Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 17(7): 784-793, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34155404

RESUMEN

Polycomb repressive complex 1 (PRC1) is an essential chromatin-modifying complex that monoubiquitinates histone H2A and is involved in maintaining the repressed chromatin state. Emerging evidence suggests PRC1 activity in various cancers, rationalizing the need for small-molecule inhibitors with well-defined mechanisms of action. Here, we describe the development of compounds that directly bind to RING1B-BMI1, the heterodimeric complex constituting the E3 ligase activity of PRC1. These compounds block the association of RING1B-BMI1 with chromatin and inhibit H2A ubiquitination. Structural studies demonstrate that these inhibitors bind to RING1B by inducing the formation of a hydrophobic pocket in the RING domain. Our PRC1 inhibitor, RB-3, decreases the global level of H2A ubiquitination and induces differentiation in leukemia cell lines and primary acute myeloid leukemia (AML) samples. In summary, we demonstrate that targeting the PRC1 RING domain with small molecules is feasible, and RB-3 represents a valuable chemical tool to study PRC1 biology.


Asunto(s)
Complejo Represivo Polycomb 1/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células K562 , Modelos Moleculares , Estructura Molecular , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Ubiquitinación/efectos de los fármacos
2.
Nat Chem Biol ; 16(12): 1403-1410, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32868895

RESUMEN

The nuclear receptor-binding SET domain (NSD) family of histone methyltransferases is associated with various malignancies, including aggressive acute leukemia with NUP98-NSD1 translocation. While NSD proteins represent attractive drug targets, their catalytic SET domains exist in autoinhibited conformation, presenting notable challenges for inhibitor development. Here, we employed a fragment-based screening strategy followed by chemical optimization, which resulted in the development of the first-in-class irreversible small-molecule inhibitors of the nuclear receptor-binding SET domain protein 1 (NSD1) SET domain. The crystal structure of NSD1 in complex with covalently bound ligand reveals a conformational change in the autoinhibitory loop of the SET domain and formation of a channel-like pocket suitable for targeting with small molecules. Our covalent lead-compound BT5-demonstrates on-target activity in NUP98-NSD1 leukemia cells, including inhibition of histone H3 lysine 36 dimethylation and downregulation of target genes, and impaired colony formation in an NUP98-NSD1 patient sample. This study will facilitate the development of the next generation of potent and selective inhibitors of the NSD histone methyltransferases.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Leucémica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucocitos/efectos de los fármacos , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Antineoplásicos/síntesis química , Sitios de Unión , Inhibidores Enzimáticos/síntesis química , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cinética , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Leucemia/genética , Leucemia/patología , Leucocitos/enzimología , Leucocitos/patología , Modelos Moleculares , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Especificidad por Sustrato , Células Tumorales Cultivadas
4.
Proc Natl Acad Sci U S A ; 111(27): 9899-904, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958854

RESUMEN

Homeobox A9 (HOXA9) is a homeodomain-containing transcription factor that plays a key role in hematopoietic stem cell expansion and is commonly deregulated in human acute leukemias. A variety of upstream genetic alterations in acute myeloid leukemia (AML) lead to overexpression of HOXA9, almost always in association with overexpression of its cofactor meis homeobox 1 (MEIS1) . A wide range of data suggests that HOXA9 and MEIS1 play a synergistic causative role in AML, although the molecular mechanisms leading to transformation by HOXA9 and MEIS1 remain elusive. In this study, we identify CCAAT/enhancer binding protein alpha (C/EBPα) as a critical collaborator required for Hoxa9/Meis1-mediated leukemogenesis. We show that C/EBPα is required for the proliferation of Hoxa9/Meis1-transformed cells in culture and that loss of C/EBPα greatly improves survival in both primary and secondary murine models of Hoxa9/Meis1-induced leukemia. Over 50% of Hoxa9 genome-wide binding sites are cobound by C/EBPα, which coregulates a number of downstream target genes involved in the regulation of cell proliferation and differentiation. Finally, we show that Hoxa9 represses the locus of the cyclin-dependent kinase inhibitors Cdkn2a/b in concert with C/EBPα to overcome a block in G1 cell cycle progression. Together, our results suggest a previously unidentified role for C/EBPα in maintaining the proliferation required for Hoxa9/Meis1-mediated leukemogenesis.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteínas de Homeodominio/fisiología , Leucemia Experimental/fisiopatología , Proteínas de Neoplasias/fisiología , Animales , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Regiones Promotoras Genéticas , Unión Proteica
5.
Blood ; 124(25): 3730-7, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25305204

RESUMEN

Lens epithelium-derived growth factor (LEDGF) is a chromatin-associated protein implicated in leukemia and HIV type 1 infection. LEDGF associates with mixed-lineage leukemia (MLL) fusion proteins and menin and is required for leukemic transformation. To better understand the molecular mechanism underlying the LEDGF integrase-binding domain (IBD) interaction with MLL fusion proteins in leukemia, we determined the solution structure of the MLL-IBD complex. We found a novel MLL motif, integrase domain binding motif 2 (IBM2), which binds to a well-defined site on IBD. Point mutations within IBM2 abolished leukemogenic transformation by MLL-AF9, validating that this newly identified motif is essential for the oncogenic activity of MLL fusion proteins. Interestingly, the IBM2 binding site on IBD overlaps with the binding site for the HIV integrase (IN), and IN was capable of efficiently sequestering IBD from the menin-MLL complex. A short IBM2 peptide binds to IBD directly and inhibits both the IBD-MLL/menin and IBD-IN interactions. Our findings show that the same site on IBD is involved in binding to MLL and HIV-IN, revealing an attractive approach to simultaneously target LEDGF in leukemia and HIV.


Asunto(s)
Infecciones por VIH/metabolismo , Integrasa de VIH/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia Bifenotípica Aguda/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , Sitios de Unión/genética , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , N-Metiltransferasa de Histona-Lisina , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Leucemia Bifenotípica Aguda/tratamiento farmacológico , Espectroscopía de Resonancia Magnética , Ratones Endogámicos C57BL , Modelos Moleculares , Terapia Molecular Dirigida , Mutación , Proteína de la Leucemia Mieloide-Linfoide/química , Proteína de la Leucemia Mieloide-Linfoide/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
6.
J Biol Chem ; 289(19): 13000-9, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24675075

RESUMEN

We have recently shown that the non-coding RNA, steroid receptor RNA activator (SRA), functions as a transcriptional coactivator of PPARγ and promotes adipocyte differentiation in vitro. To assess SRA function in vivo, we have generated a whole mouse Sra1 gene knock-out (SRA(-/-)). Here, we show that the Sra1 gene is an important regulator of adipose tissue mass and function. SRA is expressed at a higher level in adipose tissue than other organs in wild type mice. SRA(-/-) mice are resistant to high fat diet-induced obesity, with decreased fat mass and increased lean content. This lean phenotype of SRA(-/-) mice is associated with decreased expression of a subset of adipocyte marker genes and reduced plasma TNFα levels. The SRA(-/-) mice are more insulin sensitive, as evidenced by reduced fasting insulin, and lower blood glucoses in response to IP glucose and insulin. In addition, the livers of SRA(-/-) mice have fewer lipid droplets after high fat diet feeding, and the expression of lipogenesis-associated genes is decreased. To our knowledge, these data are the first to indicate a functional role for SRA in adipose tissue biology and glucose homeostasis in vivo.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Glucemia/metabolismo , Grasas de la Dieta/efectos adversos , Obesidad/metabolismo , ARN Largo no Codificante/metabolismo , Adipocitos/patología , Tejido Adiposo/patología , Animales , Glucemia/genética , Grasas de la Dieta/farmacología , Homeostasis/genética , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , ARN Largo no Codificante/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
7.
EMBO J ; 30(7): 1357-75, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21407177

RESUMEN

The endoplasmic reticulum (ER) is the cellular organelle responsible for protein folding and assembly, lipid and sterol biosynthesis, and calcium storage. The unfolded protein response (UPR) is an adaptive intracellular stress response to accumulation of unfolded or misfolded proteins in the ER. In this study, we show that the most conserved UPR sensor inositol-requiring enzyme 1 α (IRE1α), an ER transmembrane protein kinase/endoribonuclease, is required to maintain hepatic lipid homeostasis under ER stress conditions through repressing hepatic lipid accumulation and maintaining lipoprotein secretion. To elucidate physiological roles of IRE1α-mediated signalling in the liver, we generated hepatocyte-specific Ire1α-null mice by utilizing an albumin promoter-controlled Cre recombinase-mediated deletion. Deletion of Ire1α caused defective induction of genes encoding functions in ER-to-Golgi protein transport, oxidative protein folding, and ER-associated degradation (ERAD) of misfolded proteins, and led to selective induction of pro-apoptotic UPR trans-activators. We show that IRE1α is required to maintain the secretion efficiency of selective proteins. In the absence of ER stress, mice with hepatocyte-specific Ire1α deletion displayed modest hepatosteatosis that became profound after induction of ER stress. Further investigation revealed that IRE1α represses expression of key metabolic transcriptional regulators, including CCAAT/enhancer-binding protein (C/EBP) ß, C/EBPδ, peroxisome proliferator-activated receptor γ (PPARγ), and enzymes involved in triglyceride biosynthesis. IRE1α was also found to be required for efficient secretion of apolipoproteins upon disruption of ER homeostasis. Consistent with a role for IRE1α in preventing intracellular lipid accumulation, mice with hepatocyte-specific deletion of Ire1α developed severe hepatic steatosis after treatment with an ER stress-inducing anti-cancer drug Bortezomib, upon expression of a misfolding-prone human blood clotting factor VIII, or after partial hepatectomy. The identification of IRE1α as a key regulator to prevent hepatic steatosis provides novel insights into ER stress mechanisms in fatty liver diseases associated with toxic liver injuries.


Asunto(s)
Retículo Endoplásmico/metabolismo , Endorribonucleasas/metabolismo , Hígado Graso/prevención & control , Proteínas Serina-Treonina Quinasas/metabolismo , Respuesta de Proteína Desplegada , Animales , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados
8.
Blood ; 120(9): 1923-32, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22613792

RESUMEN

Factor VIII and factor V share structural homology and bind to phospholipid membranes via tandem, lectin-like C domains. Their respective C2 domains bind via 2 pairs of hydrophobic amino acids and an amphipathic cluster. In contrast, the factor V-like, homologous subunit (Pt-FV) of a prothrombin activator from Pseudonaja textilis venom is reported to function without membrane binding. We hypothesized that the distinct membrane-interactive amino acids of these proteins contribute to the differing membrane-dependent properties. We prepared mutants in which the C2 domain hydrophobic amino acid pairs were changed to the homologous residues of the other protein and a factor V mutant with 5 amino acids changed to those from Pt-FV (FV(MTTS/Y)). Factor VIII mutants were active on additional membrane sites and had altered apparent affinities for factor X. Some factor V mutants, including FV(MTTS/Y), had increased membrane interaction and apparent membrane-independent activity that was the result of phospholipid retained during purification. Phospholipid-free FV(MTTS/Y) showed increased activity, particularly a 10-fold increase in activity on membranes lacking phosphatidylserine. The reduced phosphatidylserine requirement correlated to increased activity on resting and stimulated platelets. We hypothesize that altered membrane binding contributes to toxicity of Pt-FV.


Asunto(s)
Factor VIII/química , Factor V/química , Mutación , Fosfolípidos/química , Estructura Terciaria de Proteína , Algoritmos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Unión Competitiva , Membrana Celular/química , Membrana Celular/metabolismo , Secuencia Conservada , Venenos Elapídicos/química , Venenos Elapídicos/genética , Venenos Elapídicos/metabolismo , Factor V/genética , Factor V/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Datos de Secuencia Molecular , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Fosfolípidos/metabolismo , Unión Proteica , Homología de Secuencia de Aminoácido
9.
Leukemia ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890447

RESUMEN

Chromosomal translocations of the nucleoporin 98 (NUP98) gene are found in acute myeloid leukemia (AML) patients leading to very poor outcomes. The oncogenic activity of NUP98 fusion proteins is dependent on the interaction between Mixed Lineage Leukemia 1 and menin. NUP98-rearranged (NUP98-r) leukemia cells also rely on specific kinases, including CDK6 and/or FLT3, suggesting that simultaneous targeting of these kinases and menin could overcome limited sensitivity to single agents. Here, we found that combinations of menin inhibitor, MI-3454, with kinase inhibitors targeting either CDK6 (Palbociclib) or FLT3 (Gilteritinib) strongly enhance the anti-leukemic effect of menin inhibition in NUP98-r leukemia models. We found strong synergistic effects of both combinations on cell growth, colony formation and differentiation in patient samples with NUP98 translocations. These combinations also markedly augmented anti-leukemic efficacy of menin inhibitor in Patient Derived Xenograft models of NUP98-r leukemia. Despite inhibiting two unrelated kinases, when Palbociclib or Gilteritinib were combined with the menin inhibitor, they affected similar pathways relevant to leukemogenesis, including cell cycle regulation, cell proliferation and differentiation. This study provides strong rationale for clinical translation of the combination of menin and kinase inhibitors as novel treatments for NUP98-r leukemia, supporting the unexplored combinations of epigenetic drugs with kinase inhibitors.

10.
Blood ; 117(11): 3181-9, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21156843

RESUMEN

Factor VIII binds to phosphatidylserine (PS)-containing membranes through its tandem, lectin-homology, C1 and C2 domains. However, the details of C1 domain membrane binding have not been delineated. We prepared 4 factor VIII C1 mutations localized to a hypothesized membrane-interactive surface (Arg2090Ala/Gln2091Ala, Lys2092Ala/Phe2093Ala, Gln2042Ala/Tyr2043Ala, and Arg2159Ala). Membrane binding and cofactor activity were measured using membranes with 15% PS, mimicking platelets stimulated by thrombin plus collagen, and 4% PS, mimicking platelets stimulated by thrombin. All mutants had at least 10-fold reduced affinities for membranes of 4% PS, and 3 mutants also had decreased apparent affinity for factor X. Monoclonal antibodies against the C2 domain produced different relative impairment of mutants compared with wild-type factor VIII. Monoclonal antibody ESH4 decreased the V(max) for all mutants but only the apparent membrane affinity for wild-type factor VIII. Monoclonal antibody BO2C11 decreased the V(max) of wild-type factor VIII by 90% but decreased the activity of 3 mutants more than 98%. These results identify a membrane-binding face of the factor VIII C1 domain, indicate an influence of the C1 domain on factor VIII binding to factor X, and indicate that cooperation between the C1 and C2 domains is necessary for full activity of the factor Xase complex.


Asunto(s)
Membrana Celular/metabolismo , Factor VIII/química , Factor VIII/metabolismo , Factor X/metabolismo , Aminoácidos/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Bovinos , Membrana Celular/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas de Neoplasias/metabolismo , Fosfolípidos/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Propiedades de Superficie/efectos de los fármacos , Factor de von Willebrand/metabolismo
11.
Zhonghua Yi Xue Za Zhi ; 93(8): 606-9, 2013 Feb 26.
Artículo en Zh | MEDLINE | ID: mdl-23663343

RESUMEN

OBJECTIVE: To explore the cell signal transduction pathway of calcium-sensing receptor (CaSR) mediated hypoxia-induced proliferation of rat pulmonary artery smooth muscle cells (PASMCs). METHODS: The expressions of proliferating cell nuclear antigen (PCNA) and extracellular signal-regulated protein kinase 1, 2 (ERK1, 2) were analyzed by Western blot. Cell proliferation was tested by a 5-bromo-2-deoxyuridine (BrdU) incorporation assay. Cell cycle and proliferation index (PI) were analyzed by flow cytometry. RESULTS: Hypoxia significantly increased the expression of PCNA (0.528 ± 0.028), p-ERK1, 2 (1.12 ± 0.05, 0.91 ± 0.06), BrdU incorporation (143.3 ± 4.2) and cell proliferation index (12.5 ± 0.9) (all P < 0.05, versus control group, 0.243 ± 0.025, 0.47 ± 0.03, 0.40 ± 0.03, 100.0 ± 5.4, 7.5 ± 1.2). Gadolinium chloride (GdCl3, a CaSR agonist) amplified the effect of hypoxia (0.770 ± 0.039, 1.50 ± 0.06, 1.61 ± 0.05, 187.4 ± 3.9, 19.8 ± 0.6, all P < 0.05). PD98059 (a MEK1 inhibitor) decreased the up-regulation of PCNA expression, BrdU incorporation and the increase of cell proliferation index induced by hypoxia and GdCl3 in PASMCs (0.441 ± 0.020, 0.71 ± 0.07, 0.72 ± 0.06, 115.5 ± 4.0, 9.3 ± 1.1, all P < 0.05). CONCLUSION: Calcium-sensing receptor mediates hypoxia-induced proliferation of rat pulmonary artery smooth muscle cells through ERK1, 2 pathways.


Asunto(s)
MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos del Músculo Liso/citología , Animales , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Miocitos del Músculo Liso/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Ratas , Ratas Wistar
12.
Cancer Discov ; 13(3): 724-745, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36455589

RESUMEN

Nucleophosmin (NPM1) is a ubiquitously expressed nucleolar protein with a wide range of biological functions. In 30% of acute myeloid leukemia (AML), the terminal exon of NPM1 is often found mutated, resulting in the addition of a nuclear export signal and a shift of the protein to the cytoplasm (NPM1c). AMLs carrying this mutation have aberrant expression of the HOXA/B genes, whose overexpression leads to leukemogenic transformation. Here, for the first time, we comprehensively prove that NPM1c binds to a subset of active gene promoters in NPM1c AMLs, including well-known leukemia-driving genes-HOXA/B cluster genes and MEIS1. NPM1c sustains the active transcription of key target genes by orchestrating a transcription hub and maintains the active chromatin landscape by inhibiting the activity of histone deacetylases. Together, these findings reveal the neomorphic function of NPM1c as a transcriptional amplifier for leukemic gene expression and open up new paradigms for therapeutic intervention. SIGNIFICANCE: NPM1 mutation is the most common mutation in AML, yet the mechanism of how the mutant protein results in AML remains unclear. Here, for the first time, we prove mutant NPM1 directly binds to active chromatin regions and hijacks the transcription of AML-driving genes. See related article by Uckelmann et al., p. 746. This article is highlighted in the In This Issue feature, p. 517.


Asunto(s)
Leucemia Mieloide Aguda , Nucleofosmina , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Mutación , Cromatina/genética
13.
Blood ; 116(26): 6114-22, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20852129

RESUMEN

Ectopically expressed, human B-domainless (hB) factor 8 (F8) in platelets improves hemostasis in hemophilia A mice in several injury models. However, in both a cuticular bleeding model and a cremaster laser arteriole/venule injury model, there were limitations to platelet-derived (p) hBF8 efficacy, including increased clot embolization. We now address whether variants of F8 with enhanced activity, inactivation resistant F8 (IR8) and canine (c) BF8, would improve clotting efficacy. In both transgenic and lentiviral murine model approaches, pIR8 expressed at comparable levels to phBF8, but pcBF8 expressed at only approximately 30%. Both variants were more effective than hBF8 in cuticular bleeding and FeCl(3) carotid artery models. However, in the cremaster injury model, only pcBF8 was more effective, markedly decreasing clot embolization. Because inhibitors of F8 are stored in platelet granules and IR8 is not protected by binding to von Willebrand factor, we also tested whether pIR8 was effective in the face of inhibitors and found that pIR8 is protected from the inhibitors. In summary, pF8 variants with high specific activity are more effective in controlling bleeding, but this improved efficacy was inconsistent between bleeding models, perhaps reflecting the underlying mechanism(s) for the increased specific activity of the studied F8 variants.


Asunto(s)
Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Factor VIII/genética , Hemofilia A/prevención & control , Hemorragia/prevención & control , Trombosis/prevención & control , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Perros , Factor VIII/metabolismo , Humanos , Ratones , Ratones Transgénicos
14.
PeerJ ; 10: e13658, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35833015

RESUMEN

Restoration is the natural and intervention-assisted set of processes designed to promote and facilitate the recovery of an ecosystem that has been degraded, damaged, or destroyed. However, it can also have an adverse effect on the environment. Thus, assessing an ecological restoration project's impact is crucial to determining its success and optimum management strategies. We performed a meta-analysis concerning the environmental outcomes during the years 2000-2015 resulting from the "Grain for Green" Project (GFGP) implementation in the Loess Plateau (LP). Data were gathered from 40 peer-reviewed English-language articles chosen from a pool of 332 articles. The results showed that, on average, GFGP increased forest coverage by 35.7% (95% CI [24.15-47.52%]), and grassland by 1.05% (95% CI [0.8-1.28%]). At the same time, GFGP has a positive impact on soil carbon (C) sequestration, net ecosystem production (NEP), and net primary production (NPP), from the years 2000 to 2015 by an average of 36% (95% CI [28.96-43.18%]), 22.7% (95% CI [9.10-36.79%]), and 13.5% (95% CI [9.44-17.354%]), respectively. Soil erosion, sediment load, runoff coefficient, and water yield were reduced by 13.3% (95% CI [0.27-25.76%]), 21.5% (95% CI [1.50-39.99%]), 22.4% (95% CI [5.28-40.45%]) and 43.3% (95% CI [27.03-82.86%]), respectively, from the years 2000 to 2015. Our results indicate that water supply decreased with the increase of vegetation coverage. Therefore, to balance the needs for green space, GFGP policies and strategies should recover, enhance, and sustain more resilient ecosystems.


Asunto(s)
Ecosistema , Agua , Suelo , Bosques , Abastecimiento de Agua
16.
Proc Natl Acad Sci U S A ; 105(47): 18525-30, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19011102

RESUMEN

Protein misfolding in the endoplasmic reticulum (ER) contributes to the pathogenesis of many diseases. Although oxidative stress can disrupt protein folding, how protein misfolding and oxidative stress impact each other has not been explored. We have analyzed expression of coagulation factor VIII (FVIII), the protein deficient in hemophilia A, to elucidate the relationship between protein misfolding and oxidative stress. Newly synthesized FVIII misfolds in the ER lumen, activates the unfolded protein response (UPR), causes oxidative stress, and induces apoptosis in vitro and in vivo in mice. Strikingly, antioxidant treatment reduces UPR activation, oxidative stress, and apoptosis, and increases FVIII secretion in vitro and in vivo. The findings indicate that reactive oxygen species are a signal generated by misfolded protein in the ER that cause UPR activation and cell death. Genetic or chemical intervention to reduce reactive oxygen species improves protein folding and cell survival and may provide an avenue to treat and/or prevent diseases of protein misfolding.


Asunto(s)
Antioxidantes/farmacología , Retículo Endoplásmico/efectos de los fármacos , Factor VIII/metabolismo , Animales , Apoptosis , Células CHO , Cricetinae , Cricetulus , Retículo Endoplásmico/metabolismo , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Desnaturalización Proteica
17.
Environ Pollut ; 287: 117598, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34147777

RESUMEN

A comprehensive understanding of the patterns and controlling factors of nitrate accumulation in intensive vegetable production is essential to solve this problem. For the first time, the national patterns and controlling factors of nitrate accumulation in soil of vegetable systems in China were analysed by compiling 1262 observations from 117 published articles. The results revealed that the nitrate accumulation at 0-100 cm, 100-200 cm, 200-300 cm, and >300 cm were 504, 390, 349, and 244 kg N ha-1, with accumulation rates of 62, 54, 19, and 16 kg N ha-1 yr-1 for plastic greenhouse vegetables (PG); for open field vegetables (OF), they were 264, 217, 228, and 242 kg N ha-1 with accumulation rates of 26, 24, 18, and 10 kg N ha-1 yr-1, respectively. Nitrate accumulation at 0-100 cm, 0-200 cm, and 0-400 cm accounted for 5%, 11%, and 17% of accumulated nitrogen (N) inputs for PG, and represented 4%, 9%, and 13% of accumulated N inputs for OF. Nitrogen input rates and soil pH had positive effects and soil organic carbon, water input rate, and carbon to nitrogen ratio (C/N) had negative effects on nitrate accumulation in root zone (0-100 cm soil). Nitrate accumulation in deep vadose zone (>100 cm soil) was positively correlated with N and water input rates, and was negatively correlated with soil organic carbon, C/N, and the clay content. Thus, for a given vegetable soil with relatively stable soil pH and soil clay content, reducing N and water inputs, and increasing soil organic carbon and C/N are effective measures to control nitrate accumulation.


Asunto(s)
Nitratos , Suelo , Agricultura , Carbono , China , Fertilizantes/análisis , Nitratos/análisis , Nitrógeno/análisis , Verduras
18.
Nat Commun ; 12(1): 2792, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990599

RESUMEN

ASH1L histone methyltransferase plays a crucial role in the pathogenesis of different diseases, including acute leukemia. While ASH1L represents an attractive drug target, developing ASH1L inhibitors is challenging, as the catalytic SET domain adapts an inactive conformation with autoinhibitory loop blocking the access to the active site. Here, by applying fragment-based screening followed by medicinal chemistry and a structure-based design, we developed first-in-class small molecule inhibitors of the ASH1L SET domain. The crystal structures of ASH1L-inhibitor complexes reveal compound binding to the autoinhibitory loop region in the SET domain. When tested in MLL leukemia models, our lead compound, AS-99, blocks cell proliferation, induces apoptosis and differentiation, downregulates MLL fusion target genes, and reduces the leukemia burden in vivo. This work validates the ASH1L SET domain as a druggable target and provides a chemical probe to further study the biological functions of ASH1L as well as to develop therapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Animales , Antineoplásicos/química , Dominio Catalítico/efectos de los fármacos , Dominio Catalítico/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Diseño de Fármacos , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Femenino , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Leucemia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Proteína de la Leucemia Mieloide-Linfoide/genética , Oncogenes , Dominios Proteicos , Proteínas Recombinantes de Fusión/genética
19.
J Clin Invest ; 130(2): 981-997, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31855575

RESUMEN

The protein-protein interaction between menin and mixed lineage leukemia 1 (MLL1) plays a critical role in acute leukemias with translocations of the MLL1 gene or with mutations in the nucleophosmin 1 (NPM1) gene. As a step toward clinical translation of menin-MLL1 inhibitors, we report development of MI-3454, a highly potent and orally bioavailable inhibitor of the menin-MLL1 interaction. MI-3454 profoundly inhibited proliferation and induced differentiation in acute leukemia cells and primary patient samples with MLL1 translocations or NPM1 mutations. When applied as a single agent, MI-3454 induced complete remission or regression of leukemia in mouse models of MLL1-rearranged or NPM1-mutated leukemia, including patient-derived xenograft models, through downregulation of key genes involved in leukemogenesis. We also identified MEIS1 as a potential pharmacodynamic biomarker of treatment response with MI-3454 in leukemia, and demonstrated that this compound is well tolerated and did not impair normal hematopoiesis in mice. Overall, this study demonstrates, for the first time to our knowledge, profound activity of the menin-MLL1 inhibitor as a single agent in clinically relevant PDX models of leukemia. These data provide a strong rationale for clinical translation of MI-3454 or its analogs for leukemia patients with MLL1 rearrangements or NPM1 mutations.


Asunto(s)
Antineoplásicos/farmacología , N-Metiltransferasa de Histona-Lisina , Leucemia , Mutación , Proteína de la Leucemia Mieloide-Linfoide , Neoplasias Experimentales , Proteínas Nucleares , Proteínas Proto-Oncogénicas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Células K562 , Leucemia/tratamiento farmacológico , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Inducción de Remisión , Células U937
20.
ACS Chem Biol ; 13(9): 2739-2746, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30071723

RESUMEN

GAS41 is a chromatin-associated protein that belongs to the YEATS family and is involved in the recognition of acetyl-lysine in histone proteins. A unique feature of GAS41 is the presence of a C-terminal coiled-coil domain, which is responsible for protein dimerization. Here, we characterized the specificity of the GAS41 YEATS domain and found that it preferentially binds to acetylated H3K18 and H3K27 peptides. Interestingly, we found that full-length, dimeric GAS41 binds to diacetylated H3 peptides with an enhanced affinity when compared to those for monoacetylated peptides, through a bivalent binding mode. We determined the crystal structure of the GAS41 YEATS domain with H3K23acK27ac to visualize the molecular basis of diacetylated histone binding. Our results suggest a unique binding mode in which full-length GAS41 is a reader of diacetylated histones.


Asunto(s)
Histonas/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Sitios de Unión , Células HEK293 , Histonas/química , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA