Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
1.
Int J Radiat Oncol Biol Phys ; 114(4): 792-802, 2022 11 15.
Article de Anglais | MEDLINE | ID: mdl-35896145

RÉSUMÉ

PURPOSE: The oligometastatic state is observed in patients across many malignancies, with increased recognition regarding improved outcomes after local therapies. However, there is limited data specifically regarding pancreatic ductal adenocarcinoma. We hypothesized that an oligometastatic pancreatic ductal adenocarcinoma (OPanc) phenotype would benefit from stereotactic ablative radiation therapy (SABR) to all active metastatic sites. Here, we report our institutional experience of SABR-treated OPanc to evaluate the feasibility of the approach. METHODS AND MATERIALS: A retrospective review of patients with synchronous or metachronous OPanc (1 to 5 metastases) who received SABR to all active metastatic sites was performed. We identified a comparable group of patients with similar metastatic burden, range of CA19-9 levels, and no progression for at least 5 months who did not receive SABR. We compared overall survival as the primary outcome, and polyprogression-free survival and time off chemotherapy as the secondary exploratory assessments. A third group presenting with stage IV pancreatic ductal adenocarcinoma and more than 5 distant lesions (polymetastatic) was identified to help define expected outcomes after polyprogression. RESULTS: Our study included 20 patients with OPanc receiving SABR and 21 who did not. SABR was delivered to 38 metastatic tumors. Out of the 20 SABR-treated OPanc patients, 17 (85%) had 6 or more months of time off chemotherapy, compared with 7 patients (33.3%) among the chemotherapy-treated group. Median polyprogression-free survival was 40 and 14 months (hazard ratio = 0.2; 95% confidence interval, 0.07-0.54; P = .0009), and overall survival was 42 and 18 months (hazard ratio = 0.21; 95% confidence interval, 0.08-0.53; P = .0003), for SABR- and chemotherapy-treated cohorts, respectively. CONCLUSIONS: Management of OPanc with SABR as local regional therapy could improve outcomes in a selected population and warrants prospective evaluation.


Sujet(s)
Adénocarcinome , Tumeurs du pancréas , Radiochirurgie , Antigène CA 19-9 , Humains , Tumeurs du pancréas/radiothérapie , Radiochirurgie/méthodes , Tumeurs du pancréas
2.
Cancer Cell ; 40(6): 656-673.e7, 2022 06 13.
Article de Anglais | MEDLINE | ID: mdl-35523176

RÉSUMÉ

Recent studies have identified a unique cancer-associated fibroblast (CAF) population termed antigen-presenting CAFs (apCAFs), characterized by the expression of major histocompatibility complex class II molecules, suggesting a function in regulating tumor immunity. Here, by integrating multiple single-cell RNA-sequencing studies and performing robust lineage-tracing assays, we find that apCAFs are derived from mesothelial cells. During pancreatic cancer progression, mesothelial cells form apCAFs by downregulating mesothelial features and gaining fibroblastic features, a process induced by interleukin-1 and transforming growth factor ß. apCAFs directly ligate and induce naive CD4+ T cells into regulatory T cells (Tregs) in an antigen-specific manner. Moreover, treatment with an antibody targeting the mesothelial cell marker mesothelin can effectively inhibit mesothelial cell to apCAF transition and Treg formation induced by apCAFs. Taken together, our study elucidates how mesothelial cells may contribute to immune evasion in pancreatic cancer and provides insight on strategies to enhance cancer immune therapy.


Sujet(s)
Fibroblastes associés au cancer , Tumeurs du pancréas , Fibroblastes associés au cancer/métabolisme , Fibroblastes , Humains , Tumeurs du pancréas/anatomopathologie , Lymphocytes T régulateurs , Facteur de croissance transformant bêta/métabolisme , Tumeurs du pancréas
3.
Cancer Discov ; 12(2): 542-561, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34551968

RÉSUMÉ

The degree of metastatic disease varies widely among patients with cancer and affects clinical outcomes. However, the biological and functional differences that drive the extent of metastasis are poorly understood. We analyzed primary tumors and paired metastases using a multifluorescent lineage-labeled mouse model of pancreatic ductal adenocarcinoma (PDAC)-a tumor type in which most patients present with metastases. Genomic and transcriptomic analysis revealed an association between metastatic burden and gene amplification or transcriptional upregulation of MYC and its downstream targets. Functional experiments showed that MYC promotes metastasis by recruiting tumor-associated macrophages, leading to greater bloodstream intravasation. Consistent with these findings, metastatic progression in human PDAC was associated with activation of MYC signaling pathways and enrichment for MYC amplifications specifically in metastatic patients. Collectively, these results implicate MYC activity as a major determinant of metastatic burden in advanced PDAC. SIGNIFICANCE: Here, we investigate metastatic variation seen clinically in patients with PDAC and murine PDAC tumors and identify MYC as a major driver of this heterogeneity.This article is highlighted in the In This Issue feature, p. 275.


Sujet(s)
Adénocarcinome/génétique , Carcinome du canal pancréatique/génétique , Régulation de l'expression des gènes tumoraux , Gènes myc , Métastase tumorale , Tumeurs du pancréas/génétique , Adénocarcinome/secondaire , Animaux , Carcinome du canal pancréatique/secondaire , Modèles animaux de maladie humaine , Humains , Souris , Tumeurs du pancréas/anatomopathologie
4.
EMBO Rep ; 22(9): e51872, 2021 09 06.
Article de Anglais | MEDLINE | ID: mdl-34324787

RÉSUMÉ

Epithelial plasticity, or epithelial-to-mesenchymal transition (EMT), is a well-recognized form of cellular plasticity, which endows tumor cells with invasive properties and alters their sensitivity to various agents, thus representing a major challenge to cancer therapy. It is increasingly accepted that carcinoma cells exist along a continuum of hybrid epithelial-mesenchymal (E-M) states and that cells exhibiting such partial EMT (P-EMT) states have greater metastatic competence than those characterized by either extreme (E or M). We described recently a P-EMT program operating in vivo by which carcinoma cells lose their epithelial state through post-translational programs. Here, we investigate the underlying mechanisms and report that prolonged calcium signaling induces a P-EMT characterized by the internalization of membrane-associated E-cadherin (ECAD) and other epithelial proteins as well as an increase in cellular migration and invasion. Signaling through Gαq-associated G-protein-coupled receptors (GPCRs) recapitulates these effects, which operate through the downstream activation of calmodulin-Camk2b signaling. These results implicate calcium signaling as a trigger for the acquisition of hybrid/partial epithelial-mesenchymal states in carcinoma cells.


Sujet(s)
Signalisation calcique , Transition épithélio-mésenchymateuse , Cadhérines/génétique , Cadhérines/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire , Plasticité cellulaire
5.
Nat Cancer ; 2(12): 1338-1356, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-35121902

RÉSUMÉ

Despite efforts in understanding its underlying mechanisms, the etiology of chromosomal instability (CIN) remains unclear for many tumor types. Here, we identify CIN initiation as a previously undescribed function for APOBEC3A (A3A), a cytidine deaminase upregulated across cancer types. Using genetic mouse models of pancreatic ductal adenocarcinoma (PDA) and genomics analyses in human tumor cells we show that A3A-induced CIN leads to aggressive tumors characterized by enhanced early dissemination and metastasis in a STING-dependent manner and independently of the canonical deaminase functions of A3A. We show that A3A upregulation recapitulates numerous copy number alterations commonly observed in patients with PDA, including co-deletions in DNA repair pathway genes, which in turn render these tumors susceptible to poly (ADP-ribose) polymerase inhibition. Overall, our results demonstrate that A3A plays an unexpected role in PDA as a specific driver of CIN, with significant effects on disease progression and treatment.


Sujet(s)
Cytidine deaminase , Tumeurs du pancréas , Animaux , Instabilité des chromosomes/génétique , Cytidine deaminase/génétique , Humains , Souris , Tumeurs du pancréas/génétique , Protéines/génétique , Tumeurs du pancréas
6.
Cancers (Basel) ; 12(9)2020 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-32957515

RÉSUMÉ

Cancer-associated fibroblasts (CAFs) are indispensable architects of the tumor microenvironment. They perform the essential functions of extracellular matrix deposition, stromal remodeling, tumor vasculature modulation, modification of tumor metabolism, and participation in crosstalk between cancer and immune cells. In this review, we discuss our current understanding of the principal differences between normal fibroblasts and CAFs, the origin of CAFs, their functions, and ultimately, highlight the intimate connection of CAFs to virtually all of the hallmarks of cancer. We address the remarkable degree of functional diversity and phenotypic plasticity displayed by CAFs and strive to stratify CAF biology among different tumor types into practical functional groups. Finally, we summarize the status of recent and ongoing trials of CAF-directed therapies and contend that the paucity of trials resulting in Food and Drug Administration (FDA) approvals thus far is a consequence of the failure to identify targets exclusive of pro-tumorigenic CAF phenotypes that are mechanistically linked to specific CAF functions. We believe that the development of a unified CAF nomenclature, the standardization of functional assays to assess the loss-of-function of CAF properties, and the establishment of rigorous definitions of CAF subpopulations and their mechanistic functions in cancer progression will be crucial to fully realize the promise of CAF-targeted therapies.

7.
PLoS One ; 13(8): e0201751, 2018.
Article de Anglais | MEDLINE | ID: mdl-30092011

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the US. Despite multiple large-scale genetic sequencing studies, identification of predictors of patient survival remains challenging. We performed a comprehensive assessment and integrative analysis of large-scale gene expression datasets, across multiple platforms, to enable discovery of a prognostic gene signature for patient survival in pancreatic cancer. PDAC RNA-Sequencing data from The Cancer Genome Atlas was stratified into Survival+ (>2-year survival) and Survival-(<1-year survival) cohorts (n = 47). Comparisons of RNA expression profiles between survival groups and normal pancreatic tissue expression data from the Gene Expression Omnibus generated an initial PDAC specific prognostic differential expression gene list. The candidate prognostic gene list was then trained on the Australian pancreatic cancer dataset from the ICGC database (n = 103), using iterative sampling based algorithms, to derive a gene signature predictive of patient survival. The gene signature was validated in 2 independent patient cohorts and against existing PDAC subtype classifications. We identified 707 candidate prognostic genes exhibiting differential expression in tumor versus normal tissue. A substantial fraction of these genes was also found to be differentially methylated between survival groups. From the candidate gene list, a 5-gene signature (ADM, ASPM, DCBLD2, E2F7, and KRT6A) was identified. Our signature demonstrated significant power to predict patient survival in two distinct patient cohorts and was independent of AJCC TNM staging. Cross-validation of our gene signature reported a better ROC AUC (≥ 0.8) when compared to existing PDAC survival signatures. Furthermore, validation of our signature through immunohistochemical analysis of patient tumor tissue and existing gene expression subtyping data in PDAC, demonstrated a correlation to the presence of vascular invasion and the aggressive squamous tumor subtype. Assessment of these genes in patient biopsies could help further inform risk-stratification and treatment decisions in pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/mortalité , Pancréas/métabolisme , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/mortalité , Sujet âgé , Algorithmes , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Études de cohortes , Méthylation de l'ADN , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Immunohistochimie , Mâle , Analyse sur microréseau , Adulte d'âge moyen , Modèles biologiques , Pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Pronostic , Analyse de séquence d'ARN , Analyse de survie
8.
Dev Cell ; 45(6): 681-695.e4, 2018 06 18.
Article de Anglais | MEDLINE | ID: mdl-29920274

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) is strongly implicated in tumor cell invasion and metastasis. EMT is thought to be regulated primarily at the transcriptional level through the repressive activity of EMT transcription factors. However, these classical mechanisms have been parsed out almost exclusively in vitro, leaving questions about the programs driving EMT in physiological contexts. Here, using a lineage-labeled mouse model of pancreatic ductal adenocarcinoma to study EMT in vivo, we found that most tumors lose their epithelial phenotype through an alternative program involving protein internalization rather than transcriptional repression, resulting in a "partial EMT" phenotype. Carcinoma cells utilizing this program migrate as clusters, contrasting with the single-cell migration pattern associated with traditionally defined EMT mechanisms. Moreover, many breast and colorectal cancer cell lines utilize this alternative program to undergo EMT. Collectively, these results suggest that carcinoma cells have different ways of losing their epithelial program, resulting in distinct modes of invasion and dissemination.


Sujet(s)
Plasticité cellulaire/physiologie , Cellules épithéliales/physiologie , Transition épithélio-mésenchymateuse/physiologie , Animaux , Cadhérines/métabolisme , Cadhérines/physiologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Souris , Invasion tumorale/génétique , Tumeurs du pancréas/métabolisme , Transduction du signal , Facteurs de transcription/métabolisme , Tumeurs du pancréas
9.
Dev Cell ; 45(6): 696-711.e8, 2018 06 18.
Article de Anglais | MEDLINE | ID: mdl-29920275

RÉSUMÉ

The regulation of metastatic organotropism in pancreatic ductal a denocarcinoma (PDAC) remains poorly understood. We demonstrate, using multiple mouse models, that liver and lung metastatic organotropism is dependent upon p120catenin (p120ctn)-mediated epithelial identity. Mono-allelic p120ctn loss accelerates KrasG12D-driven pancreatic cancer formation and liver metastasis. Importantly, one p120ctn allele is sufficient for E-CADHERIN-mediated cell adhesion. By contrast, cells with bi-allelic p120ctn loss demonstrate marked lung organotropism; however, rescue with p120ctn isoform 1A restores liver metastasis. In a p120ctn-independent PDAC model, mosaic loss of E-CADHERIN expression reveals selective pressure for E-CADHERIN-positive liver metastasis and E-CADHERIN-negative lung metastasis. Furthermore, human PDAC and liver metastases support the premise that liver metastases exhibit predominantly epithelial characteristics. RNA-seq demonstrates differential induction of pathways associated with metastasis and epithelial-to-mesenchymal transition in p120ctn-deficient versus p120ctn-wild-type cells. Taken together, P120CTN and E-CADHERIN mediated epithelial plasticity is an addition to the conceptual framework underlying metastatic organotropism in pancreatic cancer.


Sujet(s)
Caténines/métabolisme , Plasticité cellulaire/physiologie , Tumeurs du pancréas/anatomopathologie , Animaux , Cadhérines/métabolisme , Adhérence cellulaire , Lignée cellulaire tumorale , Cellules épithéliales/métabolisme , Transition épithélio-mésenchymateuse/physiologie , Régulation de l'expression des gènes tumoraux/génétique , Humains , Tumeurs du foie/génétique , Tumeurs du poumon/génétique , Souris , Métastase tumorale/physiopathologie , Conduits pancréatiques/métabolisme , Tumeurs du pancréas/métabolisme , Phosphoprotéines/métabolisme , Isoformes de protéines/métabolisme ,
10.
Cancer Cell ; 29(3): 247-248, 2016 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-26977875

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDA) has a dismal prognosis and is minimally responsive to current chemotherapies. In this issue of Cancer Cell, Xie et al. (2016) identify the transcription factor KLF4 as essential for the early stages of pancreatic carcinogenesis, expanding the repertoire of targets for early intervention strategies.


Sujet(s)
Cellules acineuses/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Facteurs de transcription Krüppel-like/métabolisme , Pancréas/anatomopathologie , Conduits pancréatiques/anatomopathologie , États précancéreux/génétique , Animaux , Humains , Facteur-4 de type Kruppel
11.
Cancer Discov ; 5(10): 1086-97, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26209539

RÉSUMÉ

UNLABELLED: Studies of the cancer genome have demonstrated that tumors are composed of multiple subclones with varied genetic and phenotypic properties. However, little is known about how metastases arise and evolve from these subclones. To understand the cellular dynamics that drive metastasis, we used multicolor lineage-tracing technology in an autochthonous mouse model of pancreatic cancer. Here, we report that precursor lesions exhibit significant clonal heterogeneity but that this diversity decreases during premalignant progression. Furthermore, we present evidence that a significant fraction of metastases are polyclonally seeded by distinct tumor subclones. Finally, we show that clonality during metastatic growth-leading to either monoclonal or polyclonal expansion-differs based on the site of metastatic invasion. These results provide an unprecedented window into the cellular dynamics of tumor evolution and suggest that heterotypic interactions between tumor subpopulations contribute to metastatic progression in native tumors. SIGNIFICANCE: Studies of tumor heterogeneity indicate that distinct tumor subclones interact during cancer progression. Here, we demonstrate by lineage tracing that metastases often involve seeding by more than one clone and that subsequent cellular outgrowth depends on the metastatic site. These findings provide insight into clonal diversity and evolution in metastatic disease.


Sujet(s)
Évolution clonale/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Allèles , Animaux , Marqueurs biologiques tumoraux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Muscle diaphragme/anatomopathologie , Modèles animaux de maladie humaine , Humains , Tumeurs du foie/secondaire , Tumeurs du poumon/secondaire , Souris , Souris transgéniques , Mutation , Métastase tumorale , Tumeurs du péritoine/secondaire
12.
Gastroenterology ; 144(7): 1543-53, 1553.e1, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23454691

RÉSUMÉ

BACKGROUND & AIMS: The Hippo signaling pathway is a context-dependent regulator of cell proliferation, differentiation, and apoptosis in species ranging from Drosophila to humans. In this study, we investigated the role of the core Hippo kinases-Mst1 and Mst2-in pancreatic development and homeostasis. METHODS: We used a Cre/LoxP system to create mice with pancreas-specific disruptions in Mst1 and Mst2 (Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice), the mammalian orthologs of Drosophila Hippo. We used a transgenic approach to overexpress Yap, the downstream mediator of Hippo signaling, in the developing pancreas of mice. RESULTS: Contrary to expectations, the pancreatic mass of Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice was reduced compared with wild-type mice, largely because of postnatal de-differentiation of acinar cells into duct-like cells. Development of this phenotype coincided with postnatal reactivation of YAP expression. Ectopic expression of YAP during the secondary transition (a stage at which YAP is normally absent) blocked differentiation of the endocrine and exocrine compartments, whereas loss of a single Yap allele reduced acinar de-differentiation. The phenotype of Pdx1-Cre;Mst1(-/-);Mst2(fl/fl) mice recapitulated cellular and molecular changes observed during chemical-induced pancreatitis in mice. CONCLUSIONS: The mammalian Hippo kinases, and YAP, maintain postnatal pancreatic acinar differentiation in mice.


Sujet(s)
Protéines adaptatrices de la transduction du signal/physiologie , Pancréas exocrine/croissance et développement , Phosphoprotéines/physiologie , Protein-Serine-Threonine Kinases/physiologie , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Protéines du cycle cellulaire , Différenciation cellulaire , Prolifération cellulaire , Souris , Souris transgéniques , Pancréas exocrine/physiologie , Phosphoprotéines/génétique , Protein-Serine-Threonine Kinases/génétique , Serine-threonine kinase-3 , Transduction du signal , Protéines de signalisation YAP
13.
J Exp Med ; 208(3): 519-33, 2011 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-21282379

RÉSUMÉ

Reactive oxygen species (ROS) have an established role in inflammation and host defense, as they kill intracellular bacteria and have been shown to activate the NLRP3 inflammasome. Here, we find that ROS generated by mitochondrial respiration are important for normal lipopolysaccharide (LPS)-driven production of several proinflammatory cytokines and for the enhanced responsiveness to LPS seen in cells from patients with tumor necrosis factor receptor-associated periodic syndrome (TRAPS), an autoinflammatory disorder caused by missense mutations in the type 1 TNF receptor (TNFR1). We find elevated baseline ROS in both mouse embryonic fibroblasts and human immune cells harboring TRAPS-associated TNFR1 mutations. A variety of antioxidants dampen LPS-induced MAPK phosphorylation and inflammatory cytokine production. However, gp91(phox) and p22(phox) reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits are dispensable for inflammatory cytokine production, indicating that NADPH oxidases are not the source of proinflammatory ROS. TNFR1 mutant cells exhibit altered mitochondrial function with enhanced oxidative capacity and mitochondrial ROS generation, and pharmacological blockade of mitochondrial ROS efficiently reduces inflammatory cytokine production after LPS stimulation in cells from TRAPS patients and healthy controls. These findings suggest that mitochondrial ROS may be a novel therapeutic target for TRAPS and other inflammatory diseases.


Sujet(s)
Syndromes périodiques associés à la cryopyrine/immunologie , Cytokines/physiologie , Espèces réactives de l'oxygène/métabolisme , Récepteur au facteur de nécrose tumorale de type I/physiologie , Animaux , Syndromes périodiques associés à la cryopyrine/génétique , Syndromes périodiques associés à la cryopyrine/physiopathologie , Cytokines/immunologie , Humains , Inflammation/immunologie , Inflammation/physiopathologie , Interleukine-6/immunologie , Interleukine-6/physiologie , Souris , Souches mutantes de souris , Mitochondries/immunologie , Mitochondries/métabolisme , Mitochondries/physiologie , Mitogen-Activated Protein Kinase Kinases/immunologie , Mitogen-Activated Protein Kinase Kinases/physiologie , Mutation , Monoxyde d'azote/physiologie , Récepteur au facteur de nécrose tumorale de type I/génétique , Transduction du signal/immunologie , Transduction du signal/physiologie
14.
Proc Natl Acad Sci U S A ; 107(21): 9801-6, 2010 May 25.
Article de Anglais | MEDLINE | ID: mdl-20457915

RÉSUMÉ

TNF, acting through p55 tumor necrosis factor receptor 1 (TNFR1), contributes to the pathogenesis of many inflammatory diseases. TNFR-associated periodic syndrome (TRAPS, OMIM 142680) is an autosomal dominant autoinflammatory disorder characterized by prolonged attacks of fevers, peritonitis, and soft tissue inflammation. TRAPS is caused by missense mutations in the extracellular domain of TNFR1 that affect receptor folding and trafficking. These mutations lead to loss of normal function rather than gain of function, and thus the pathogenesis of TRAPS is an enigma. Here we show that mutant TNFR1 accumulates intracellularly in peripheral blood mononuclear cells of TRAPS patients and in multiple cell types from two independent lines of knockin mice harboring TRAPS-associated TNFR1 mutations. Mutant TNFR1 did not function as a surface receptor for TNF but rather enhanced activation of MAPKs and secretion of proinflammatory cytokines upon stimulation with LPS. Enhanced inflammation depended on autocrine TNF secretion and WT TNFR1 in mouse and human myeloid cells but not in fibroblasts. Heterozygous TNFR1-mutant mice were hypersensitive to LPS-induced septic shock, whereas homozygous TNFR1-mutant mice resembled TNFR1-deficient mice and were resistant to septic shock. Thus WT and mutant TNFR1 act in concert from distinct cellular locations to potentiate inflammation in TRAPS. These findings establish a mechanism of pathogenesis in autosomal dominant diseases where full expression of the disease phenotype depends on functional cooperation between WT and mutant proteins and also may explain partial responses of TRAPS patients to TNF blockade.


Sujet(s)
Maladies auto-inflammatoires héréditaires/immunologie , Maladies auto-inflammatoires héréditaires/métabolisme , Mutation , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Animaux , Maladies auto-inflammatoires héréditaires/génétique , Humains , Lipopolysaccharides/immunologie , MAP Kinase Kinase 4/métabolisme , Souris , Souris de lignée C57BL , Souris transgéniques , Récepteur au facteur de nécrose tumorale de type I/déficit , Récepteur au facteur de nécrose tumorale de type I/génétique , p38 Mitogen-Activated Protein Kinases/métabolisme
15.
Nano Lett ; 5(4): 603-7, 2005 Apr.
Article de Anglais | MEDLINE | ID: mdl-15826094

RÉSUMÉ

Quantum dots (QDs) could serve as fluorescent scaffolds for effecting specific physiological and pharmacological responses in cells. Here, we conjugate the peptide ligand betaNGF to QD surfaces, and confirm surface modification and single QD nanostructure using AFM. We show that betaNGF-QDs retain bioactivity, activate TrkA receptors, and initiate neuronal differentiation in PC12 cells. Receptor-evoked activity of QD-immobilized ligands has wide-ranging implications for the development of molecular tools and therapeutics targeted at understanding and regulating cell function.


Sujet(s)
Facteur de croissance nerveuse/pharmacologie , Neurites/effets des médicaments et des substances chimiques , Neurones/cytologie , Peptides/composition chimique , Boîtes quantiques , Récepteur trkA/agonistes , Animaux , Différenciation cellulaire , Fluorescence , Ligands , Microscopie à force atomique , Facteur de croissance nerveuse/composition chimique , Cellules PC12 , Rats , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...