disordered Regions) analysis suggested and CD confirmed that P311 is an intrinsically disordered protein, therefore requiring an interacting partner to acquire tertiary structure and function. Immunoprecipitation coupled with mass spectroscopy identified eIF3 subunit b (eIF3b) as a novel P311 binding partner. Immunohistochemical colocalization, GST pulldown, and surface plasmon resonance studies revealed that P311-eIF3b interaction is direct and has a Kd of 1.26 µm. Binding sites were mapped to the non-canonical RNA recognition motif of eIF3b and a central 11-amino acid-long region of P311, here referred to as eIF3b binding motif. Disruption of P311-eIF3b binding inhibited translation of TGF-ß1, 2, and 3, as indicated by luciferase reporter assays, polysome fractionation studies, and Western blot analysis. RNA precipitation assays after UV cross-linking and RNA-protein EMSA demonstrated that P311 binds directly to TGF-ß 5'UTRs mRNAs through a previously unidentified RNA recognition motif-like motif. Our results demonstrate that P311 is a novel RNA-binding protein that, by interacting with TGF-ßs 5'UTRs and eIF3b, stimulates the translation of TGF-ß1, 2, and 3.


Asunto(s)
Factor 3 de Iniciación Eucariótica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas/metabolismo , Biosíntesis de Proteínas , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Regiones no Traducidas 5' , Secuencia de Aminoácidos , Animales , Sitios de Unión , Escherichia coli/genética , Escherichia coli/metabolismo , Factor 3 de Iniciación Eucariótica/química , Factor 3 de Iniciación Eucariótica/genética , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Unión Proteica , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transducción de Señal , Factor de Crecimiento Transformador beta1/química , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta2/química , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta3/química , Factor de Crecimiento Transformador beta3/genética
11.
J Biol Chem ; 287(47): 39911-24, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23035120

RESUMEN

Tau is a major member of the neuronal microtubule-associated proteins. It promotes tubulin assembly and stabilizes axonal microtubules. Previous studies have demonstrated that Tau forms cross-bridges between microtubules, with some particles located on cross-bridges, suggesting that some proteins interact with Tau and might be involved in regulating Tau-related microtubule dynamics. This study reports that PACSIN1 interacts with Tau in axon. PACSIN1 blockade results in impaired axonal elongation and a higher number of primary axonal branches in mouse dorsal root ganglia neurons, which is induced by increasing the binding ability of Tau to microtubules. In PACSIN1-blocked dorsal root ganglia neurons, a greater amount of Tau is inclined to accumulate in the central domain of growth cones, and it promotes the stability of the microtubule network. Taken together, these results suggest that PACSIN1 is an important Tau binding partner in regulating microtubule dynamics and forming axonal plasticity.


Asunto(s)
Ganglios Espinales/metabolismo , Conos de Crecimiento/metabolismo , Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas tau/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Ganglios Espinales/citología , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Microtúbulos/genética , Neuropéptidos/genética , Fosfoproteínas/genética , Proteínas tau/genética
12.
J Clin Invest ; 133(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37317963

RESUMEN

RAS mutations are among the most prevalent oncogenic drivers in cancers. RAS proteins propagate signals only when associated with cellular membranes as a consequence of lipid modifications that impact their trafficking. Here, we discovered that RAB27B, a RAB family small GTPase, controlled NRAS palmitoylation and trafficking to the plasma membrane, a localization required for activation. Our proteomic studies revealed RAB27B upregulation in CBL- or JAK2-mutated myeloid malignancies, and its expression correlated with poor prognosis in acute myeloid leukemias (AMLs). RAB27B depletion inhibited the growth of CBL-deficient or NRAS-mutant cell lines. Strikingly, Rab27b deficiency in mice abrogated mutant but not WT NRAS-mediated progenitor cell growth, ERK signaling, and NRAS palmitoylation. Further, Rab27b deficiency significantly reduced myelomonocytic leukemia development in vivo. Mechanistically, RAB27B interacted with ZDHHC9, a palmitoyl acyltransferase that modifies NRAS. By regulating palmitoylation, RAB27B controlled c-RAF/MEK/ERK signaling and affected leukemia development. Importantly, RAB27B depletion in primary human AMLs inhibited oncogenic NRAS signaling and leukemic growth. We further revealed a significant correlation between RAB27B expression and sensitivity to MEK inhibitors in AMLs. Thus, our studies presented a link between RAB proteins and fundamental aspects of RAS posttranslational modification and trafficking, highlighting future therapeutic strategies for RAS-driven cancers.


Asunto(s)
Leucemia Mieloide , Lipoilación , Humanos , Animales , Ratones , Proteómica , Transducción de Señal , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas de la Membrana/genética , GTP Fosfohidrolasas
13.
J Leukoc Biol ; 112(5): 1089-1104, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35648675

RESUMEN

Langerhans cell histiocytosis (LCH) is an inflammatory disease characterized by abnormal dendritic cells (DCs) with hyperactive ERK signaling, called "LCH cells." Since DCs rely on ERK signaling to produce inflammatory molecules in response to pathogenic cues, we hypothesized that hyperactive ERK enhances DCs inflammatory responses. We specifically investigated TLR4-induced TNFα production in LCH cells by utilizing the BRAF-V600Efl/+ :CD11c-Cre mouse model of LCH, which hyperactivates ERK in DCs. We measured LPS-induced TNFα production both in vivo and in vitro using splenic CD11c+ cells and bone marrow-derived DCs with or without pharmacologic BRAFV600E inhibition. We observed a reversible increase in secreted TNFα and a partially reversible increase in TNFα protein per cell, despite a decrease in TLR4 signaling and Tnfa transcripts compared with controls. We examined ERK-driven, posttranscriptional mechanisms that contribute to TNFα production and secretion using biochemical and cellular assays. We identified a reversible increase in TACE activation, the enzyme required for TNFα secretion, and most strikingly, an increase in protein translation, including TNFα. Defining the translatome through polysome-bound RNA sequencing revealed up-regulated translation of the LPS-response program. These data suggest hyperactive ERK signaling utilizes multiple posttranscriptional mechanisms to amplify inflammatory responses in DCs, advancing our understanding of LCH and basic DC biology.


Asunto(s)
Histiocitosis de Células de Langerhans , Proteínas Proto-Oncogénicas B-raf , Ratones , Animales , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Lipopolisacáridos/farmacología , Factor de Necrosis Tumoral alfa/genética , Receptor Toll-Like 4/genética , Mutación , Histiocitosis de Células de Langerhans/metabolismo , Células Dendríticas/metabolismo
14.
Cell Rep ; 40(12): 111361, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36130501

RESUMEN

Hematopoietic stem/progenitor cells (HSPCs) are supported and regulated by niche cells in the bone marrow with an important characterization of physiological hypoxia. However, how hypoxia regulates HSPCs is still unclear. Here, we find that meteorin (Metrn) from hypoxic macrophages restrains HSPC mobilization. Hypoxia-induced factor 1α and Yin Yang 1 induce the high expression of Metrn in macrophages, and macrophage-specific Metrn knockout increases HSPC mobilization through modulating HSPC proliferation and migration. Mechanistically, Metrn interacts with its receptor 5-hydroxytryptamine receptor 2b (Htr2b) to regulate the reactive oxygen species levels in HSPCs through targeting phospholipase C signaling. The reactive oxygen species levels are reduced in HSPCs of macrophage-specific Metrn knockout mice with activated phospholipase C signaling. Targeting the Metrn/Htr2b axis could therefore be a potential strategy to improve HSPC mobilization for stem cell-based therapy.


Asunto(s)
Células de la Médula Ósea , Médula Ósea , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso , Especies Reactivas de Oxígeno/metabolismo , Receptores de Serotonina/metabolismo , Fosfolipasas de Tipo C/metabolismo
15.
Cell Stem Cell ; 28(7): 1275-1290.e9, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33711283

RESUMEN

Impaired ribosome function is the underlying etiology in a group of bone marrow failure syndromes called ribosomopathies. However, how ribosomes are regulated remains poorly understood, as are approaches to restore hematopoietic stem cell (HSC) function loss because of defective ribosome biogenesis. Here we reveal a role of the E3 ubiquitin ligase HectD1 in regulating HSC function via ribosome assembly and protein translation. Hectd1-deficient HSCs exhibit a striking defect in transplantation ability and ex vivo maintenance concomitant with reduced protein synthesis and growth rate under stress conditions. Mechanistically, HectD1 ubiquitinates and degrades ZNF622, an assembly factor for the ribosomal 60S subunit. Hectd1 loss leads to accumulation of ZNF622 and the anti-association factor eIF6 on 60S, resulting in 60S/40S joining defects. Importantly, Znf622 depletion in Hectd1-deficient HSCs restored ribosomal subunit joining, protein synthesis, and HSC reconstitution capacity. These findings highlight the importance of ubiquitin-coordinated ribosome assembly in HSC regeneration.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas , Células Madre Hematopoyéticas , Ribosomas/metabolismo
16.
Nat Cell Biol ; 22(10): 1162-1169, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32958856

RESUMEN

Stem cells need to be protected from genotoxic and proteotoxic stress to maintain a healthy pool throughout life1-3. Little is known about the proteostasis mechanism that safeguards stem cells. Here we report endoplasmic reticulum-associated degradation (ERAD) as a protein quality checkpoint that controls the haematopoietic stem cell (HSC)-niche interaction and determines the fate of HSCs. The SEL1L-HRD1 complex, the most conserved branch of ERAD4, is highly expressed in HSCs. Deletion of Sel1l led to niche displacement of HSCs and a complete loss of HSC identity, and allowed highly efficient donor-HSC engraftment without irradiation. Mechanistic studies identified MPL, the master regulator of HSC identity5, as a bona fide ERAD substrate that became aggregated in the endoplasmic reticulum following ERAD deficiency. Restoration of MPL signalling with an agonist partially rescued the number and reconstitution capacity of Sel1l-deficient HSCs. Our study defines ERAD as an essential proteostasis mechanism to safeguard a healthy stem cell pool by regulating the stem cell-niche interaction.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Receptores de Trombopoyetina/metabolismo , Nicho de Células Madre , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Trombopoyetina/genética , Ubiquitina-Proteína Ligasas/genética
17.
Nat Commun ; 9(1): 3915, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254368

RESUMEN

Fanconi anemia (FA) is a bone marrow failure (BMF) syndrome that arises from mutations in a network of FA genes essential for DNA interstrand crosslink (ICL) repair and replication stress tolerance. While allogeneic stem cell transplantation can replace defective HSCs, interventions to mitigate HSC defects in FA do not exist. Remarkably, we reveal here that Lnk (Sh2b3) deficiency restores HSC function in Fancd2-/- mice. Lnk deficiency does not impact ICL repair, but instead stabilizes stalled replication forks in a manner, in part, dependent upon alleviating blocks to cytokine-mediated JAK2 signaling. Lnk deficiency restores proliferation and survival of Fancd2-/- HSCs, while reducing replication stress and genomic instability. Furthermore, deletion of LNK in human FA-like HSCs promotes clonogenic growth. These findings highlight a new role for cytokine/JAK signaling in promoting replication fork stability, illuminate replication stress as a major underlying origin of BMF in FA, and have strong therapeutic implications.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Trasplante de Médula Ósea , Proliferación Celular/genética , Células Cultivadas , Reparación del ADN/genética , Replicación del ADN/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Proteínas de la Membrana , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados
18.
J Zhejiang Univ Sci B ; 15(12): 1032-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25471832

RESUMEN

The p53 tumor suppressor protein coordinates the cellular responses to a broad range of cellular stresses, leading to DNA repair, cell cycle arrest or apoptosis. The stability of p53 is essential for its tumor suppressor function, which is tightly controlled by ubiquitin-dependent degradation primarily through its negative regulator murine double minute 2 (Mdm2). To better understand the regulation of p53, we tested the interaction between p53 and USP11 using co-immunoprecipitation. The results show that USP11, an ubiquitin-specific protease, forms specific complexes with p53 and stabilizes p53 by deubiquitinating it. Moreover, down-regulation of USP11 dramatically attenuated p53 induction in response to DNA damage stress. These findings reveal that USP11 is a novel regulator of p53, which is required for p53 activation in response to DNA damage.


Asunto(s)
Daño del ADN , Tioléster Hidrolasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Cicloheximida/química , Reparación del ADN , Células HEK293 , Humanos , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Interferencia de ARN , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA