Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nat Chem Biol ; 20(1): 93-102, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37679459

RESUMEN

Molecular glue degraders are an effective therapeutic modality, but their design principles are not well understood. Recently, several unexpectedly diverse compounds were reported to deplete cyclin K by linking CDK12-cyclin K to the DDB1-CUL4-RBX1 E3 ligase. Here, to investigate how chemically dissimilar small molecules trigger cyclin K degradation, we evaluated 91 candidate degraders in structural, biophysical and cellular studies and reveal all compounds acquire glue activity via simultaneous CDK12 binding and engagement of DDB1 interfacial residues, in particular Arg928. While we identify multiple published kinase inhibitors as cryptic degraders, we also show that these glues do not require pronounced inhibitory properties for activity and that the relative degree of CDK12 inhibition versus cyclin K degradation is tuneable. We further demonstrate cyclin K degraders have transcriptional signatures distinct from CDK12 inhibitors, thereby offering unique therapeutic opportunities. The systematic structure-activity relationship analysis presented herein provides a conceptual framework for rational molecular glue design.


Asunto(s)
Ciclinas , Ubiquitina-Proteína Ligasas , Ciclinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteolisis , Relación Estructura-Actividad
2.
Nature ; 588(7836): 164-168, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33208943

RESUMEN

Effective and sustained inhibition of non-enzymatic oncogenic driver proteins is a major pharmacological challenge. The clinical success of thalidomide analogues demonstrates the therapeutic efficacy of drug-induced degradation of transcription factors and other cancer targets1-3, but a substantial subset of proteins are resistant to targeted degradation using existing approaches4,5. Here we report an alternative mechanism of targeted protein degradation, in which a small molecule induces the highly specific, reversible polymerization of a target protein, followed by its sequestration into cellular foci and subsequent degradation. BI-3802 is a small molecule that binds to the Broad-complex, Tramtrack and Bric-à-brac (BTB) domain of the oncogenic transcription factor B cell lymphoma 6 (BCL6) and leads to the proteasomal degradation of BCL66. We use cryo-electron microscopy to reveal how the solvent-exposed moiety of a BCL6-binding molecule contributes to a composite ligand-protein surface that engages BCL6 homodimers to form a supramolecular structure. Drug-induced formation of BCL6 filaments facilitates ubiquitination by the SIAH1 E3 ubiquitin ligase. Our findings demonstrate that a small molecule such as BI-3802 can induce polymerization coupled to highly specific protein degradation, which in the case of BCL6 leads to increased pharmacological activity compared to the effects induced by other BCL6 inhibitors. These findings open new avenues for the development of therapeutic agents and synthetic biology.


Asunto(s)
Polimerizacion/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/química , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Microscopía por Crioelectrón , Humanos , Técnicas In Vitro , Ligandos , Modelos Moleculares , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/ultraestructura , Solventes , Biología Sintética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
3.
Haematologica ; 104(11): 2215-2224, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975913

RESUMEN

Somatic mutations in acute myeloid leukemia are acquired sequentially and hierarchically. First, pre-leukemic mutations, such as t(8;21) that encodes AML1-ETO, are acquired within the hematopoietic stem cell (HSC) compartment, while signaling pathway mutations, including KRAS activating mutations, are late events acquired during transformation of leukemic progenitor cells and are rarely detectable in HSC. This raises the possibility that signaling pathway mutations are detrimental to clonal expansion of pre-leukemic HSC. To address this hypothesis, we used conditional genetics to introduce Aml1-ETO and K-RasG12D into murine HSC, either individually or in combination. In the absence of activated Ras, Aml1-ETO-expressing HSC conferred a competitive advantage. However, activated K-Ras had a marked detrimental effect on Aml1-ETO-expressing HSC, leading to loss of both phenotypic and functional HSC. Cell cycle analysis revealed a loss of quiescence in HSC co-expressing Aml1-ETO and K-RasG12D, accompanied by an enrichment in E2F and Myc target gene expression and depletion of HSC self-renewal-associated gene expression. These findings provide a mechanistic basis for the observed absence of KRAS signaling mutations in the pre-malignant HSC compartment.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Animales , Proliferación Celular/genética , Expresión Génica , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Transgénicos , Modelos Animales , Modelos Biológicos , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo
4.
Br J Haematol ; 163(2): 235-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23889083

RESUMEN

Whole exome sequencing was performed in a patient with myelodysplastic syndrome before and after progression to acute myeloid leukaemia. Mutations in several genes, including SETBP1, were identified following leukaemic transformation. Screening of 328 patients with myeloid disorders revealed SETBP1 mutations in 14 patients (4·3%), 7 of whom had -7/del(7q) and 3 had i(17)(q10), cytogenetic markers associated with shortened overall survival and increased risk of leukaemic evolution. SETBP1 mutations were frequently acquired at the time of leukaemic evolution, coinciding with increase of leukaemic blasts. These data suggest that SETBP1 mutations may play a role in MDS and chronic myelomonocytic leukaemia disease progression.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas Portadoras/genética , Aberraciones Cromosómicas , Mutación , Síndromes Mielodisplásicos/genética , Proteínas Nucleares/genética , Transformación Celular Neoplásica/genética , Deleción Cromosómica , Cromosomas Humanos Par 7 , Progresión de la Enfermedad , Exoma , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia Mielomonocítica Crónica/genética , Masculino , Recurrencia
5.
Exp Hematol ; 97: 6-13, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33600869

RESUMEN

Acute erythroid leukemia (AEL) is an acute leukemia characterized by erythroid lineage transformation. The World Health Organization (WHO) 2008 classification recognized two subtypes of AEL: bilineage erythroleukemia (erythroid/myeloid leukemia) and pure erythroid leukemia. The erythroleukemia subtype was removed in the updated 2016 WHO classification, with about half of cases reclassified as myelodysplastic syndrome (MDS) and half as acute myeloid leukemia (AML). Diagnosis and classification are currently based on morphology using standard blast cutoffs, without integration of underlying genomic and other molecular features. Key outstanding questions are therefore whether AEL can be accurately diagnosed based solely on morphology or whether genetic or other molecular criteria should be included in its classification, and whether considering AEL as an entity distinct from AML and MDS is clinically relevant. We discuss recent work on the molecular basis of AEL, including the identification of mutations causative of AEL and of transcriptional and epigenetic features that can be used to distinguish AEL from MDS and nonerythroid AML, and the prognostic value of these molecular features.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Animales , Epigénesis Genética , Células Eritroides/metabolismo , Células Eritroides/patología , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Eritroblástica Aguda/patología , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Mutación , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética
6.
Nat Commun ; 11(1): 4075, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796847

RESUMEN

Hematopoietic ageing involves declining erythropoiesis and lymphopoiesis, leading to frequent anaemia and decreased adaptive immunity. How intrinsic changes to the hematopoietic stem cells (HSCs), an altered microenvironment and systemic factors contribute to this process is not fully understood. Here we use bone marrow stromal cells as sensors of age-associated changes to the bone marrow microenvironment, and observe up-regulation of IL-6 and TGFß signalling-induced gene expression in aged bone marrow stroma. Inhibition of TGFß signalling leads to reversal of age-associated HSC platelet lineage bias, increased generation of lymphoid progenitors and rebalanced HSC lineage output in transplantation assays. In contrast, decreased erythropoiesis is not an intrinsic property of aged HSCs, but associated with decreased levels and functionality of erythroid progenitor populations, defects ameliorated by TGFß-receptor and IL-6 inhibition, respectively. These results show that both HSC-intrinsic and -extrinsic mechanisms are involved in age-associated hematopoietic decline, and identify therapeutic targets that promote their reversal.


Asunto(s)
Envejecimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-6/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Envejecimiento/genética , Animales , Médula Ósea , Ciclo Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Células Precursoras Eritroides , Eritropoyesis/genética , Eritropoyesis/fisiología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hematopoyesis , Interleucina-6/genética , Linfopoyesis/genética , Linfopoyesis/fisiología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Células Mieloides , Transducción de Señal , Nicho de Células Madre , Factor de Crecimiento Transformador beta1/genética
7.
Cancer Cell ; 37(5): 690-704.e8, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32330454

RESUMEN

Acute erythroid leukemia (AEL) commonly involves both myeloid and erythroid lineage transformation. However, the mutations that cause AEL and the cell(s) that sustain the bilineage leukemia phenotype remain unknown. We here show that combined biallelic Cebpa and Gata2 zinc finger-1 (ZnF1) mutations cooperatively induce bilineage AEL, and that the major leukemia-initiating cell (LIC) population has a neutrophil-monocyte progenitor (NMP) phenotype. In pre-leukemic NMPs Cebpa and Gata2 mutations synergize by increasing erythroid transcription factor (TF) expression and erythroid TF chromatin access, respectively, thereby installing ectopic erythroid potential. This erythroid-permissive chromatin conformation is retained in bilineage LICs. These results demonstrate that synergistic transcriptional and epigenetic reprogramming by leukemia-initiating mutations can generate neomorphic pre-leukemic progenitors, defining the lineage identity of the resulting leukemia.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Linaje de la Célula , Transformación Celular Neoplásica/patología , Células Precursoras Eritroides/patología , Factor de Transcripción GATA2/genética , Leucemia Eritroblástica Aguda/patología , Mutación , Neutrófilos/patología , Anciano , Alelos , Animales , Diferenciación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Células Precursoras Eritroides/metabolismo , Femenino , Factor de Transcripción GATA1/genética , Humanos , Leucemia Eritroblástica Aguda/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neutrófilos/metabolismo , Dedos de Zinc
8.
Nat Commun ; 10(1): 5455, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784538

RESUMEN

Acute Myeloid Leukemia (AML) develops due to the acquisition of mutations from multiple functional classes. Here, we demonstrate that activating mutations in the granulocyte colony stimulating factor receptor (CSF3R), cooperate with loss of function mutations in the transcription factor CEBPA to promote acute leukemia development. The interaction between these distinct classes of mutations occurs at the level of myeloid lineage enhancers where mutant CEBPA prevents activation of a subset of differentiation associated enhancers. To confirm this enhancer-dependent mechanism, we demonstrate that CEBPA mutations must occur as the initial event in AML initiation. This improved mechanistic understanding will facilitate therapeutic development targeting the intersection of oncogene cooperativity.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Leucemia Mieloide Aguda/genética , Receptores del Factor Estimulante de Colonias/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Humanos , Células K562 , Mutación con Pérdida de Función , Ratones , Mutación
10.
J Exp Med ; 214(7): 2005-2021, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28637883

RESUMEN

Although previous studies suggested that the expression of FMS-like tyrosine kinase 3 (Flt3) initiates downstream of mouse hematopoietic stem cells (HSCs), FLT3 internal tandem duplications (FLT3 ITDs) have recently been suggested to intrinsically suppress HSCs. Herein, single-cell interrogation found Flt3 mRNA expression to be absent in the large majority of phenotypic HSCs, with a strong negative correlation between Flt3 and HSC-associated gene expression. Flt3-ITD knock-in mice showed reduced numbers of phenotypic HSCs, with an even more severe loss of long-term repopulating HSCs, likely reflecting the presence of non-HSCs within the phenotypic HSC compartment. Competitive transplantation experiments established that Flt3-ITD compromises HSCs through an extrinsically mediated mechanism of disrupting HSC-supporting bone marrow stromal cells, with reduced numbers of endothelial and mesenchymal stromal cells showing increased inflammation-associated gene expression. Tumor necrosis factor (TNF), a cell-extrinsic potent negative regulator of HSCs, was overexpressed in bone marrow niche cells from FLT3-ITD mice, and anti-TNF treatment partially rescued the HSC phenotype. These findings, which establish that Flt3-ITD-driven myeloproliferation results in cell-extrinsic suppression of the normal HSC reservoir, are of relevance for several aspects of acute myeloid leukemia biology.


Asunto(s)
Proliferación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Mutación , Nicho de Células Madre/genética , Tirosina Quinasa 3 Similar a fms/genética , Animales , Antiinflamatorios no Esteroideos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Etanercept/farmacología , Perfilación de la Expresión Génica/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de la Célula Individual/métodos , Secuencias Repetidas en Tándem/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
PLoS Negl Trop Dis ; 7(3): e2152, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23556030

RESUMEN

BACKGROUND: Wolbachia inherited intracellular bacteria can manipulate the reproduction of their insect hosts through cytoplasmic incompatibility (CI), and certain strains have also been shown to inhibit the replication or dissemination of viruses. Wolbachia strains also vary in their relative fitness effects on their hosts and this is a particularly important consideration with respect to the potential of newly created transinfections for use in disease control. METHODOLOGY/PRINCIPAL FINDINGS: In Aedes albopictus mosquitoes transinfected with the wMel strain from Drosophila melanogaster, which we previously reported to be unable to transmit dengue in lab challenges, no significant detrimental effects were observed on egg hatch rate, fecundity, adult longevity or male mating competitiveness. All these parameters influence the population dynamics of Wolbachia, and the data presented are favourable with respect to the aim of taking wMel to high population frequency. Challenge with the chikungunya (CHIKV) virus, for which Ae. albopictus is an important vector, was conducted and the presence of wMel abolished CHIKV dissemination to the saliva. CONCLUSIONS/SIGNIFICANCE: Taken together, these data suggest that introducing wMel into natural Ae. albopictus populations using bidirectional CI could be an efficient strategy for preventing or reducing the transmission of arboviruses by this species.


Asunto(s)
Aedes/microbiología , Aedes/virología , Virus Chikungunya/crecimiento & desarrollo , Interacciones Microbianas , Wolbachia/crecimiento & desarrollo , Aedes/fisiología , Animales , Drosophila melanogaster/microbiología , Femenino , Masculino , Saliva/virología , Wolbachia/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA