Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Genome Biol ; 25(1): 143, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822412

RESUMEN

BACKGROUND: Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence. RESULTS: Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers. CONCLUSIONS: Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Pirimidinonas/farmacología , Pirimidinonas/uso terapéutico , Proteína de la Leucemia Mieloide-Linfoide/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética
2.
Sci Rep ; 14(1): 9104, 2024 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643249

RESUMEN

Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common among children. AML is characterized by aberrant proliferation of myeloid blasts in the bone marrow and impaired normal hematopoiesis. Despite the introduction of new drugs and allogeneic bone marrow transplantation, patients have poor overall survival rate with relapse as the major challenge, driving the demand for new therapeutic strategies. AML patients with high expression of the very long/long chain fatty acid transporter CD36 have poorer survival and very long chain fatty acid metabolism is critical for AML cell survival. Here we show that fatty acids are transferred from human primary adipocytes to AML cells upon co-culturing. A drug-like small molecule (SMS121) was identified by receptor-based virtual screening and experimentally demonstrated to target the lipid uptake protein CD36. SMS121 reduced the uptake of fatty acid into AML cells that could be reversed by addition of free fatty acids and caused decreased cell viability. The data presented here serves as a framework for the development of CD36 inhibitors to be used as future therapeutics against AML.


Asunto(s)
Ácidos Grasos , Leucemia Mieloide Aguda , Adulto , Niño , Humanos , Ácidos Grasos/uso terapéutico , Leucemia Mieloide Aguda/metabolismo , Médula Ósea/metabolismo , Enfermedad Aguda , Técnicas de Cocultivo
3.
Blood Adv ; 7(18): 5382-5395, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37505194

RESUMEN

Acute myeloid leukemia (AML) is initiated and propagated by leukemia stem cells (LSCs), a self-renewing population of leukemia cells responsible for therapy resistance. Hence, there is an urgent need to identify new therapeutic opportunities targeting LSCs. Here, we performed an in vivo CRISPR knockout screen to identify potential therapeutic targets by interrogating cell surface dependencies of LSCs. The facilitated glucose transporter type 1 (GLUT1) emerged as a critical in vivo metabolic dependency for LSCs in a murine MLL::AF9-driven model of AML. GLUT1 disruption by genetic ablation or pharmacological inhibition led to suppression of leukemia progression and improved survival of mice that received transplantation with LSCs. Metabolic profiling revealed that Glut1 inhibition suppressed glycolysis, decreased levels of tricarboxylic acid cycle intermediates and increased the levels of amino acids. This metabolic reprogramming was accompanied by an increase in autophagic activity and apoptosis. Moreover, Glut1 disruption caused transcriptional, morphological, and immunophenotypic changes, consistent with differentiation of AML cells. Notably, dual inhibition of GLUT1 and oxidative phosphorylation (OXPHOS) exhibited synergistic antileukemic effects in the majority of tested primary AML patient samples through restraining of their metabolic plasticity. In particular, RUNX1-mutated primary leukemia cells displayed striking sensitivity to the combination treatment compared with normal CD34+ bone marrow and cord blood cells. Collectively, our study reveals a GLUT1 dependency of murine LSCs in the bone marrow microenvironment and demonstrates that dual inhibition of GLUT1 and OXPHOS is a promising therapeutic approach for AML.


Asunto(s)
Leucemia Mieloide Aguda , Fosforilación Oxidativa , Animales , Ratones , Apoptosis , Médula Ósea/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Leucemia Mieloide Aguda/genética , Microambiente Tumoral
4.
Hemasphere ; 6(10): e785, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36204688

RESUMEN

Infant acute lymphoblastic leukemia (ALL) with KMT2A-gene rearrangements (KMT2A-r) have few mutations and a poor prognosis. To uncover mutations that are below the detection of standard next-generation sequencing (NGS), a combination of targeted duplex sequencing and NGS was applied on 20 infants and 7 children with KMT2A-r ALL, 5 longitudinal and 6 paired relapse samples. Of identified nonsynonymous mutations, 87 had been previously implicated in cancer and targeted genes recurrently altered in KMT2A-r leukemia and included mutations in KRAS, NRAS, FLT3, TP53, PIK3CA, PAX5, PIK3R1, and PTPN11, with infants having fewer such mutations. Of identified cancer-associated mutations, 62% were below the resolution of standard NGS. Only 33 of 87 mutations exceeded 2% of cellular prevalence and most-targeted PI3K/RAS genes (31/33) and typically KRAS/NRAS. Five patients only had low-frequency PI3K/RAS mutations without a higher-frequency signaling mutation. Further, drug-resistant clones with FLT3 D835H or NRAS G13D/G12S mutations that comprised only 0.06% to 0.34% of diagnostic cells, expanded at relapse. Finally, in longitudinal samples, the relapse clone persisted as a minor subclone from diagnosis and through treatment before expanding during the last month of disease. Together, we demonstrate that infant and childhood KMT2A-r ALL harbor low-frequency cancer-associated mutations, implying a vast subclonal genetic landscape.

7.
J Alzheimers Dis ; 64(s1): S547-S554, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29758944

RESUMEN

Spanning over three decades of extensive drug discovery research, the efforts to develop a potent and selective GSK3 inhibitor as a therapeutic for the treatment of type 2 diabetes, Alzheimer's disease (AD), bipolar disorders and cancer have been futile. Since its initial discovery in 1980 and subsequent decades of research, one cannot underscore the importance of the target and the promise of a game changing disease modifier. Several pharmaceutical companies, biotech companies, and academic institutions raged in a quest to unravel the biology and discover potent and selective GSK3 inhibitors, some of which went through clinical trials. However, the conundrum of what happened to the fate of the AstraZeneca's GSK3 inhibitors and the undertaking to find a therapeutic that could control glycogen metabolism and aberrant tau hyperphosphorylation in the brain, and rescue synaptic dysfunction has largely been untold. AstraZeneca was in the forefront of GSK3 drug discovery research with six GSK3 drug candidates, one of which progressed up to Phase II clinical trials in the quest to untangle the tau hypothesis for AD. Analysis of key toxicity issues, serendipitous findings and efficacy, and biomarker considerations in relation to safety margins have limited the potential of small molecule therapeutics as a way forward. To guide future innovation of this important target, we reveal the roller coaster journey comprising of two decades of preclinical and clinical GSK3 drug discovery at AstraZeneca; the understanding of which could lead to improved GSK3 therapies for disease. These learnings in combination with advances in achieving kinase selectivity, different modes of action as well as the recent discovery of novel conjugated peptide technology targeting specific tissues have potentially provided a venue for scientific innovation and a new beginning for GSK3 drug discovery.


Asunto(s)
Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Animales , Ensayos Clínicos como Asunto , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos
8.
Nat Commun ; 9(1): 1770, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29720585

RESUMEN

Activating signaling mutations are common in acute leukemia with KMT2A (previously MLL) rearrangements (KMT2A-R). These mutations are often subclonal and their biological impact remains unclear. Using a retroviral acute myeloid mouse leukemia model, we demonstrate that FLT3 ITD , FLT3 N676K , and NRAS G12D accelerate KMT2A-MLLT3 leukemia onset. Further, also subclonal FLT3 N676K mutations accelerate disease, possibly by providing stimulatory factors. Herein, we show that one such factor, MIF, promotes survival of mouse KMT2A-MLLT3 leukemia initiating cells. We identify acquired de novo mutations in Braf, Cbl, Kras, and Ptpn11 in KMT2A-MLLT3 leukemia cells that favored clonal expansion. During clonal evolution, we observe serial genetic changes at the Kras G12D locus, consistent with a strong selective advantage of additional Kras G12D . KMT2A-MLLT3 leukemias with signaling mutations enforce Myc and Myb transcriptional modules. Our results provide new insight into the biology of KMT2A-R leukemia with subclonal signaling mutations and highlight the importance of activated signaling as a contributing driver.


Asunto(s)
Evolución Clonal , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide/genética , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Enfermedad Aguda , Animales , Línea Celular Tumoral , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
9.
Nat Genet ; 48(12): 1551-1556, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27798625

RESUMEN

Acute myeloid leukemia (AML) comprises a heterogeneous group of leukemias frequently defined by recurrent cytogenetic abnormalities, including rearrangements involving the core-binding factor (CBF) transcriptional complex. To better understand the genomic landscape of CBF-AMLs, we analyzed both pediatric (n = 87) and adult (n = 78) samples, including cases with RUNX1-RUNX1T1 (n = 85) or CBFB-MYH11 (n = 80) rearrangements, by whole-genome or whole-exome sequencing. In addition to known mutations in the Ras pathway, we identified recurrent stabilizing mutations in CCND2, suggesting a previously unappreciated cooperating pathway in CBF-AML. Outside of signaling alterations, RUNX1-RUNX1T1 and CBFB-MYH11 AMLs demonstrated remarkably different spectra of cooperating mutations, as RUNX1-RUNX1T1 cases harbored recurrent mutations in DHX15 and ZBTB7A, as well as an enrichment of mutations in epigenetic regulators, including ASXL2 and the cohesin complex. This detailed analysis provides insights into the pathogenesis and development of CBF-AML, while highlighting dramatic differences in the landscapes of cooperating mutations for these related AML subtypes.


Asunto(s)
Biomarcadores de Tumor/genética , Factores de Unión al Sitio Principal/genética , Genómica/métodos , Leucemia Mieloide Aguda/genética , Mutación/genética , Proteínas de Fusión Oncogénica/genética , Adulto , Niño , Humanos
10.
J Pharm Sci ; 105(9): 2913-2920, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27431012

RESUMEN

Cyclodextrins (CD) have the ability to form inclusion complexes with drugs and can be used as excipients to enhance solubility of poorly soluble drugs. To make accurate estimations of the potency of the drug, knowledge of the free drug concentration is important. The aim of this study was to evaluate the applicability of calculated free drug concentrations toward response measurements in a transient receptor potential vanilloid receptor-1 cell-based in vitro assay. This included accounting for potential competitive CD binding of 2 transient receptor potential vanilloid receptor-1 active entities: 1 antagonist, and 1 agonist (capsaicin). Solubility of the CD-drug complexes was measured, and the ligand to substrate affinity in CD formulations was determined according to the phase-solubility technique. The total concentration of antagonist, agonist, CD, and the binding constants between ligands and CD were used to calculate the free concentration of CD ligands. For capsaicin and 2 of the 3 investigated model drugs, the calculated free drug concentration was consistent with the experimental in vitro data while it was overestimated for one of the compounds. In conclusion, the suggested approach can be used to calculate free drug concentration and competitive binding in CD formulations for the application of cell-based drug functionality assays.


Asunto(s)
Ciclodextrinas/química , Preparaciones Farmacéuticas/análisis , Animales , Células CHO , Capsaicina/química , Capsaicina/farmacología , Línea Celular , Química Farmacéutica , Cricetinae , Cricetulus , Composición de Medicamentos , Excipientes/química , Humanos , Ligandos , Solubilidad , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores
11.
Genes Chromosomes Cancer ; 55(11): 847-54, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27240832

RESUMEN

Myelodysplastic/myeloproliferative neoplasms, unclassifiable (MDS/MPN-U) are rare genetically heterogeneous hematologic diseases associated with older age and a poor prognosis. If the disease progresses into acute myeloid leukemia (AML), it is often refractory to treatment. To gain insight into genetic alterations associated with disease progression, whole exome sequencing and single nucleotide polymorphism arrays were used to characterize the bone marrow and blood samples from a 39-year-old woman at MDS/MPN-U diagnosis and at AML progression, in which routine genetic diagnostics had not identified any genetic alterations. The data revealed the presence of a partial tandem duplication of the MLL gene as the only detectable copy number change and 11 non-silent somatic mutations, including DNMT3A R882H and NRAS G13D. All somatic lesions were present both at initial MDS/MPN-U diagnosis and at AML presentation at similar mutant allele frequencies. The patient has since had two extramedullary relapses and is at high risk of a future bone marrow relapse. A directed ex vivo drug sensitivity analysis showed that the patient's AML cells are sensitive to, for example, the MEK inhibitor trametinib and the proteasome inhibitor bortezomib, indicating that she may benefit from treatment with these drugs. © 2016 Wiley Periodicals, Inc.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , GTP Fosfohidrolasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de la Membrana/genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Proteína de la Leucemia Mieloide-Linfoide/genética , Adulto , Bortezomib/administración & dosificación , ADN Metiltransferasa 3A , Progresión de la Enfermedad , Femenino , Duplicación de Gen , Frecuencia de los Genes , Heterogeneidad Genética , Genoma Humano , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación
12.
Nat Commun ; 7: 11790, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27265895

RESUMEN

Fusion genes are potent driver mutations in cancer. In this study, we delineate the fusion gene landscape in a consecutive series of 195 paediatric B-cell precursor acute lymphoblastic leukaemia (BCP ALL). Using RNA sequencing, we find in-frame fusion genes in 127 (65%) cases, including 27 novel fusions. We describe a subtype characterized by recurrent IGH-DUX4 or ERG-DUX4 fusions, representing 4% of cases, leading to overexpression of DUX4 and frequently co-occurring with intragenic ERG deletions. Furthermore, we identify a subtype characterized by an ETV6-RUNX1-like gene-expression profile and coexisting ETV6 and IKZF1 alterations. Thus, this study provides a detailed overview of fusion genes in paediatric BCP ALL and adds new pathogenetic insights, which may improve risk stratification and provide therapeutic options for this disease.


Asunto(s)
Reordenamiento Génico/genética , Proteínas de Homeodominio/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Empalme Alternativo/genética , Niño , Rotura Cromosómica , Análisis por Conglomerados , Estudios de Cohortes , Análisis Mutacional de ADN , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Análisis de Componente Principal
13.
Nat Genet ; 47(4): 330-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25730765

RESUMEN

Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole-genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non-MLL-R cases) and 20 older children (MLL-R cases) with leukemia. Our data show that infant MLL-R ALL has one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite this paucity of mutations, we detected activating mutations in kinase-PI3K-RAS signaling pathway components in 47% of cases. Surprisingly, these mutations were often subclonal and were frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (mean of 6.5 mutations/case versus 1.3 mutations/case, P = 7.15 × 10(-5)) and had frequent mutations (45%) in epigenetic regulators, a category of genes that, with the exception of MLL, was rarely mutated in infant MLL-R ALL.


Asunto(s)
Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Desequilibrio Alélico/genética , Estudios de Cohortes , Análisis Mutacional de ADN , Frecuencia de los Genes , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Proteínas de Fusión Oncogénica/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/genética , Proteínas ras/genética , Proteínas ras/metabolismo
14.
J Clin Invest ; 123(7): 3099-111, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23921123

RESUMEN

Acute lymphoblastic leukemia (ALL) is the commonest childhood malignancy and is characterized by recurring structural genetic alterations. Previous studies of DNA methylation suggest epigenetic alterations may also be important, but an integrated genome-wide analysis of genetic and epigenetic alterations in ALL has not been performed. We analyzed 137 B-lineage and 30 T-lineage childhood ALL cases using microarray analysis of DNA copy number alterations and gene expression, and genome-wide cytosine methylation profiling using the HpaII tiny fragment enrichment by ligation-mediated PCR (HELP) assay. We found that the different genetic subtypes of ALL are characterized by distinct DNA methylation signatures that exhibit significant correlation with gene expression profiles. We also identified an epigenetic signature common to all cases, with correlation to gene expression in 65% of these genes, suggesting that a core set of epigenetically deregulated genes is central to the initiation or maintenance of lymphoid transformation. Finally, we identified aberrant methylation in multiple genes also targeted by recurring DNA copy number alterations in ALL, suggesting that these genes are inactivated far more frequently than suggested by structural genomic analyses alone. Together, these results demonstrate subtype- and disease-specific alterations in cytosine methylation in ALL that influence transcriptional activity, and are likely to exert a key role in leukemogenesis.


Asunto(s)
Epigénesis Genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transformación Celular Neoplásica/genética , Niño , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Metilación de ADN , Regulación Leucémica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Transducción de Señal , Transcriptoma
15.
Cancer Cell ; 22(5): 683-97, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23153540

RESUMEN

To define the mutation spectrum in non-Down syndrome acute megakaryoblastic leukemia (non-DS-AMKL), we performed transcriptome sequencing on diagnostic blasts from 14 pediatric patients and validated our findings in a recurrency/validation cohort consisting of 34 pediatric and 28 adult AMKL samples. Our analysis identified a cryptic chromosome 16 inversion (inv(16)(p13.3q24.3)) in 27% of pediatric cases, which encodes a CBFA2T3-GLIS2 fusion protein. Expression of CBFA2T3-GLIS2 in Drosophila and murine hematopoietic cells induced bone morphogenic protein (BMP) signaling and resulted in a marked increase in the self-renewal capacity of hematopoietic progenitors. These data suggest that expression of CBFA2T3-GLIS2 directly contributes to leukemogenesis.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Niño , Inversión Cromosómica , Cromosomas Humanos Par 16 , Drosophila/genética , Drosophila/crecimiento & desarrollo , Perfilación de la Expresión Génica , Humanos , Leucemia Megacarioblástica Aguda/clasificación , Leucemia Megacarioblástica Aguda/diagnóstico , Ratones , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Pronóstico , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/fisiología , Análisis de Secuencia de ARN , Transducción de Señal
16.
Bone ; 50(3): 619-27, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22142634

RESUMEN

Glycogen synthase kinase 3ß (GSK-3ß) actions are central in the canonical Wnt pathway, important in many biological processes and a potential drug target for treating several diseases. It is appreciated that a balanced Wnt canonical signaling is crucial for the maintenance of normal bone mass. In this study we investigated the effects of a potent orally active GSK-3 inhibitor, AZD2858, on bone mass in rats. Treatment (1 µM) of human osteoblast cells with AZD2858 in vitro increased ß-catenin levels after a short period of time. In rats, oral AZD2858 treatment caused a dose-dependent increase in trabecular bone mass compared to control after a two-week treatment with a maximum effect at a dose of 20 mg/kg once daily (total BMC: 172% of control; p<0.001). A small but significant effect was also seen at cortical sites (total BMC: 111% of control; p<0.001). Biomechanical testing demonstrated an increase in both vertebral compression strength at a dose of 20 mg/kg once daily (Load at failure: 370% of control, p<0.001) and diaphyseal strength of femora subjected to a three point bending test (Load at failure: 115% of control; p<0.01). Furthermore, histomorphometry showed a dramatic increase in bone formation indices, and serum markers of both bone formation (Osteocalcin, 146% of control; p<0.001) and resorption (CTX, 189% of control; p<0.001) were elevated. Our conclusion is that a GSK-3 inhibitor drug may prove effective as an anabolic strategy in the treatment of diseases characterized by low bone mass, since AZD2858 has extensive bone building effects at predominantly trabecular sites.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Osteoblastos/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Animales , Remodelación Ósea/efectos de los fármacos , Huesos/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Osteoblastos/metabolismo , Ratas , Ratas Sprague-Dawley , beta Catenina/metabolismo
17.
Arthritis Rheum ; 63(9): 2617-29, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21618462

RESUMEN

OBJECTIVE: To assess the role of the activating receptor NKG2D in arthritis. METHODS: Levels of NKG2D and its ligands were determined by fluorescence-activated cell sorting, real-time polymerase chain reaction, and immunohistochemistry in rheumatoid arthritis (RA) synovial membrane tissue and in paw tissue from arthritic mice. Arthritis was induced in DBA/1 mice by immunization with type II collagen, and mice were treated intraperitoneally with a blocking anti-NKG2D antibody (CX5) on days 1, 5, and 8 after clinical onset and were monitored for 10 days. RESULTS: We demonstrated expression of NKG2D and its ligands on human RA synovial cells and extended this finding to the paws of arthritic mice. Expression of messenger RNA for the NKG2D ligand Rae-1 was up-regulated, and NKG2D was present predominantly on natural killer (NK) and CD4+ T cells, in arthritic paw cell isolates. NKG2D was down-modulated during the progression of collagen-induced arthritis (CIA). NKG2D expression in arthritic paws was demonstrated by immunohistochemistry. Blockade of NKG2D ameliorated established CIA, with significant reductions in clinical scores and paw swelling. Histologic analysis of arthritic joints from anti-NKG2D-treated mice demonstrated significant joint protection, compared with control mice. Moreover, anti-NKG2D treatment significantly reduced both interleukin-17 production from CD4+ T cells in arthritic paws and splenic NK cell cytotoxic effector functions in vivo and in vitro. CONCLUSION: Our findings indicate that blockade of NKG2D in a murine model and in human explants has beneficial therapeutic potential that merits further investigation in RA.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Subfamilia K de Receptores Similares a Lectina de Células NK/antagonistas & inhibidores , Membrana Sinovial/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Artritis Experimental/inmunología , Artritis Experimental/patología , Células Cultivadas , Técnicas de Cocultivo , Articulaciones/inmunología , Articulaciones/patología , Células Asesinas Naturales/inmunología , Masculino , Ratones , Ratones Endogámicos DBA , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Membrana Sinovial/patología
18.
Accid Anal Prev ; 43(1): 284-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21094326

RESUMEN

Winter weather can be a significant cause of road traffic accidents. This paper uses UKCIP climate change scenarios and a temporal analogue to investigate the relationship between temperature and severe road accidents in the West Midlands, UK. This approach also allows quantification of the changes in the severity of the winter season over the next century in the region. It is demonstrated that the predicted reduction in the number of frost days should in turn reduce the number of road accidents caused due to slipperiness by approximately 50%. However, the paper concludes by warning against complacency in winter maintenance regimes. A warmer climate may result in budget cuts for highway maintenance which in turn may well reverse declining accident trends.


Asunto(s)
Accidentes de Tránsito/prevención & control , Accidentes de Tránsito/estadística & datos numéricos , Cambio Climático , Clima Frío/efectos adversos , Planificación Ambiental , Administración de la Seguridad , Estaciones del Año , Accidentes de Tránsito/tendencias , Estudios Transversales , Inglaterra , Predicción , Humanos , Modelos Estadísticos , Riesgo
19.
Neurochem Res ; 34(3): 556-65, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18716864

RESUMEN

Even though ammonia is considered to underlie nervous system symptoms of dysfunction during hyperammonemia, lactate, which increases as a metabolic consequence of high ammonia levels, might also be a contributing factor. The data presented here show that NH4Cl (5 mM) mediates astroglial cell swelling, and that treatment with NH4Cl or lactate (25 mM) causes rearrangements of actin filaments and reduces astroglial glutamate uptake capacity. Co-application with BaCl2, which blocks astroglial uptake of NH4+, prevents NH4Cl-mediated cell swelling and rearrangement of actin filaments, but does not reduce NH4Cl-induced glutamate uptake capacity inhibition. Neither NH4Cl nor lactate affected glutamate uptake or protein expression in microglial cultures, indicating that astroglial cells are more susceptible to the neurotoxic affects of ammonia. Our results suggest that ammonium underlies brain edema, but that lactate can contribute to some of the cellular dysfunctions associated with elevated cerebral levels of ammonia.


Asunto(s)
Amoníaco/metabolismo , Astrocitos/metabolismo , Hiperamonemia/metabolismo , Ácido Láctico/metabolismo , Citoesqueleto de Actina/ultraestructura , Cloruro de Amonio/farmacología , Animales , Astrocitos/patología , Calcio/metabolismo , Tamaño de la Célula , Células Cultivadas , Técnicas de Cocultivo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Hiperamonemia/patología , Ácido Láctico/farmacología , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley
20.
Arthritis Res Ther ; 10(2): 204, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18373887

RESUMEN

Progress into the understanding of immunopathology in rheumatoid arthritis is reviewed in the present article with regard to pro-inflammatory cytokine production, cell activation and recruitment, and osteoclastogenesis. Studies highlight the potential importance of T helper 17 cells and regulatory T cells in driving and suppressing inflammation in rheumatoid arthritis, respectively, and highlight other potential T-cell therapeutic targets. The genetic associations of the HLA shared epitope alleles with antibodies to citrullinated peptides in rheumatoid arthritis patients indicate that T cells are providing help to B cells to produce autoantibodies, and there is increasing evidence that these autoantibodies are pathogenic in rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/inmunología , Linfocitos T/inmunología , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Predisposición Genética a la Enfermedad , Humanos , Activación de Linfocitos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...