Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
1.
PLoS Genet ; 18(4): e1010093, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35381001

RÉSUMÉ

Novel drug targets for sustained reduction in body mass index (BMI) are needed to curb the epidemic of obesity, which affects 650 million individuals worldwide and is a causal driver of cardiovascular and metabolic disease and mortality. Previous studies reported that the Arg95Ter nonsense variant of GPR151, an orphan G protein-coupled receptor, is associated with reduced BMI and reduced risk of Type 2 Diabetes (T2D). Here, we further investigate GPR151 with the Pakistan Genome Resource (PGR), which is one of the largest exome biobanks of human homozygous loss-of-function carriers (knockouts) in the world. Among PGR participants, we identify eleven GPR151 putative loss-of-function (plof) variants, three of which are present at homozygosity (Arg95Ter, Tyr99Ter, and Phe175LeufsTer7), with a cumulative allele frequency of 2.2%. We confirm these alleles in vitro as loss-of-function. We test if GPR151 plof is associated with BMI, T2D, or other metabolic traits and find that GPR151 deficiency in complete human knockouts is not associated with clinically significant differences in these traits. Relative to Gpr151+/+ mice, Gpr151-/- animals exhibit no difference in body weight on normal chow and higher body weight on a high-fat diet. Together, our findings indicate that GPR151 antagonism is not a compelling therapeutic approach to treatment of obesity.


Sujet(s)
Diabète de type 2 , Récepteurs couplés aux protéines G/métabolisme , Animaux , Indice de masse corporelle , Diabète de type 2/traitement médicamenteux , Diabète de type 2/génétique , Exome , Fréquence d'allèle , Humains , Souris , Obésité/génétique
2.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Article de Anglais | MEDLINE | ID: mdl-32551603

RÉSUMÉ

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Sujet(s)
Facteur XIa/antagonistes et inhibiteurs , Facteur XIa/génétique , Inhibiteurs du facteur Xa/administration et posologie , Inhibiteurs du facteur Xa/composition chimique , Administration par voie orale , Séquence d'acides aminés , Animaux , Biodisponibilité , Chiens , Évaluation préclinique de médicament/méthodes , Humains , Mâle , Souris , Souris de lignée C57BL , Rats , Rat Sprague-Dawley , Relation structure-activité
3.
Cancer Cell ; 22(3): 359-72, 2012 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-22975378

RÉSUMÉ

Diffuse large B cell lymphoma (DLBCL) is a clinically and biologically heterogeneous disease with a high proliferation rate. By integrating copy number data with transcriptional profiles and performing pathway analysis in primary DLBCLs, we identified a comprehensive set of copy number alterations (CNAs) that decreased p53 activity and perturbed cell cycle regulation. Primary tumors either had multiple complementary alterations of p53 and cell cycle components or largely lacked these lesions. DLBCLs with p53 and cell cycle pathway CNAs had decreased abundance of p53 target transcripts and increased expression of E2F target genes and the Ki67 proliferation marker. CNAs of the CDKN2A-TP53-RB-E2F axis provide a structural basis for increased proliferation in DLBCL, predict outcome with current therapy, and suggest targeted treatment approaches.


Sujet(s)
Cycle cellulaire/génétique , Variations de nombre de copies de segment d'ADN , Gènes p53 , Lymphome B diffus à grandes cellules/génétique , Protéine p53 suppresseur de tumeur/génétique , Prolifération cellulaire , Inhibiteur p16 de kinase cycline-dépendante/génétique , Facteurs de transcription E2F/biosynthèse , Facteurs de transcription E2F/génétique , Analyse de profil d'expression de gènes , Gènes p16 , Humains , Antigène KI-67/biosynthèse , Lymphome B diffus à grandes cellules/métabolisme , Lymphome B diffus à grandes cellules/anatomopathologie , Données de séquences moléculaires , Protéine p53 suppresseur de tumeur/métabolisme
4.
Clin Cancer Res ; 18(22): 6122-35, 2012 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-22966017

RÉSUMÉ

PURPOSE: B-cell receptor (BCR)-mediated signaling is important in the pathogenesis of a subset of diffuse large B-cell lymphomas (DLBCL) and the BCR-associated kinases SYK and BTK have recently emerged as potential therapeutic targets. We sought to identify a signature of activated BCR signaling in DLBCL to aid the identification of tumors that may be most likely to respond to BCR-pathway inhibition. EXPERIMENTAL DESIGN: We applied quantitative immunofluorescence (qIF) using antibodies to phosphorylated forms of proximal BCR signaling kinases LYN, SYK, and BTK and antibody to BCR-associated transcription factor FOXO1 on BCR-cross-linked formalin-fixed paraffin-embedded (FFPE) DLBCL cell lines as a model system and on two clinical cohorts of FFPE DLBCL specimens (n = 154). RESULTS: A robust signature of active BCR signaling was identified and validated in BCR-cross-linked DLBCL cell lines and in 71/154 (46%) of the primary DLBCL patient specimens. Further analysis of the primary biopsy samples revealed increased nuclear exclusion of FOXO1 among DLBCL with qIF evidence of active BCR signaling compared with those without (P = 0.004). Nuclear exclusion of FOXO1 was also detected in a subset of DLBCL without evidence of proximal BCR signaling suggesting that alternative mechanisms for PI3K/AKT activation may mediate FOXO1 subcellular localization in these cases. CONCLUSION: This study establishes the feasibility of detecting BCR activation in primary FFPE biopsy specimens of DLBCL. It lays a foundation for future dissection of signal transduction networks in DLBCL and provides a potential platform for evaluating individual tumors in patients receiving novel therapies targeting the BCR pathway.


Sujet(s)
Lymphome B diffus à grandes cellules/métabolisme , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Adulte , Agammaglobulinaemia tyrosine kinase , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire tumorale , Femelle , Technique d'immunofluorescence indirecte , Protéine O1 à motif en tête de fourche , Facteurs de transcription Forkhead/métabolisme , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Lymphome B diffus à grandes cellules/anatomopathologie , Mâle , Microscopie de fluorescence , Adulte d'âge moyen , Phosphoprotéines/métabolisme , Phosphorylation , Maturation post-traductionnelle des protéines , Transport des protéines , Protein-tyrosine kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Syk kinase , Analyse sur puce à tissus , src-Family kinases/métabolisme
5.
Cell ; 148(4): 739-51, 2012 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-22341446

RÉSUMÉ

B cells infected by Epstein-Barr virus (EBV), a transforming virus endemic in humans, are rapidly cleared by the immune system, but some cells harboring the virus persist for life. Under conditions of immunosuppression, EBV can spread from these cells and cause life-threatening pathologies. We have generated mice expressing the transforming EBV latent membrane protein 1 (LMP1), mimicking a constitutively active CD40 coreceptor, specifically in B cells. Like human EBV-infected cells, LMP1+ B cells were efficiently eliminated by T cells, and breaking immune surveillance resulted in rapid, fatal lymphoproliferation and lymphomagenesis. The lymphoma cells expressed ligands for a natural killer (NK) cell receptor, NKG2D, and could be targeted by an NKG2D-Fc fusion protein. These experiments indicate a central role for LMP1 in the surveillance and transformation of EBV-infected B cells in vivo, establish a preclinical model for B cell lymphomagenesis in immunosuppressed patients, and validate a new therapeutic approach.


Sujet(s)
Modèles animaux de maladie humaine , Herpèsvirus humain de type 4 , Surveillance immunologique , Lymphomes/immunologie , Lymphomes/thérapie , Protéines de la matrice virale/métabolisme , Animaux , Lymphocytes B/immunologie , Lymphocytes B/anatomopathologie , Humains , Immunothérapie , Lymphomes/anatomopathologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie , Protéines de la matrice virale/génétique
6.
Blood ; 118(10): 2857-67, 2011 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-21753187

RÉSUMÉ

MSI2 is highly expressed in human myeloid leukemia (AML) cell lines, and high expression of MSI2 mRNA is associated with decreased survival in AML, suggesting its use as a new prognostic marker. To test this, we measured MSI2 protein level by immunohistochemistry in 120 AML patients. Most cases (70%) showed some nuclear or cytoplasmic positivity, but the percentage of positive cells was low in most cases. Despite this, MSI2 protein expression was negatively associated with outcome, particularly for patients with good cytogenetic subgroup. For practical diagnostic purposes, the strongest significance of association was seen in cases with > 1% of cells showing strong MSI2 staining, these having a very poor outcome (P < .0001). Multivariate analysis with cytogenetic category, age, white cell count, and French-American-British subtype demonstrated that nuclear MSI2 levels were independently predictive of outcome (P = .0497). These results confirm the association of MSI2 expression with outcome in AML at the protein level and demonstrate the utility of MSI2 protein as a clinical prognostic biomarker. In addition, although positive at some level in most cases, its prognostic power derived from few positive cells, supporting its role in control of normal hematopoietic stem cell function and highlighting its role in disease progression.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Évolution de la maladie , Femelle , Études de suivi , Humains , Techniques immunoenzymatiques , Mâle , Adulte d'âge moyen , Pronostic , Taux de survie , Analyse sur puce à tissus , Jeune adulte
7.
Blood ; 117(16): 4315-22, 2011 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-21300977

RÉSUMÉ

Posttransplant lymphoproliferative disorders (PTLDs) are potentially fatal, EBV-driven B-cell malignancies that develop in immunocompromised solid organ or hematopoietic stem cell recipients. In PTLD, the expression of EBV proteins, including latent membrane protein 1 (LMP1) and LMP2A, viral immune evasion strategies, and impaired host immune surveillance foster the proliferation of EBV-transformed B cells. Current PTLD treatment strategies include reduction of immunosuppression, which increases the risk of graft rejection, anti-CD20 treatment, combination chemotherapy, and administration of EBV-specific cytotoxic T cells. In the present study, we report that EBV-transformed lymphoblastoid B-cell lines (LCLs) and primary PTLDs overexpress galectin-1 (Gal1), a carbohydrate-binding lectin that induces tolerogenic dendritic cells and triggers the selective apoptosis of CD4(+) Th1 and Th17 cells and cytotoxic T cells. In transcriptional reporter assays, LMP2A and LMP1 each increased Gal1-driven luciferase expression, and the combination of LMP2A and LMP1 was additive. In addition, small interfering RNA (siRNA)-mediated depletion of LMP2A decreased Gal1 protein abundance in EBV-transformed LCLs. Gal1 expression in LCLs was dependent on both activating protein 1 (AP-1) and PI3K. A newly developed neutralizing Gal1 mAb selectively inhibited Gal1-mediated apoptosis of EBV-specific CD8(+) T cells. Given the tolerogenic and immunosuppressive function of Gal1, antibody-mediated Gal1 neutralization may represent a novel immunotherapeutic strategy for PTLD and other Gal1-expressing tumors.


Sujet(s)
Transformation cellulaire virale , Galectine 1/génétique , Herpèsvirus humain de type 4/physiologie , Syndromes lymphoprolifératifs/virologie , Animaux , Anticorps monoclonaux/immunologie , Apoptose , Lignée de cellules transformées , Lignée cellulaire tumorale , Galectine 1/immunologie , Régulation de l'expression des gènes tumoraux , Humains , Syndromes lymphoprolifératifs/génétique , Souris , Lymphocytes T cytotoxiques/cytologie , Facteur de transcription AP-1/métabolisme , Régulation positive , Protéines de la matrice virale/métabolisme
8.
Blood ; 117(9): 2567-76, 2011 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-21068437

RÉSUMÉ

Haploinsufficiency for ribosomal protein genes has been implicated in the pathophysiology of Diamond-Blackfan anemia (DBA) and the 5q-syndrome, a subtype of myelodysplastic syndrome. The p53 pathway is activated by ribosome dysfunction, but the molecular basis for selective impairment of the erythroid lineage in disorders of ribosome function has not been determined. We found that p53 accumulates selectively in the erythroid lineage in primary human hematopoietic progenitor cells after expression of shRNAs targeting RPS14, the ribosomal protein gene deleted in the 5q-syndrome, or RPS19, the most commonly mutated gene in DBA. Induction of p53 led to lineage-specific accumulation of p21 and consequent cell cycle arrest in erythroid progenitor cells. Pharmacologic inhibition of p53 rescued the erythroid defect, whereas nutlin-3, a compound that activates p53 through inhibition of HDM2, selectively impaired erythropoiesis. In bone marrow biopsies from patients with DBA or del(5q) myelodysplastic syndrome, we found an accumulation of nuclear p53 staining in erythroid progenitor cells that was not present in control samples. Our findings indicate that the erythroid lineage has a low threshold for the induction of p53, providing a basis for the failure of erythropoiesis in the 5q-syndrome, DBA, and perhaps other bone marrow failure syndromes.


Sujet(s)
Précurseurs érythroïdes/métabolisme , Haploinsuffisance/génétique , Protéines ribosomiques/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Anémie de Blackfan-Diamond/génétique , Anémie de Blackfan-Diamond/anatomopathologie , Anémie macrocytaire/génétique , Anémie macrocytaire/anatomopathologie , Animaux , Benzothiazoles/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignage cellulaire/effets des médicaments et des substances chimiques , Nucléole/effets des médicaments et des substances chimiques , Nucléole/métabolisme , Délétion de segment de chromosome , Chromosomes humains de la paire 5/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Précurseurs érythroïdes/effets des médicaments et des substances chimiques , Précurseurs érythroïdes/anatomopathologie , Hématopoïèse/effets des médicaments et des substances chimiques , Humains , Imidazoles/métabolisme , Souris , Souris de lignée BALB C , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/anatomopathologie , Pipérazines/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-mdm2/métabolisme , Petit ARN interférent/métabolisme , Protéines ribosomiques/déficit , Protéines ribosomiques/métabolisme , Toluène/analogues et dérivés , Toluène/pharmacologie
9.
Blood ; 116(17): 3268-77, 2010 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-20628145

RÉSUMÉ

Classical Hodgkin lymphoma (cHL) and mediastinal large B-cell lymphoma (MLBCL) are lymphoid malignancies with certain shared clinical, histologic, and molecular features. Primary cHLs and MLBCLs include variable numbers of malignant cells within an inflammatory infiltrate, suggesting that these tumors escape immune surveillance. Herein, we integrate high-resolution copy number data with transcriptional profiles and identify the immunoregulatory genes, PD-L1 and PD-L2, as key targets at the 9p24.1 amplification peak in HL and MLBCL cell lines. We extend these findings to laser-capture microdissected primary Hodgkin Reed-Sternberg cells and primary MLBCLs and find that programmed cell death-1 (PD-1) ligand/9p24.1 amplification is restricted to nodular sclerosing HL, the cHL subtype most closely related to MLBCL. Using quantitative immunohistochemical methods, we document the association between 9p24.1 copy number and PD-1 ligand expression in primary tumors. In cHL and MLBCL, the extended 9p24.1 amplification region also included the Janus kinase 2 (JAK2) locus. Of note, JAK2 amplification increased protein expression and activity, specifically induced PD-1 ligand transcription and enhanced sensitivity to JAK2 inhibition. Therefore, 9p24.1 amplification is a disease-specific structural alteration that increases both the gene dosage of PD-1 ligands and their induction by JAK2, defining the PD-1 pathway and JAK2 as complementary rational therapeutic targets.


Sujet(s)
Antigènes CD/génétique , Chromosomes humains de la paire 9/génétique , Dosage génique , Maladie de Hodgkin/génétique , Kinase Janus-2/génétique , Lymphome B diffus à grandes cellules/génétique , Antigène CD274 , Lignée cellulaire tumorale , Prolifération cellulaire , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux , Humains , Protéines et peptides de signalisation intercellulaire/génétique , Kinase Janus-2/antagonistes et inhibiteurs , Kinase Janus-2/métabolisme , Ligand-2 de la protéine-1 de mort cellulaire programmée , Cellules cancéreuses en culture
10.
Clin Cancer Res ; 14(11): 3338-44, 2008 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-18519761

RÉSUMÉ

PURPOSE: Galectin-1 (Gal1) is an immunomodulatory glycan-binding protein regulated by an AP1-dependent enhancer in Hodgkin Reed-Sternberg cells. We recently found that Reed-Sternberg cell Gal1 promotes the immunosuppressive T-helper 2/T-regulatory cell-skewed microenvironment in classical Hodgkin lymphoma (cHL). We sought to investigate whether the coordinate expression of activated AP1 pathway components and Gal1 serves as a diagnostic signature of cHL. In addition, because there are common signaling and survival pathways in cHL and additional non-Hodgkin lymphomas, we also evaluated whether the AP1/Gal1 signature is shared by other molecularly or morphologically related lymphomas. EXPERIMENTAL DESIGN: We evaluated 225 cases of primary cHL and non-Hodgkin lymphoma for evidence of a functional AP1/Gal1 signature by immunohistochemical techniques. RESULTS: Gal1 is selectively expressed by malignant Reed-Sternberg cells in >90% of primary cHLs, and Gal1 expression is concordant with the activated AP1 component, c-Jun. In contrast, diffuse large B-cell lymphoma, primary mediastinal large B-cell lymphoma, and another Hodgkin-related entity, nodular lymphocyte-predominant Hodgkin lymphoma, do not express Gal1. However, anaplastic large cell lymphoma (ALCL), consistently expresses both Gal1 and its transcriptional regulator, c-Jun. The presence of activated c-Jun, indicative of functional AP1 activity, was confirmed by phospho-c-Jun immunostaining in cHL and ALCL. CONCLUSIONS: These findings establish a functional AP1 signature that includes Gal1 expression in cHL and ALCL and suggests a common mechanism for tumor immunotolerance in these diseases. In addition, the combination of Gal1 and c-Jun serve as diagnostic biomarkers that delineate cHL and ALCL from other lymphomas with shared morphologic and/or molecular features.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Galectine 1/biosynthèse , Maladie de Hodgkin/métabolisme , Lymphome à grandes cellules anaplasiques/métabolisme , Syndromes lymphoprolifératifs/métabolisme , Facteur de transcription AP-1/métabolisme , Diagnostic différentiel , Expression des gènes , Maladie de Hodgkin/diagnostic , Maladie de Hodgkin/génétique , Humains , Immunohistochimie , Lymphome à grandes cellules anaplasiques/diagnostic , Lymphome à grandes cellules anaplasiques/génétique , Syndromes lymphoprolifératifs/diagnostic , Syndromes lymphoprolifératifs/génétique , Protéines proto-oncogènes c-jun/métabolisme , Cellules de Reed-Sternberg/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE