Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 5(19): 3794-3798, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34471932

RESUMEN

T cell-engaging immunotherapies exert unprecedented single-agent activity in multiple myeloma (MM), thereby putting a yet unexplored selective pressure on the clonal architecture. In this study, we report on homozygous BCMA (TNFRSF17) gene deletion after BCMA-targeting T cell-redirecting bispecific antibody therapy in a heavily pretreated MM patient. Loss of BCMA protein expression persisted over subsequent relapses, with no response to treatment with anti-BCMA antibody drug conjugate. In light of the multiple alternative targets that are emerging in addition to BCMA, we extended our analyses to delineate a more complete picture of genetic alterations that may have an impact on immunotherapy targets in MM. We performed whole-genome sequencing and RNA sequencing in 100 MM patients (50 were newly diagnosed; 50 were relapsed/refractory) and identified a significant proportion of patients with aberrations in genes encoding immunotherapy targets; GPRC5D ranked first with 15% heterozygous deletions, followed by CD38 (10%), SDC1 (5%), and TNFRSF17 (4%). Notably, these heterozygous deletions did not lower the expression levels of respective genes, but they may represent a first hit that drives the acquisition of homozygous deletions and subsequent antigen-loss relapse upon targeted immunotherapy. In summary, we show preexisting vulnerability in genes encoding immunotargets before and homozygous deletions after T cell-engaging immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos , Mieloma Múltiple , Antígeno de Maduración de Linfocitos B , Humanos , Inmunoterapia , Mieloma Múltiple/genética , Mieloma Múltiple/terapia , Linfocitos T
3.
PLoS One ; 9(4): e93852, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24718418

RESUMEN

The implementation of expanded newborn screening programs reduced mortality and morbidity in medium-chain acyl-CoA dehydrogenase deficiency (MCADD) caused by mutations in the ACADM gene. However, the disease is still potentially fatal. Missense induced MCADD is a protein misfolding disease with a molecular loss-of-function phenotype. Here we established a comprehensive experimental setup to analyze the structural consequences of eight ACADM missense mutations (p.Ala52Val, p.Tyr67His, p.Tyr158His, p.Arg206Cys, p.Asp266Gly, p.Lys329Glu, p.Arg334Lys, p.Arg413Ser) identified after newborn screening and linked the corresponding protein misfolding phenotype to the site of side-chain replacement with respect to the domain. With fever being the crucial risk factor for metabolic decompensation of patients with MCADD, special emphasis was put on the analysis of structural and functional derangements related to thermal stress. Based on protein conformation, thermal stability and kinetic stability, the molecular phenotype in MCADD depends on the structural region that is affected by missense-induced conformational changes with the central ß-domain being particularly prone to structural derangement and destabilization. Since systematic classification of conformational derangements induced by ACADM mutations may be a helpful tool in assessing the clinical risk of patients, we scored the misfolding phenotype of the variants in comparison to p.Lys329Glu (K304E), the classical severe mutation, and p.Tyr67His (Y42H), discussed to be mild. Experiments assessing the impact of thermal stress revealed that mutations in the ACADM gene lower the temperature threshold at which MCAD loss-of-function occurs. Consequently, increased temperature as it occurs during intercurrent infections, significantly increases the risk of further conformational derangement and loss of function of the MCAD enzyme explaining the life-threatening clinical courses observed during fever episodes. Early and aggressive antipyretic treatment thus may be life-saving in patients suffering from MCADD.


Asunto(s)
Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Pliegue de Proteína , Temperatura , Animales , Células COS , Chlorocebus aethiops , Dicroismo Circular , Activación Enzimática , Flavina-Adenina Dinucleótido/metabolismo , Fluorescencia , Calor , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Mutación Missense/genética , Fenotipo , Agregado de Proteínas , Desnaturalización Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
4.
J Biol Chem ; 285(40): 30686-97, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20667834

RESUMEN

Protein misfolding with loss-of-function of the enzyme phenylalanine hydroxylase (PAH) is the molecular basis of phenylketonuria in many individuals carrying missense mutations in the PAH gene. PAH is complexly regulated by its substrate L-Phenylalanine and its natural cofactor 6R-L-erythro-5,6,7,8-tetrahydrobiopterin (BH(4)). Sapropterin dihydrochloride, the synthetic form of BH(4), was recently approved as the first pharmacological chaperone to correct the loss-of-function phenotype. However, current knowledge about enzyme function and regulation in the therapeutic setting is scarce. This illustrates the need for comprehensive analyses of steady state kinetics and allostery beyond single residual enzyme activity determinations to retrace the structural impact of missense mutations on the phenylalanine hydroxylating system. Current standard PAH activity assays are either indirect (NADH) or discontinuous due to substrate and product separation before detection. We developed an automated fluorescence-based continuous real-time PAH activity assay that proved to be faster and more efficient but as precise and accurate as standard methods. Wild-type PAH kinetic analyses using the new assay revealed cooperativity of activated PAH toward BH(4), a previously unknown finding. Analyses of structurally preactivated variants substantiated BH(4)-dependent cooperativity of the activated enzyme that does not rely on the presence of l-Phenylalanine but is determined by activating conformational rearrangements. These findings may have implications for an individualized therapy, as they support the hypothesis that the patient's metabolic state has a more significant effect on the interplay of the drug and the conformation and function of the target protein than currently appreciated.


Asunto(s)
Biopterinas/análogos & derivados , Coenzimas/química , Fenilalanina Hidroxilasa/química , Fenilalanina/química , Regulación Alostérica/genética , Biopterinas/química , Biopterinas/metabolismo , Biopterinas/uso terapéutico , Coenzimas/metabolismo , Coenzimas/uso terapéutico , Activación Enzimática/genética , Fluorescencia , Humanos , Cinética , Mutación Missense , Fenilalanina/genética , Fenilalanina/metabolismo , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/enzimología , Fenilcetonurias/genética
5.
Hum Mol Genet ; 18(9): 1612-23, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19224950

RESUMEN

Newborn screening (NBS) for medium-chain acyl-CoA dehydrogenase deficiency (MCADD) revealed a higher birth prevalence and genotypic variability than previously estimated, including numerous novel missense mutations in the ACADM gene. On average, these mutations are associated with milder biochemical phenotypes raising the question about their pathogenic relevance. In this study, we analyzed the impact of 10 ACADM mutations identified in NBS (A27V, Y42H, Y133H, R181C, R223G, D241G, K304E, R309K, I331T and R388S) on conformation, stability and enzyme kinetics of the corresponding proteins. Partial to total rescue of aggregation by co-overexpression of GroESL indicated protein misfolding. This was confirmed by accelerated thermal unfolding in all variants, as well as decreased proteolytic stability and accelerated thermal inactivation in most variants. Catalytic function varied from high residual activity to markedly decreased activity or substrate affinity. Mutations mapping to the beta-domain of the protein predisposed to severe destabilization. In silico structural analyses of the affected amino acid residues revealed involvement in functionally relevant networks. Taken together, our results substantiate the hypothesis of protein misfolding with loss-of-function being the common molecular basis in MCADD. Moreover, considerable structural alterations in all analyzed variants do not support the view that novel mutations found in NBS bear a lower risk of metabolic decompensation than that associated with mutations detected in clinically ascertained patients. Finally, the detailed insight into how ACADM missense mutations induce loss of MCAD function may provide guidance for risk assessment and counseling of patients, and in future may assist delineation of novel pharmacological strategies.


Asunto(s)
Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo Lipídico/enzimología , Tamizaje Neonatal , Pliegue de Proteína , Acil-CoA Deshidrogenasa/genética , Sustitución de Aminoácidos , Estabilidad de Enzimas , Femenino , Humanos , Recién Nacido , Cinética , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Conformación Molecular , Datos de Secuencia Molecular , Mutación Missense
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...