Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(49): e2315096120, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38011564

RESUMEN

Hidradenitis suppurativa (HS) is a complex inflammatory skin disease with undefined mechanistic underpinnings. Here, we investigated HS epithelial cells and demonstrated that HS basal progenitors modulate their lineage restriction and give rise to pathogenic keratinocyte clones, resulting in epidermal hyperproliferation and dysregulated inflammation in HS. When comparing to healthy epithelial stem/progenitor cells, in HS, we identified changes in gene signatures that revolve around the mitotic cell cycle, DNA damage response and repair, as well as cell-cell adhesion and chromatin remodeling. By reconstructing cell differentiation trajectory and CellChat modeling, we identified a keratinocyte population specific to HS. This population is marked by S100A7/8/9 and KRT6 family members, triggering IL1, IL10, and complement inflammatory cascades. These signals, along with HS-specific proinflammatory cytokines and chemokines, contribute to the recruitment of certain immune cells during the disease progression. Furthermore, we revealed a previously uncharacterized role of S100A8 in regulating the local chromatin environment of target loci in HS keratinocytes. Through the integration of genomic and epigenomic datasets, we identified genome-wide chromatin rewiring alongside the switch of transcription factors (TFs), which mediated HS transcriptional profiles. Importantly, we identified numerous clinically relevant inflammatory enhancers and their coordinated TFs in HS basal CD49fhigh cells. The disruption of the S100A enhancer using the CRISPR/Cas9-mediated approach or the pharmacological inhibition of the interferon regulatory transcription factor 3 (IRF3) efficiently reduced the production of HS-associated inflammatory regulators. Our study not only uncovers the plasticity of epidermal progenitor cells in HS but also elucidates the epigenetic mechanisms underlying HS pathogenesis.


Asunto(s)
Hidradenitis Supurativa , Humanos , Hidradenitis Supurativa/genética , Piel/metabolismo , Epigenómica , Epigénesis Genética , Células Madre/metabolismo , Cromatina/metabolismo
2.
Genes Dev ; 27(14): 1581-95, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23824326

RESUMEN

Post-translational histone modifications play important roles in regulating chromatin structure and function. Histone H2B ubiquitination and deubiquitination have been implicated in transcriptional regulation, but the function of H2B deubiquitination is not well defined, particularly in higher eukaryotes. Here we report the purification of ubiquitin-specific peptidase 49 (USP49) as a histone H2B-specific deubiquitinase and demonstrate that H2B deubiquitination by USP49 is required for efficient cotranscriptional splicing of a large set of exons. USP49 forms a complex with RuvB-like1 (RVB1) and SUG1 and specifically deubiquitinates histone H2B in vitro and in vivo. USP49 knockdown results in small changes in gene expression but affects the abundance of >9000 isoforms. Exons down-regulated in USP49 knockdown cells show both elevated levels of alternative splicing and a general decrease in splicing efficiency. Importantly, USP49 is relatively enriched at this set of exons. USP49 knockdown increased H2B ubiquitination (uH2B) levels at these exons as well as upstream 3' and downstream 5' intronic splicing elements. Change in H2B ubiquitination level, as modulated by USP49, regulates U1A and U2B association with chromatin and binding to nascent pre-mRNA. Although H3 levels are relatively stable after USP49 depletion, H2B levels at these exons are dramatically increased, suggesting that uH2B may enhance nucleosome stability. Therefore, this study identifies USP49 as a histone H2B-specific deubiquitinase and uncovers a critical role for H2B deubiquitination in cotranscriptional pre-mRNA processing events.


Asunto(s)
Histonas/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN Helicasas/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas con Dominio LIM/metabolismo , Complejo de la Endopetidasa Proteasomal , Factores de Transcripción/metabolismo , Ubiquitina Tiolesterasa/aislamiento & purificación , Ubiquitinación
3.
Circulation ; 139(23): 2654-2663, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905171

RESUMEN

BACKGROUND: Nitrosation of a conserved cysteine residue at position 93 in the hemoglobin ß chain (ß93C) to form S-nitroso (SNO) hemoglobin (Hb) is claimed to be essential for export of nitric oxide (NO) bioactivity by the red blood cell (RBC) to mediate hypoxic vasodilation and cardioprotection. METHODS: To test this hypothesis, we used RBCs from mice in which the ß93 cysteine had been replaced with alanine (ß93A) in a number of ex vivo and in vivo models suitable for studying export of NO bioactivity. RESULTS: In an ex vivo model of cardiac ischemia/reperfusion injury, perfusion of a mouse heart with control RBCs (ß93C) pretreated with an arginase inhibitor to facilitate export of RBC NO bioactivity improved cardiac recovery after ischemia/reperfusion injury, and the response was similar with ß93A RBCs. Next, when human platelets were coincubated with RBCs and then deoxygenated in the presence of nitrite, export of NO bioactivity was detected as inhibition of ADP-induced platelet activation. This effect was the same in ß93C and ß93A RBCs. Moreover, vascular reactivity was tested in rodent aortas in the presence of RBCs pretreated with S-nitrosocysteine or with hemolysates or purified Hb treated with authentic NO to form nitrosyl(FeII)-Hb, the proposed precursor of SNO-Hb. SNO-RBCs or NO-treated Hb induced vasorelaxation, with no differences between ß93C and ß93A RBCs. Finally, hypoxic microvascular vasodilation was studied in vivo with a murine dorsal skin-fold window model. Exposure to acute systemic hypoxia caused vasodilatation, and the response was similar in ß93C and ß93A mice. CONCLUSIONS: RBCs clearly have the fascinating ability to export NO bioactivity, but this occurs independently of SNO formation at the ß93 cysteine of Hb.


Asunto(s)
Plaquetas/metabolismo , Eritrocitos/metabolismo , Hemoglobinas/metabolismo , Daño por Reperfusión Miocárdica/sangre , Óxido Nítrico/sangre , Piel/irrigación sanguínea , Globinas beta/metabolismo , Alanina , Sustitución de Aminoácidos , Animales , Transporte Biológico , Cisteína , Modelos Animales de Enfermedad , Hemoglobinas/genética , Humanos , Hipoxia/sangre , Hipoxia/fisiopatología , Preparación de Corazón Aislado , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Daño por Reperfusión Miocárdica/fisiopatología , Activación Plaquetaria , Ratas Sprague-Dawley , Vasodilatación , Función Ventricular Izquierda , Presión Ventricular , Globinas beta/genética
4.
Proc Natl Acad Sci U S A ; 113(1): E51-60, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26699484

RESUMEN

Epigenetic mechanisms play important regulatory roles in hematopoiesis and hematopoietic stem cell (HSC) function. Subunits of polycomb repressive complex 1 (PRC1), the major histone H2A ubiquitin ligase, are critical for both normal and pathological hematopoiesis; however, it is unclear which of the several counteracting H2A deubiquitinases functions along with PRC1 to control H2A ubiquitination (ubH2A) level and regulates hematopoiesis in vivo. Here we investigated the function of Usp16 in mouse hematopoiesis. Conditional deletion of Usp16 in bone marrow resulted in a significant increase of global ubH2A level and lethality. Usp16 deletion did not change HSC number but was associated with a dramatic reduction of mature and progenitor cell populations, revealing a role in governing HSC lineage commitment. ChIP- and RNA-sequencing studies in HSC and progenitor cells revealed that Usp16 bound to many important hematopoietic regulators and that Usp16 deletion altered the expression of genes in transcription/chromosome organization, immune response, hematopoietic/lymphoid organ development, and myeloid/leukocyte differentiation. The altered gene expression was partly rescued by knockdown of PRC1 subunits, suggesting that Usp16 and PRC1 counterbalance each other to regulate cellular ubH2A level and gene expression in the hematopoietic system. We further discovered that knocking down Cdkn1a (p21cip1), a Usp16 target and regulated gene, rescued the altered cell cycle profile and differentiation defect of Usp16-deleted HSCs. Collectively, these studies identified Usp16 as one of the histone H2A deubiquitinases, which coordinates with the H2A ubiquitin ligase PRC1 to regulate hematopoiesis, and revealed cell cycle regulation by Usp16 as key for HSC differentiation.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Ubiquitina Tiolesterasa/fisiología , Proteasas Ubiquitina-Específicas/fisiología , Animales , Recuento de Células , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Endopeptidasas/genética , Endopeptidasas/fisiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Letales , Hematopoyesis/genética , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/fisiología , Transactivadores , Ubiquitina Tiolesterasa/genética , Proteasas Ubiquitina-Específicas/genética
5.
Biol Blood Marrow Transplant ; 24(8): 1554-1562, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29684562

RESUMEN

Allogeneic hematopoietic stem cell transplantation (HSCT) can be curative for patients with sickle cell disease (SCD). However, morbidity associated with myeloablative conditioning and graft-versus-host disease has limited its utility. To this end, autologous HSCT for SCD using lentiviral gene-modified bone marrow (BM) or peripheral blood stem cells has been undertaken, although toxicities of fully ablative conditioning with busulfan and incomplete engraftment have been encountered. Treosulfan, a busulfan analog with a low extramedullary toxicity profile, has been used successfully as part of a myeloablative conditioning regimen in the allogeneic setting in SCD. To further minimize toxicity of conditioning, noncytotoxic monoclonal antibodies that clear stem cells from the marrow niche, such as anti-c-Kit (ACK2), have been considered. Using a murine model of SCD, we sought to determine whether nonmyeloablative conditioning followed by transplantation with syngeneic BM cells could ameliorate the disease phenotype. Treosulfan and ACK2, in a dose-dependent manner, decreased BM cellularity and induced cytopenia in SCD mice. Conditioning with treosulfan alone at nonmyeloablative dosing (3.6 g/kg), followed by transplantation with syngeneic BM donor cells, permitted long-term mixed-donor chimerism. Level of chimerism correlated with improvement in hematologic parameters, normalization of urine osmolality, and improvement in liver and spleen pathology. Addition of ACK2 to treosulfan conditioning did not enhance engraftment. Our data suggests that pretransplant conditioning with treosulfan alone may allow sufficient erythroid engraftment to reverse manifestations of SCD, with clinical application as a preparative regimen in SCD patients undergoing gene-modified autologous HSCT.


Asunto(s)
Anemia de Células Falciformes/terapia , Trasplante de Médula Ósea/métodos , Busulfano/análogos & derivados , Acondicionamiento Pretrasplante/métodos , Animales , Anticuerpos/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Busulfano/uso terapéutico , Modelos Animales de Enfermedad , Supervivencia de Injerto , Ratones , Proteínas Proto-Oncogénicas c-kit/inmunología , Resultado del Tratamiento
6.
EMBO Rep ; 17(6): 887-900, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27118388

RESUMEN

Antisense RNAs regulate the transcription and translation of the corresponding sense genes. Here, we report that an antisense RNA, AS-RBM15, is transcribed in the opposite direction within exon 1 of RBM15 RBM15 is a regulator of megakaryocyte (MK) differentiation and is also involved in a chromosome translocation t(1;22) in acute megakaryocytic leukemia. MK terminal differentiation is enhanced by up-regulation of AS-RBM15 expression and attenuated by AS-RBM15 knockdown. At the molecular level, AS-RBM15 enhances RBM15 protein translation in a CAP-dependent manner. The region of the antisense AS-RBM15 RNA, which overlaps with the 5'UTR of RBM15, is sufficient for the up-regulation of RBM15 protein translation. In addition, we find that transcription of both RBM15 and AS-RBM15 is activated by the transcription factor RUNX1 and repressed by RUNX1-ETO, a leukemic fusion protein. Therefore, AS-RBM15 is a regulator of megakaryocyte differentiation and may play a regulatory role in leukemogenesis.


Asunto(s)
Diferenciación Celular/genética , Megacariocitos/citología , Megacariocitos/metabolismo , ARN sin Sentido , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Biosíntesis de Proteínas , Transporte de Proteínas , Eliminación de Secuencia , Transcripción Genética
7.
J Am Soc Nephrol ; 28(8): 2443-2458, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28348063

RESUMEN

Sickle cell disease (SCD)-associated nephropathy is a major source of morbidity and mortality in patients because of the lack of efficacious treatments targeting renal manifestations of the disease. Here, we describe a long-term treatment strategy with the selective endothelin-A receptor (ETA) antagonist, ambrisentan, designed to interfere with the development of nephropathy in a humanized mouse model of SCD. Ambrisentan preserved GFR at the level of nondisease controls and prevented the development of proteinuria, albuminuria, and nephrinuria. Microscopy studies demonstrated prevention of podocyte loss and structural alterations, the absence of vascular congestion, and attenuation of glomerulosclerosis in treated mice. Studies in isolated glomeruli showed that treatment reduced inflammation and oxidative stress. At the level of renal tubules, ambrisentan treatment prevented the increased excretion of urinary tubular injury biomarkers. Additionally, the treatment strategy prevented tubular brush border loss, diminished tubular iron deposition, blocked the development of interstitial fibrosis, and prevented immune cell infiltration. Furthermore, the prevention of albuminuria in treated mice was associated with preservation of cortical megalin expression. In a separate series of identical experiments, combined ETA and ETB receptor antagonism provided only some of the protection observed with ambrisentan, highlighting the importance of exclusively targeting the ETA receptor in SCD. Our results demonstrate that ambrisentan treatment provides robust protection from diverse renal pathologies in SCD mice, and suggest that long-term ETA receptor antagonism may provide a strategy for the prevention of renal complications of SCD.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Antagonistas de los Receptores de la Endotelina A/uso terapéutico , Enfermedades Renales/etiología , Enfermedades Renales/prevención & control , Fenilpropionatos/uso terapéutico , Piridazinas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Factores de Tiempo
8.
Basic Res Cardiol ; 112(2): 19, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28238121

RESUMEN

In the failing heart, iNOS is expressed by both macrophages and cardiomyocytes. We hypothesized that inflammatory cell-localized iNOS exacerbates left ventricular (LV) remodeling. Wild-type (WT) C57BL/6 mice underwent total body irradiation and reconstitution with bone marrow from iNOS-/- mice (iNOS-/-c) or WT mice (WTc). Chimeric mice underwent coronary ligation to induce large infarction and ischemic heart failure (HF), or sham surgery. After 28 days, as compared with WTc sham mice, WTc HF mice exhibited significant (p < 0.05) mortality, LV dysfunction, hypertrophy, fibrosis, oxidative/nitrative stress, inflammatory activation, and iNOS upregulation. These mice also exhibited a ~twofold increase in circulating Ly6Chi pro-inflammatory monocytes, and ~sevenfold higher cardiac M1 macrophages, which were primarily CCR2- cells. In contrast, as compared with WTc HF mice, iNOS-/-c HF mice exhibited significantly improved survival, LV function, hypertrophy, fibrosis, oxidative/nitrative stress, and inflammatory activation, without differences in overall cardiac iNOS expression. Moreover, iNOS-/-c HF mice exhibited lower circulating Ly6Chi monocytes, and augmented cardiac M2 macrophages, but with greater infiltrating monocyte-derived CCR2+ macrophages vs. WTc HF mice. Lastly, upon cell-to-cell contact with naïve cardiomyocytes, peritoneal macrophages from WT HF mice depressed contraction, and augmented cardiomyocyte oxygen free radicals and peroxynitrite. These effects were not observed upon contact with macrophages from iNOS-/- HF mice. We conclude that leukocyte iNOS is obligatory for local and systemic inflammatory activation and cardiac remodeling in ischemic HF. Activated macrophages in HF may directly induce cardiomyocyte contractile dysfunction and oxidant stress upon cell-to-cell contact; this juxtacrine response requires macrophage-localized iNOS.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Remodelación Ventricular/fisiología , Animales , Western Blotting , Ecocardiografía , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Inmunohistoquímica , Isquemia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Am J Hematol ; 92(10): 981-988, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28646491

RESUMEN

Sickle erythrocytes' (SSRBCs) unique physical adaptation to hypoxic conditions renders them able to home to hypoxic tumor niches in vivo, shut down tumor blood flow and induce tumoricidal responses. SSRBCs are also useful vehicles for transport of encapsulated drugs and oncolytic virus into hypoxic tumors with enhanced anti-tumor effects. In search of additional modes for arming sickle cells with cytotoxics, we turned to a lentiviral ß-globin vector with optimized Locus Control Region/ß-globin coding region/promoter/enhancers. We partially replaced the ß-globin coding region of this vector with genes encoding T cell cytolytics, perforin and granzyme or immune modulating superantigens SEG and SEI. These modified vectors efficiently transduced Sca+ ckit- Lin- hematopoietic stem cells (HSCs) from humanized sickle cell knockin mice. Irradiated mice reconstituted with these HSCs displayed robust expression of transgenic RNAs and proteins in host sickle cells that was sustained for more than 10 months. SSRBCs from reconstituted mice harboring SEG/SEI transgenes induced robust proliferation and a prototypical superantigen-induced cytokine reaction when exposed to human CD4+/CD8+ cells. The ß-globin lentiviral vector therefore produces a high level of functional, erythroid-specific immune modulators and cytotoxics that circulate without toxicity. Coupled with their unique ability to target and occlude hypoxic tumor vessels these armed SSRBCs constitute a potentially useful tool for treatment of solid tumors.


Asunto(s)
Anemia de Células Falciformes , Citotoxicidad Inmunológica , Eritrocitos Anormales/inmunología , Neoplasias Experimentales/inmunología , Neovascularización Patológica/inmunología , Globinas beta/genética , Anemia de Células Falciformes/sangre , Animales , Citotoxicidad Inmunológica/genética , Sistemas de Liberación de Medicamentos , Eritrocitos Anormales/metabolismo , Eritrocitos Anormales/trasplante , Técnicas de Sustitución del Gen , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Hipoxia , Lentivirus/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/terapia , Neovascularización Patológica/patología , Neovascularización Patológica/terapia
12.
Blood ; 133(21): 2245-2246, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31122935
13.
Blood ; 124(13): 2025-33, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25139354

RESUMEN

Epigenetic mechanisms, including histone modifications, have emerged as important factors influencing cell fate determination. The functional role of H3K4 methylation, however, remains largely unclear in the maintenance and differentiation of hematopoietic stem cells (HSCs)/hematopoietic progenitor cells (HPCs). Here we show that DPY30, a shared core subunit of the SET1/MLL family methyltransferase complexes and a facilitator of their H3K4 methylation activity, is important for ex vivo proliferation and differentiation of human CD34(+) HPCs. DPY30 promotes HPC proliferation by directly regulating the expression of genes critical for cell proliferation. Interestingly, while DPY30 knockdown in HPCs impaired their differentiation into the myelomonocytic lineage, it potently promoted hemoglobin production and affected the kinetics of their differentiation into the erythroid lineage. In an in vivo model, we show that morpholino-mediated dpy30 knockdown resulted in severe defects in the development of the zebrafish hematopoietic system, which could be partially rescued by coinjection of dpy30 messenger RNA. Taken together, our results establish a critical role of DPY30 in the proliferation and appropriate differentiation of hematopoietic progenitor cells and in animal hematopoiesis. Finally, we also demonstrate a crucial role of DPY30 in the growth of several MLL1-fusion-mediated leukemia cell lines.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Complejos Multiproteicos/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/metabolismo , Subunidades de Proteína/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Hematopoyesis , N-Metiltransferasa de Histona-Lisina/química , Humanos , Leucemia/genética , Leucemia/metabolismo , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Proteína de la Leucemia Mieloide-Linfoide/química , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/genética , Alineación de Secuencia , Factores de Transcripción , Pez Cebra
14.
Proc Natl Acad Sci U S A ; 109(10): 3772-7, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22362888

RESUMEN

To gain insight into mechanisms controlling SRY (sex determining region Y)-box 2 (Sox2) protein activity in mouse embryonic stem cells (ESCs), the endogenous Sox2 gene was tagged with FLAG/Hemagglutinin (HA) sequences by homologous recombination. Sox2 protein complexes were purified from Sox2/FLAG/HA knockin ESCs, and interacting proteins were defined by mass spectrometry. One protein in the complex was poly ADP-ribose polymerase I (Parp1). The results presented below demonstrate that Parp1 regulates Sox2 protein activity. In response to fibroblast growth factor (FGF)/extracellular signal-regulated kinase (ERK) signaling, Parp1 auto-poly ADP-ribosylation enhances Sox2-Parp1 interactions, and this complex inhibits Sox2 binding to octamer-binding transcription factor 4 (Oct4)/Sox2 enhancers. Based on these results, we propose a unique mechanism in which FGF signaling fine-tunes Sox2 activity through posttranslational modification of a critical interacting protein, Parp1, and balances the maintenance of ESC pluripotency and differentiation. In addition, we demonstrate that regulation of Sox2 activity by Parp1 is critical for efficient generation of induced pluripotent stem cells.


Asunto(s)
Células Madre Embrionarias/citología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Adenosina Difosfato/genética , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Marcación de Gen , Espectrometría de Masas/métodos , Ratones , Modelos Genéticos , Células Madre Pluripotentes/citología , Poli(ADP-Ribosa) Polimerasa-1 , Recombinación Genética , Transducción de Señal
15.
Circulation ; 127(12): 1317-29, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23446829

RESUMEN

BACKGROUND: Hemolytic diseases are characterized by enhanced intravascular hemolysis resulting in heme-catalyzed reactive oxygen species generation, which leads to endothelial dysfunction and oxidative damage. Hemopexin (Hx) is a plasma heme scavenger able to prevent endothelial damage and tissue congestion in a model of heme overload. Here, we tested whether Hx could be used as a therapeutic tool to counteract heme toxic effects on the cardiovascular system in hemolytic diseases. METHODS AND RESULTS: By using a model of heme overload in Hx-null mice, we demonstrated that heme excess in plasma, if not bound to Hx, promoted the production of reactive oxygen species and the induction of adhesion molecules and caused the reduction of nitric oxide availability. Then, we used ß-thalassemia and sickle cell disease mice as models of hemolytic diseases to evaluate the efficacy of an Hx-based therapy in the treatment of vascular dysfunction related to heme overload. Our data demonstrated that Hx prevented heme-iron loading in the cardiovascular system, thus limiting the production of reactive oxygen species, the induction of adhesion molecules, and the oxidative inactivation of nitric oxide synthase/nitric oxide, and promoted heme recovery and detoxification by the liver mainly through the induction of heme oxygenase activity. Moreover, we showed that in sickle cell disease mice, endothelial activation and oxidation were associated with increased blood pressure and altered cardiac function, and the administration of exogenous Hx was found to almost completely normalize these parameters. CONCLUSIONS: Hemopexin treatment is a promising novel therapy to protect against heme-induced cardiovascular dysfunction in hemolytic disorders.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Sistema Cardiovascular/fisiopatología , Endotelio Vascular/fisiopatología , Hemo/efectos adversos , Hemopexina/uso terapéutico , Talasemia beta/tratamiento farmacológico , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/fisiopatología , Animales , Sistema Cardiovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Hemo/metabolismo , Hemopexina/genética , Hemopexina/farmacología , Ratones , Ratones Noqueados , Ratones SCID , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento , Talasemia beta/metabolismo , Talasemia beta/fisiopatología
16.
Blood ; 119(22): 5276-84, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22498744

RESUMEN

Heme-regulated eIF2α kinase (Hri) is necessary for balanced synthesis of heme and globin. In addition, Hri deficiency exacerbates the phenotypic severity of ß-thalassemia intermedia in mice. Activation of Hri during heme deficiency and in ß-thalassemia increases eIF2α phosphorylation and inhibits globin translation. Under endoplasmic reticulum stress and nutrient starvation, eIF2α phosphorylation also induces the Atf4 signaling pathway to mitigate stress. Although the function of Hri in regulating globin translation is well established, its role in Atf4 signaling in erythroid precursors is not known. Here, we report the role of the Hri-activated Atf4 signaling pathway in reducing oxidative stress and in promoting erythroid differentiation during erythropoiesis. On acute oxidative stress, Hri(-/-) erythroblasts suffered from increased levels of reactive oxygen species (ROS) and apoptosis. During chronic iron deficiency in vivo, Hri is necessary both to reduce oxidative stress and to promote erythroid differentiation. Hri(-/-) mice developed ineffective erythropoiesis during iron deficiency with inhibition of differentiation at the basophilic erythroblast stage. This inhibition is recapitulated during ex vivo differentiation of Hri(-/-) fetal liver erythroid progenitors. Importantly, the Hri-eIF2αP-Atf4 pathway was activated and required for erythroid differentiation. We further demonstrate the potential of modulating Hri-eIF2αP-Atf4 signaling with chemical compounds as pharmaceutical therapies for ß-thalassemia.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Eritroblastos/metabolismo , Eritropoyesis , Estrés Oxidativo , Transducción de Señal , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Eritroblastos/patología , Feto/embriología , Feto/metabolismo , Feto/patología , Globinas/biosíntesis , Globinas/genética , Hierro/metabolismo , Deficiencias de Hierro , Hígado/embriología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Biosíntesis de Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/patología , Talasemia beta/terapia , eIF-2 Quinasa/genética
17.
J Immunol ; 186(2): 1001-10, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21148039

RESUMEN

Cells react to viral infection by exhibiting IFN-based innate immune responses and integrated stress responses, but little is known about the interrelationships between the two. In this study, we report a linkage between these two host-protective cellular mechanisms. We found that IFN regulatory factor (IRF)7, the master regulator of type I IFN gene expression, interacts with activating transcription factor (ATF)4, a key component of the integrated stress responses whose translation is induced by viral infection and various stresses. We have demonstrated that IRF7 upregulates ATF4 activity and expression, whereas ATF4 in return inhibits IRF7 activation, suggesting a cross-regulation between the IFN response and the cellular integrated stress response that controls host innate immune defense against viral infection.


Asunto(s)
Factor de Transcripción Activador 4/fisiología , Comunicación Celular/inmunología , Regulación hacia Abajo/inmunología , Factor 7 Regulador del Interferón/antagonistas & inhibidores , Interferones/biosíntesis , Estrés Fisiológico/inmunología , Factor de Transcripción Activador 4/biosíntesis , Factor de Transcripción Activador 4/deficiencia , Factor de Transcripción Activador 4/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Factor 7 Regulador del Interferón/metabolismo , Interferones/metabolismo , Interferones/fisiología , Ratones , Datos de Secuencia Molecular , Regulación hacia Arriba/inmunología , Células Vero , Virus de la Estomatitis Vesicular Indiana/inmunología
18.
PLoS One ; 18(11): e0295009, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38019847

RESUMEN

A major unmet need in the cystic fibrosis (CF) therapeutic landscape is the lack of effective treatments for nonsense CFTR mutations, which affect approximately 10% of CF patients. Correction of nonsense CFTR mutations via genomic editing represents a promising therapeutic approach. In this study, we tested whether prime editing, a novel CRISPR-based genomic editing method, can be a potential therapeutic modality to correct nonsense CFTR mutations. We generated iPSCs from a CF patient homozygous for the CFTR W1282X mutation. We demonstrated that prime editing corrected one mutant allele in iPSCs, which effectively restored CFTR function in iPSC-derived airway epithelial cells and organoids. We further demonstrated that prime editing may directly repair mutations in iPSC-derived airway epithelial cells when the prime editing machinery is efficiently delivered by helper-dependent adenovirus (HDAd). Together, our data demonstrated that prime editing may potentially be applied to correct CFTR mutations such as W1282X.


Asunto(s)
Fibrosis Quística , Células Madre Pluripotentes Inducidas , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Mutación , Fibrosis Quística/terapia , Fibrosis Quística/tratamiento farmacológico , Codón sin Sentido , Células Epiteliales
19.
Stem Cells ; 29(2): 229-40, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21732481

RESUMEN

Polycomb repressive complex two (PRC2) has been implicated in embryonic stem (ES) cell pluripotency; however, the mechanistic roles of this complex are unclear. It was assumed that ES cells contain PRC2 with the same subunit composition as that identified in HeLa cells and Drosophila embryos. Here, we report that PRC2 in mouse ES cells contains at least three additional subunits: JARID2, MTF2, and a novel protein denoted esPRC2p48. JARID2, MTF2, and esPRC2p48 are highly expressed in mouse ES cells compared to differentiated cells. Importantly, knockdowns of JARID2, MTF2, or esPRC2p48 alter the level of PRC2-mediated H3K27 methylation and result in the expression of differentiation-associated genes in ES cells. Interestingly, expression of JARID2, MTF2, and esPRC2p48 together, but not individually, enhances Oct4/Sox2/Klf4-mediated reprogramming of mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells, whereas knockdown or knockout of JARID2, MTF2, or esPRC2p48 significantly inhibits reprogramming. JARID2, MTF2, and esPRC2p48 modulate H3K27 methylation and facilitate repression of lineage-associated gene expression when transduced into MEFs, and synergistically stimulate the histone methyltransferase activity of PRC2 in vitro. Therefore, these studies identify JARID2, MTF2, and esPRC2p48 as important regulatory subunits of PRC2 in ES cells and reveal critical functions of these subunits in modulating PRC2's activity and gene expression both in ES cells and during somatic cell reprogramming.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Unión Proteica , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Factores de Transcripción SOXB1/metabolismo
20.
PLoS Biol ; 6(10): e245, 2008 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-18842067

RESUMEN

DNA replication in mammals is regulated via the coordinate firing of clusters of replicons that duplicate megabase-sized chromosome segments at specific times during S-phase. Cytogenetic studies show that these "replicon clusters" coalesce as subchromosomal units that persist through multiple cell generations, but the molecular boundaries of such units have remained elusive. Moreover, the extent to which changes in replication timing occur during differentiation and their relationship to transcription changes has not been rigorously investigated. We have constructed high-resolution replication-timing profiles in mouse embryonic stem cells (mESCs) before and after differentiation to neural precursor cells. We demonstrate that chromosomes can be segmented into multimegabase domains of coordinate replication, which we call "replication domains," separated by transition regions whose replication kinetics are consistent with large originless segments. The molecular boundaries of replication domains are remarkably well conserved between distantly related ESC lines and induced pluripotent stem cells. Unexpectedly, ESC differentiation was accompanied by the consolidation of smaller differentially replicating domains into larger coordinately replicated units whose replication time was more aligned to isochore GC content and the density of LINE-1 transposable elements, but not gene density. Replication-timing changes were coordinated with transcription changes for weak promoters more than strong promoters, and were accompanied by rearrangements in subnuclear position. We conclude that replication profiles are cell-type specific, and changes in these profiles reveal chromosome segments that undergo large changes in organization during differentiation. Moreover, smaller replication domains and a higher density of timing transition regions that interrupt isochore replication timing define a novel characteristic of the pluripotent state.


Asunto(s)
Diferenciación Celular/fisiología , Replicación del ADN/fisiología , Células Madre Embrionarias/citología , Transcripción Genética/fisiología , Animales , Ciclo Celular/fisiología , Línea Celular , Células Madre Embrionarias/fisiología , Perfilación de la Expresión Génica , Hibridación Fluorescente in Situ , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA