Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Discov ; 13(8): 1922-1947, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37191437

RESUMEN

Leukemia stem cells (LSC) possess distinct self-renewal and arrested differentiation properties that are responsible for disease emergence, therapy failure, and recurrence in acute myeloid leukemia (AML). Despite AML displaying extensive biological and clinical heterogeneity, LSC with high interleukin-3 receptor (IL3R) levels are a constant yet puzzling feature, as this receptor lacks tyrosine kinase activity. Here, we show that the heterodimeric IL3Rα/ßc receptor assembles into hexamers and dodecamers through a unique interface in the 3D structure, where high IL3Rα/ßc ratios bias hexamer formation. Importantly, receptor stoichiometry is clinically relevant as it varies across the individual cells in the AML hierarchy, in which high IL3Rα/ßc ratios in LSCs drive hexamer-mediated stemness programs and poor patient survival, while low ratios mediate differentiation. Our study establishes a new paradigm in which alternative cytokine receptor stoichiometries differentially regulate cell fate, a signaling mechanism that may be generalizable to other transformed cellular hierarchies and of potential therapeutic significance. SIGNIFICANCE: Stemness is a hallmark of many cancers and is largely responsible for disease emergence, progression, and relapse. Our finding that clinically significant stemness programs in AML are directly regulated by different stoichiometries of cytokine receptors represents a hitherto unexplained mechanism underlying cell-fate decisions in cancer stem cell hierarchies. This article is highlighted in the In This Issue feature, p. 1749.


Asunto(s)
Leucemia Mieloide Aguda , Receptores de Citocinas , Humanos , Receptores de Citocinas/uso terapéutico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Fosforilación , Transducción de Señal , Proliferación Celular , Células Madre Neoplásicas
2.
EMBO Mol Med ; 15(1): e16236, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36468184

RESUMEN

C-reactive protein (CRP) is an early-stage acute phase protein and highly upregulated in response to inflammatory reactions. We recently identified a novel mechanism that leads to a conformational change from the native, functionally relatively inert, pentameric CRP (pCRP) structure to a pentameric CRP intermediate (pCRP*) and ultimately to the monomeric CRP (mCRP) form, both exhibiting highly pro-inflammatory effects. This transition in the inflammatory profile of CRP is mediated by binding of pCRP to activated/damaged cell membranes via exposed phosphocholine lipid head groups. We designed a tool compound as a low molecular weight CRP inhibitor using the structure of phosphocholine as a template. X-ray crystallography revealed specific binding to the phosphocholine binding pockets of pCRP. We provide in vitro and in vivo proof-of-concept data demonstrating that the low molecular weight tool compound inhibits CRP-driven exacerbation of local inflammatory responses, while potentially preserving pathogen-defense functions of CRP. The inhibition of the conformational change generating pro-inflammatory CRP isoforms via phosphocholine-mimicking compounds represents a promising, potentially broadly applicable anti-inflammatory therapy.


Asunto(s)
Proteína C-Reactiva , Fosforilcolina , Humanos , Fosforilcolina/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Membrana Celular/metabolismo , Antiinflamatorios
3.
Semin Immunol ; 54: 101513, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-34836771

RESUMEN

Our understanding of the biological role of the ßc family of cytokines has evolved enormously since their initial identification as bone marrow colony stimulating factors in the 1960's. It has become abundantly clear over the intervening decades that this family of cytokines has truly astonishing pleiotropic capacity, capable of regulating not only hematopoiesis but also many other normal and pathological processes such as development, inflammation, allergy and cancer. As noted in the current pandemic, ßc cytokines contribute to the cytokine storm seen in acutely ill COVID-19 patients. Ongoing studies to discover how these cytokines activate their receptor are revealing insights into the fundamental mechanisms that give rise to cytokine pleiotropy and are providing tantalizing glimpses of how discrete signaling pathways may be dissected for activation with novel ligands for therapeutic benefit.


Asunto(s)
COVID-19 , Objetivos , Humanos , SARS-CoV-2
4.
Sci Rep ; 9(1): 7851, 2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31110193

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

5.
Sci Rep ; 8(1): 12457, 2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-30127368

RESUMEN

A direct interaction between the erythropoietin (EPOR) and the beta-common (ßc) receptors to form an Innate Repair Receptor (IRR) is controversial. On one hand, studies have shown a functional link between EPOR and ßc receptor in tissue protection while others have shown no involvement of the ßc receptor in tissue repair. To date there is no biophysical evidence to confirm a direct association of the two receptors either in vitro or in vivo. We investigated the existence of an interaction between the extracellular regions of EPOR and the ßc receptor in silico and in vitro (either in the presence or absence of EPO or EPO-derived peptide ARA290). Although a possible interaction between EPOR and ßc was suggested by our computational and genomic studies, our in vitro biophysical analysis demonstrates that the extracellular regions of the two receptors do not specifically associate. We also explored the involvement of the ßc receptor gene (Csf2rb) under anaemic stress conditions and found no requirement for the ßc receptor in mice. In light of these studies, we conclude that the extracellular regions of the EPOR and the ßc receptor do not directly interact and that the IRR is not involved in anaemic stress.

6.
MAbs ; 10(7): 1018-1029, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29969365

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that can stimulate a variety of cells, but its overexpression leads to excessive production and activation of granulocytes and macrophages with many pathogenic effects. This cytokine is a therapeutic target in inflammatory diseases, and several anti-GM-CSF antibodies have advanced to Phase 2 clinical trials in patients with such diseases, e.g., rheumatoid arthritis. GM-CSF is also an essential factor in preventing pulmonary alveolar proteinosis (PAP), a disease associated with GM-CSF malfunction arising most typically through the presence of GM-CSF neutralizing auto-antibodies. Understanding the mechanism of action for neutralizing antibodies that target GM-CSF is important for improving their specificity and affinity as therapeutics and, conversely, in devising strategies to reduce the effects of GM-CSF auto-antibodies in PAP. We have solved the crystal structures of human GM-CSF bound to antigen-binding fragments of two neutralizing antibodies, the human auto-antibody F1 and the mouse monoclonal antibody 4D4. Coordinates and structure factors of the crystal structures of the GM-CSF:F1 Fab and the GM-CSF:4D4 Fab complexes have been deposited in the RCSB Protein Data Bank under the accession numbers 6BFQ and 6BFS, respectively. The structures show that these antibodies bind to mutually exclusive epitopes on GM-CSF; however, both prevent the cytokine from interacting with its alpha receptor subunit and hence prevent receptor activation. Importantly, identification of the F1 epitope together with functional analyses highlighted modifications to GM-CSF that would abolish auto-antibody recognition whilst retaining GM-CSF function. These results provide a framework for developing novel GM-CSF molecules for PAP treatment and for optimizing current anti-GM-CSF antibodies for use in treating inflammatory disorders.


Asunto(s)
Anticuerpos Neutralizantes/química , Complejo Antígeno-Anticuerpo/química , Artritis Reumatoide/terapia , Autoanticuerpos/química , Epítopos/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia/métodos , Anticuerpos Neutralizantes/metabolismo , Artritis Reumatoide/inmunología , Autoanticuerpos/metabolismo , Autoanticuerpos/farmacología , Cristalografía por Rayos X , Citocinas/metabolismo , Epítopos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Estructura Molecular , Unión Proteica , Conformación Proteica
7.
Nat Commun ; 9(1): 386, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29374162

RESUMEN

The interleukin-3 (IL-3) receptor is a cell-surface heterodimer that links the haemopoietic, vascular and immune systems and is overexpressed in acute and chronic myeloid leukaemia progenitor cells. It belongs to the type I cytokine receptor family in which the α-subunits consist of two fibronectin III-like domains that bind cytokine, and a third, evolutionarily unrelated and topologically conserved, N-terminal domain (NTD) with unknown function. Here we show by crystallography that, while the NTD of IL3Rα is highly mobile in the presence of IL-3, it becomes surprisingly rigid in the presence of IL-3 K116W. Mutagenesis, biochemical and functional studies show that the NTD of IL3Rα regulates IL-3 binding and signalling and reveal an unexpected role in preventing spontaneous receptor dimerisation. Our work identifies a dual role for the NTD in this cytokine receptor family, protecting against inappropriate signalling and dynamically regulating cytokine receptor binding and function.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-3/química , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Dominios Proteicos , Transducción de Señal , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Cristalografía por Rayos X , Células HEK293 , Humanos , Interleucina-3/química , Interleucina-3/genética , Interleucina-3/metabolismo , Subunidad alfa del Receptor de Interleucina-3/genética , Simulación de Dinámica Molecular , Mutación , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA