Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(6): 1265-1281.e24, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827681

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease that resides within a complex microenvironment, complicating efforts to understand how different cell types contribute to disease progression. We combined single-cell RNA sequencing and genotyping to profile 38,410 cells from 40 bone marrow aspirates, including 16 AML patients and five healthy donors. We then applied a machine learning classifier to distinguish a spectrum of malignant cell types whose abundances varied between patients and between subclones in the same tumor. Cell type compositions correlated with prototypic genetic lesions, including an association of FLT3-ITD with abundant progenitor-like cells. Primitive AML cells exhibited dysregulated transcriptional programs with co-expression of stemness and myeloid priming genes and had prognostic significance. Differentiated monocyte-like AML cells expressed diverse immunomodulatory genes and suppressed T cell activity in vitro. In conclusion, we provide single-cell technologies and an atlas of AML cell states, regulators, and markers with implications for precision medicine and immune therapies. VIDEO ABSTRACT.


Asunto(s)
Leucemia Mieloide Aguda/genética , Transcriptoma/genética , Adulto , Secuencia de Bases/genética , Médula Ósea , Células de la Médula Ósea/citología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Genotipo , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/fisiopatología , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , ARN , Transducción de Señal , Análisis de la Célula Individual/métodos , Microambiente Tumoral , Secuenciación del Exoma/métodos
2.
Nature ; 613(7944): 565-574, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36410718

RESUMEN

Programming T cells to distinguish self from non-self is a vital, multi-step process that occurs in the thymus1-4. Signalling through the pre-T cell receptor (preTCR), a CD3-associated heterodimer comprising an invariant pTα chain and a clone-specific ß chain, is a critical early checkpoint in thymocyte development within the αß T cell lineage5,6. PreTCRs arrayed on CD4-CD8- double-negative thymocytes ligate peptides bound to major histocompatibility complex molecules (pMHC) on thymic stroma, similar to αß T cell receptors that appear on CD4+CD8+ double-positive thymocytes, but via a different molecular docking strategy7-10. Here we show the consequences of these distinct interactions for thymocyte progression using synchronized fetal thymic progenitor cultures that differ in the presence or absence of pMHC on support stroma, and single-cell transcriptomes at key thymocyte developmental transitions. Although major histocompatibility complex (MHC)-negative stroma fosters αß T cell differentiation, the absence of preTCR-pMHC interactions leads to deviant thymocyte transcriptional programming associated with dedifferentiation. Highly proliferative double-negative and double-positive thymocyte subsets emerge, with antecedent characteristics of T cell lymphoblastic and myeloid malignancies. Compensatory upregulation of diverse MHC class Ib proteins in B2m/H2-Ab1 MHC-knockout mice partially safeguards in vivo thymocyte progression, although disseminated double-positive thymic tumours may develop with ageing. Thus, as well as promoting ß chain repertoire broadening for subsequent αß T cell receptor utilization, preTCR-pMHC interactions limit cellular plasticity to facilitate normal thymocyte differentiation and proliferation that, if absent, introduce developmental vulnerabilities.


Asunto(s)
Desdiferenciación Celular , Antígenos de Histocompatibilidad Clase I , Receptores de Antígenos de Linfocitos T , Timocitos , Animales , Ratones , Ratones Noqueados , Simulación del Acoplamiento Molecular , Péptidos/inmunología , Péptidos/metabolismo , Timocitos/citología , Timocitos/inmunología , Timo/citología , Timo/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo
3.
Mol Cell ; 73(6): 1174-1190.e12, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30745086

RESUMEN

Chromatin loops enable transcription-factor-bound distal enhancers to interact with their target promoters to regulate transcriptional programs. Although developmental transcription factors such as active forms of Notch can directly stimulate transcription by activating enhancers, the effect of their oncogenic subversion on the 3D organization of cancer genomes is largely undetermined. By mapping chromatin looping genome-wide in Notch-dependent triple-negative breast cancer and B cell lymphoma, we show that beyond the well-characterized role of Notch as an activator of distal enhancers, Notch regulates its direct target genes by instructing enhancer repositioning. Moreover, a large fraction of Notch-instructed regulatory loops form highly interacting enhancer and promoter spatial clusters termed "3D cliques." Loss- and gain-of-function experiments show that Notch preferentially targets hyperconnected 3D cliques that regulate the expression of crucial proto-oncogenes. Our observations suggest that oncogenic hijacking of developmental transcription factors can dysregulate transcription through widespread effects on the spatial organization of cancer genomes.


Asunto(s)
Transformación Celular Neoplásica/genética , Cromatina/genética , Linfoma de Células B/genética , Oncogenes , Receptores Notch/genética , Neoplasias de la Mama Triple Negativas/genética , Sitios de Unión , Linaje de la Célula/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Ciclina D1/genética , Ciclina D1/metabolismo , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Células HEK293 , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Mutación , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
4.
Eur J Immunol ; 53(9): e2250362, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37366295

RESUMEN

Nonhematopoietic lymph node stromal cells (LNSCs) regulate lymphocyte trafficking, survival, and function for key roles in host defense, autoimmunity, alloimmunity, and lymphoproliferative disorders. However, the study of LNSCs in human diseases is complicated by a dependence on viable lymphoid tissues, which are most often excised prior to establishment of a specific diagnosis. Here, we demonstrate that cryopreservation can be used to bank lymphoid tissue for the study of LNSCs in human disease. Using human tonsils and lymph nodes (LN), lymphoid tissue fragments were cryopreserved for subsequent enzymatic digestion and recovery of viable nonhematopoietic cells. Flow cytometry and single-cell transcriptomics identified comparable proportions of LN stromal cell types in fresh and cryopreserved tissue. Moreover, cryopreservation had little effect on transcriptional profiles, which showed significant overlap between tonsils and LN. The presence and spatial distribution of transcriptionally defined cell types were confirmed by in situ analyses. Our broadly applicable approach promises to greatly enable research into the roles of LNSCs in human disease.


Asunto(s)
Bancos de Muestras Biológicas , Criopreservación , Humanos , Linfocitos , Ganglios Linfáticos/patología , Células del Estroma
6.
Semin Cancer Biol ; 85: 95-106, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-33862222

RESUMEN

Notch receptors participate in a conserved pathway in which ligands expressed on neighboring cells trigger a series of proteolytic cleavages that allow the intracellular portion of the receptor to travel to the nucleus and form a short-lived transcription complex that turns on target gene expression. The directness and seeming simplicity of this signaling mechanism belies the complexity of the outcomes of Notch signaling in normal cells, which are highly context and dosage dependent. This complexity is reflected in the diverse roles of Notch in cancers of various types, in which Notch may be oncogenic or tumor suppressive and may have a wide spectrum of effects on tumor cells and stromal elements. This review provides an overview of the roles of Notch in cancer and discusses challenges to clinical translation of Notch targeting agents as well as approaches that may overcome these hurdles.


Asunto(s)
Neoplasias , Receptores Notch , Humanos , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes , Ligandos
7.
Blood ; 137(18): 2463-2480, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33227818

RESUMEN

Lineage plasticity and stemness have been invoked as causes of therapy resistance in cancer, because these flexible states allow cancer cells to dedifferentiate and alter their dependencies. We investigated such resistance mechanisms in relapsed/refractory early T-cell progenitor acute lymphoblastic leukemia (ETP-ALL) carrying activating NOTCH1 mutations via full-length single-cell RNA sequencing (scRNA-seq) of malignant and microenvironmental cells. We identified 2 highly distinct stem-like states that critically differed with regard to cell cycle and oncogenic signaling. Fast-cycling stem-like leukemia cells demonstrated Notch activation and were effectively eliminated in patients by Notch inhibition, whereas slow-cycling stem-like cells were Notch independent and rather relied on PI3K signaling, likely explaining the poor efficacy of Notch inhibition in this disease. Remarkably, we found that both stem-like states could differentiate into a more mature leukemia state with prominent immunomodulatory functions, including high expression of the LGALS9 checkpoint molecule. These cells promoted an immunosuppressive leukemia ecosystem with clonal accumulation of dysfunctional CD8+ T cells that expressed HAVCR2, the cognate receptor for LGALS9. Our study identified complex interactions between signaling programs, cellular plasticity, and immune programs that characterize ETP-ALL, illustrating the multidimensionality of tumor heterogeneity. In this scenario, combination therapies targeting diverse oncogenic states and the immune ecosystem seem most promising to successfully eliminate tumor cells that escape treatment through coexisting transcriptional programs.


Asunto(s)
Carcinogénesis , Galectinas/metabolismo , Regulación Leucémica de la Expresión Génica , Evasión Inmune , Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Análisis de la Célula Individual/métodos , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Niño , Preescolar , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Galectinas/genética , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Pronóstico , RNA-Seq/métodos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
8.
Nature ; 547(7662): 217-221, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28678778

RESUMEN

Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma/inmunología , Melanoma/terapia , Medicina de Precisión/métodos , Secuencia de Aminoácidos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos de Neoplasias/química , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/química , Secuenciación de Nucleótidos de Alto Rendimiento , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Aprendizaje Automático , Melanoma/genética , Mutación , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/prevención & control , Seguridad del Paciente , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
9.
Proc Natl Acad Sci U S A ; 117(28): 16292-16301, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601208

RESUMEN

Notch pathway signaling is implicated in several human cancers. Aberrant activation and mutations of Notch signaling components are linked to tumor initiation, maintenance, and resistance to cancer therapy. Several strategies, such as monoclonal antibodies against Notch ligands and receptors, as well as small-molecule γ-secretase inhibitors (GSIs), have been developed to interfere with Notch receptor activation at proximal points in the pathway. However, the use of drug-like small molecules to target the downstream mediators of Notch signaling, the Notch transcription activation complex, remains largely unexplored. Here, we report the discovery of an orally active small-molecule inhibitor (termed CB-103) of the Notch transcription activation complex. We show that CB-103 inhibits Notch signaling in primary human T cell acute lymphoblastic leukemia and other Notch-dependent human tumor cell lines, and concomitantly induces cell cycle arrest and apoptosis, thereby impairing proliferation, including in GSI-resistant human tumor cell lines with chromosomal translocations and rearrangements in Notch genes. CB-103 produces Notch loss-of-function phenotypes in flies and mice and inhibits the growth of human breast cancer and leukemia xenografts, notably without causing the dose-limiting intestinal toxicity associated with other Notch inhibitors. Thus, we describe a pharmacological strategy that interferes with Notch signaling by disrupting the Notch transcription complex and shows therapeutic potential for treating Notch-driven cancers.


Asunto(s)
Receptores Notch/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Activación Transcripcional/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Drosophila , Resistencia a Antineoplásicos/efectos de los fármacos , Células HeLa , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/química , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Ratones , Mutación , Fenotipo , Multimerización de Proteína , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/uso terapéutico
10.
Blood ; 136(26): 3051-3055, 2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-32961550

RESUMEN

Adult-onset hemophagocytic lymphohistiocytosis (HLH) is a rare, life-threatening disease of immune hyperactivation. Unlike pediatric HLH, adult HLH is rarely driven by germline genetic variants. Although numerous precipitating etiologies have been identified, the reason that HLH occurs in only a subset of individuals and how other factors contribute to the disease remains unknown. We hypothesized that clonal hematopoiesis (CH), a state in which somatic mutations in blood cells cause an expanded population of mutant hematopoietic cells and drive an aberrant inflammatory state, could contribute to adult-onset HLH. In a highly annotated cohort of older adults with HLH we found that CH was more prevalent than in control cohorts. Using the adult-onset HLH mouse model in which repeated treatments of the TLR9 agonist, ODN1826, was delivered to the mouse, we observed that macrophages carrying mutations in Tet2, one of the most commonly mutated genes in CH, have an enhanced inflammatory response to TLR9 agonism. Finally, mice carrying Tet2 mutations in the hematopoietic compartment (a common model for CH) displayed an exaggerated response to TLR9 agonism, including worse splenomegaly and anemia. Our data suggest that CH is more common in individuals with adult-onset HLH and can contribute to the pathophysiology of this disease.


Asunto(s)
Hematopoyesis Clonal , Linfohistiocitosis Hemofagocítica/metabolismo , Mutación , Adulto , Edad de Inicio , Anciano , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Humanos , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/patología , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
11.
PLoS Genet ; 15(4): e1008039, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30970016

RESUMEN

The SWI/SNF-family chromatin remodeling protein ATRX is a tumor suppressor in sarcomas, gliomas and other malignancies. Its loss of function facilitates the alternative lengthening of telomeres (ALT) pathway in tumor cells, while it also affects Polycomb repressive complex 2 (PRC2) silencing of its target genes. To further define the role of inactivating ATRX mutations in carcinogenesis, we knocked out atrx in our previously reported p53/nf1-deficient zebrafish line that develops malignant peripheral nerve sheath tumors and gliomas. Complete inactivation of atrx using CRISPR/Cas9 was lethal in developing fish and resulted in an alpha-thalassemia-like phenotype including reduced alpha-globin expression. In p53/nf1-deficient zebrafish neither peripheral nerve sheath tumors nor gliomas showed accelerated onset in atrx+/- fish, but these fish developed various tumors that were not observed in their atrx+/+ siblings, including epithelioid sarcoma, angiosarcoma, undifferentiated pleomorphic sarcoma and rare types of carcinoma. These cancer types are included in the AACR Genie database of human tumors associated with mutant ATRX, indicating that our zebrafish model reliably mimics a role for ATRX-loss in the early pathogenesis of these human cancer types. RNA-seq of p53/nf1- and p53/nf1/atrx-deficient tumors revealed that down-regulation of telomerase accompanied ALT-mediated lengthening of the telomeres in atrx-mutant samples. Moreover, inactivating mutations in atrx disturbed PRC2-target gene silencing, indicating a connection between ATRX loss and PRC2 dysfunction in cancer development.


Asunto(s)
Sarcoma Experimental/etiología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína Nuclear Ligada al Cromosoma X/deficiencia , Proteína Nuclear Ligada al Cromosoma X/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Carcinogénesis/genética , Carcinogénesis/metabolismo , Modelos Animales de Enfermedad , Eritropoyesis , Femenino , Técnicas de Inactivación de Genes , Globinas/genética , Humanos , Mutación con Pérdida de Función , Masculino , Neurofibromina 1/deficiencia , Neurofibromina 1/genética , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Homeostasis del Telómero/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
12.
Genes Dev ; 28(6): 576-93, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24637115

RESUMEN

Notch1 is required to generate the earliest embryonic hematopoietic stem cells (HSCs); however since Notch-deficient embryos die early in gestation, additional functions for Notch in embryonic HSC biology have not been described. We used two complementary genetic models to address this important biological question. Unlike Notch1-deficient mice, mice lacking the conserved Notch1 transcriptional activation domain (TAD) show attenuated Notch1 function in vivo and survive until late gestation, succumbing to multiple cardiac abnormalities. Notch1 TAD-deficient HSCs emerge and successfully migrate to the fetal liver but are decreased in frequency by embryonic day 14.5. In addition, TAD-deficient fetal liver HSCs fail to compete with wild-type HSCs in bone marrow transplant experiments. This phenotype is independently recapitulated by conditional knockout of Rbpj, a core Notch pathway component. In vitro analysis of Notch1 TAD-deficient cells shows that the Notch1 TAD is important to properly assemble the Notch1/Rbpj/Maml trimolecular transcription complex. Together, these studies reveal an essential role for the Notch1 TAD in fetal development and identify important cell-autonomous functions for Notch1 signaling in fetal HSC homeostasis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Receptor Notch1/metabolismo , Transducción de Señal , Animales , Línea Celular , Células Madre Fetales , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Ratones , Mutación , Estructura Terciaria de Proteína/genética , Receptor Notch1/genética , Análisis de Supervivencia
13.
Blood ; 134(17): 1430-1440, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31383641

RESUMEN

Antibodies that bind CD47 on tumor cells and prevent interaction with SIRPα on phagocytes are active against multiple cancer types including T-cell lymphoma (TCL). Here we demonstrate that surface CD47 is heterogeneously expressed across primary TCLs, whereas major histocompatibility complex (MHC) class I, which can also suppress phagocytosis, is ubiquitous. Multiple monoclonal antibodies (mAbs) that block CD47-SIRPα interaction promoted phagocytosis of TCL cells, which was enhanced by cotreatment with antibodies targeting MHC class I. Expression levels of surface CD47 and genes that modulate CD47 pyroglutamation did not correlate with the extent of phagocytosis induced by CD47 blockade in TCL lines. In vivo treatment of multiple human TCL patient-derived xenografts or an immunocompetent murine TCL model with a short course of anti-CD47 mAb markedly reduced lymphoma burden and extended survival. Depletion of macrophages reduced efficacy in vivo, whereas depletion of neutrophils had no effect. F(ab')2-only fragments of anti-CD47 antibodies failed to induce phagocytosis by human macrophages, indicating a requirement for Fc-Fcγ receptor interactions. In contrast, F(ab')2-only fragments increased phagocytosis by murine macrophages independent of SLAMF7-Mac-1 interaction. Full-length anti-CD47 mAbs also induced phagocytosis by Fcγ receptor-deficient murine macrophages. An immunoglobulin G1 anti-CD47 mAb induced phagocytosis and natural killer cell-mediated cytotoxicity of TCL cells that was augmented by cotreatment with mogamulizumab, an anti-CCR4 mAb, or a mAb blocking MHC class I. These studies help explain the disparate activity of monotherapy with agents that block CD47 in murine models compared with patients. They also have direct translational implications for the deployment of anti-CD47 mAbs alone or in combination.


Asunto(s)
Antígenos de Diferenciación/inmunología , Antineoplásicos Inmunológicos/farmacología , Antígeno CD47/inmunología , Linfoma de Células T/tratamiento farmacológico , Receptores de IgG/inmunología , Receptores Inmunológicos/inmunología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno CD47/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Ratones , Receptores Fc/inmunología
14.
Circulation ; 139(1): 78-96, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30586693

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) increases cardiovascular risk. Underlying mechanisms, however, remain obscure. The uremic toxin indoxyl sulfate is an independent cardiovascular risk factor in CKD. We explored the potential impact of indoxyl sulfate on proinflammatory activation of macrophages and its underlying mechanisms. METHODS: We examined in vitro the effects of clinically relevant concentrations of indoxyl sulfate on proinflammatory responses of macrophages and the roles of organic anion transporters and organic anion transporting polypeptides (OATPs). A systems approach, involving unbiased global proteomics, bioinformatics, and network analysis, then explored potential key pathways. To address the role of Delta-like 4 (Dll4) in indoxyl sulfate-induced macrophage activation and atherogenesis in CKD in vivo, we used 5/6 nephrectomy and Dll4 antibody in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. To further determine the relative contribution of OATP2B1 or Dll4 to proinflammatory activation of macrophages and atherogenesis in vivo, we used siRNA delivered by macrophage-targeted lipid nanoparticles in mice. RESULTS: We found that indoxyl sulfate-induced proinflammatory macrophage activation is mediated by its uptake through transporters, including OATP2B1, encoded by the SLCO2B1 gene. The global proteomics identified potential mechanisms, including Notch signaling and the ubiquitin-proteasome pathway, that mediate indoxyl sulfate-triggered proinflammatory macrophage activation. We chose the Notch pathway as an example of key candidates for validation of our target discovery platform and for further mechanistic studies. As predicted computationally, indoxyl sulfate triggered Notch signaling, which was preceded by the rapid induction of Dll4 protein. Dll4 induction may result from inhibition of the ubiquitin-proteasome pathway, via the deubiquitinating enzyme USP5. In mice, macrophage-targeted OATP2B1/Slco2b1 silencing and Dll4 antibody inhibited proinflammatory activation of peritoneal macrophages induced by indoxyl sulfate. In low-density lipoprotein receptor-deficient mice, Dll4 antibody abolished atherosclerotic lesion development accelerated in Ldlr-/- mice. Moreover, coadministration of indoxyl sulfate and OATP2B1/Slco2b1 or Dll4 siRNA encapsulated in macrophage-targeted lipid nanoparticles in Ldlr-/- mice suppressed lesion development. CONCLUSIONS: These results suggest that novel crosstalk between OATP2B1 and Dll4-Notch signaling in macrophages mediates indoxyl sulfate-induced vascular inflammation in CKD.


Asunto(s)
Aterosclerosis/metabolismo , Indicán/toxicidad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Receptores Notch/metabolismo , Insuficiencia Renal Crónica/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Proteínas de Unión al Calcio , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Transportadores de Anión Orgánico/genética , Fenotipo , Placa Aterosclerótica , Células RAW 264.7 , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Notch/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
15.
Mod Pathol ; 33(6): 1135-1145, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31896808

RESUMEN

Greater than 90% of cases of systemic mastocytosis (SM) harbor pathogenic KIT mutations, particularly KITD816V. Prognostically-significant pathogenic KIT mutations also occur in 30-40% of core binding factor-associated acute myeloid leukemia (CBF-AML), but are uncommonly associated with concurrent SM. By comparison, the occurrence of SM in other myeloid neoplasms bearing pathogenic KIT mutations, particularly those with a chronic course, is poorly understood. Review of clinical next-generation sequencing (NGS) performed at our institutions in patients with known or suspected hematologic malignancies over an 8-year period revealed 64 patients with both a pathogenic KIT mutation detected at one or more timepoints and available bone marrow biopsy materials. Patients with KITD816V-mutated myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN), or overlap MDS/MPN (n = 22) accounted for approximately one-third of our cohort (34%). Comprehensive morphologic and immunophenotypic characterization revealed that nearly all cases (n = 20, 91%) exhibited concurrent SM. In contrast, of the 18 patients (28%) with AML and KITD816V, only eight (44%) showed evidence of SM at any point in their disease course (p = 0.0021); of these eight, the AML component was characterized as AML with myelodysplasia-related changes (AML-MRC) in all but one instance (n = 7, 87%). Twelve patients (19%) had pathogenic KIT mutations other than p.D816V, all in the setting of AML (CFB-AML, n = 7; AML, not otherwise specified, n = 2; AML-MRC, n = 1; acute promyelocytic leukemia, n = 1); only two of these patients (17%), both with CBF-AML, exhibited concurrent SM. The remaining 12 patients (19%) had SM without evidence of an associated hematological neoplasm (AHN). For nearly one-third of the 30 SM-AHN patients in our cohort (n = 9, 30%), the SM component of their disease was not initially clinicopathologically recognized. We propose that identification of the KITD816V mutation in patients diagnosed with MDS, MPN, MDS/MPN, or AML-MRC should trigger reflex testing for SM.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mastocitosis/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Proteínas Proto-Oncogénicas c-kit/genética , Análisis Mutacional de ADN , Humanos , Leucemia Mieloide Aguda/patología , Mastocitosis/patología , Mutación , Síndromes Mielodisplásicos/patología , Trastornos Mieloproliferativos/patología
16.
Blood ; 132(9): 935-947, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-29769264

RESUMEN

Patients with angioimmunoblastic T-cell lymphoma (AITL) and other peripheral T-cell lymphomas that harbor features of follicular helper T (TFH) cells have a very poor prognosis. These lymphomas commonly present with paraneoplastic autoimmunity and lymphopenia. RhoA G17V mutation is present in 60% of TFH-like lymphomas, but its role in tumorigenesis is poorly understood. We generated transgenic mice that express RhoA G17V under the control of murine CD4 regulatory elements at levels comparable to a heterozygous mutation (tgRhoA mice). These mice had markedly reduced naive T cells but relatively increased TFH-cell populations. Surprisingly, naive CD4 T cells expressing RhoA G17V were hyperreactive to T-cell receptor stimulation. All tgRhoA mice developed autoimmunity that included a cellular infiltrate within ears and tails that was recapitulated in wild-type (WT) recipients after bone marrow transplantation. Older tgRhoA mice developed elevated serum titers of anti-double-stranded DNA antibodies and renal immune complex deposition. RhoA G17V mice crossed with Tet2fl/fl; Vav-Cre+ mice, which delete Tet2 throughout the hematopoietic compartment, developed T-cell lymphomas that retained histologic and immunophenotypic features of AITL and had transcriptional signatures enriched for mechanistic target of rapamycin (mTOR)-associated genes. Transplanted tumors were responsive to the mTOR inhibitor everolimus, providing a possible strategy for targeting RhoA G17V. Taken together, these data indicate that RhoA G17V contributes to both neoplastic and paraneoplastic phenotypes similar to those observed in patients with TFH lymphomas.


Asunto(s)
Linfoma de Células T , Mutación Missense , Proteínas de Neoplasias , Linfocitos T Colaboradores-Inductores , Proteínas de Unión al GTP rho , Sustitución de Aminoácidos , Animales , Anticuerpos Antinucleares/inmunología , Enfermedades Autoinmunes , Linfoma Folicular/genética , Linfoma Folicular/inmunología , Linfoma Folicular/patología , Linfoma de Células T/genética , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/inmunología , Proteína de Unión al GTP rhoA
17.
Blood ; 131(8): 888-898, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29233821

RESUMEN

Duvelisib (IPI-145) is an oral inhibitor of phosphatidylinositol 3-kinase (PI3K)-δ/γ isoforms currently in clinical development. PI3K-δ/γ inhibition may directly inhibit malignant T-cell growth, making duvelisib a promising candidate for patients with peripheral (PTCL) or cutaneous (CTCL) T-cell lymphoma. Inhibition of either isoform may also contribute to clinical responses by modulating nonmalignant immune cells. We investigated these dual effects in a TCL cohort from a phase 1, open-label study of duvelisib in patients with relapsed or refractory PTCL (n = 16) and CTCL (n = 19), along with in vitro and in vivo models of TCL. The overall response rates in patients with PTCL and CTCL were 50.0% and 31.6%, respectively (P = .32). There were 3 complete responses, all among patients with PTCL. Activity was seen across a wide spectrum of subtypes. The most frequently observed grade 3 and 4 adverse events were transaminase increases (40% alanine aminotransferase, 17% aspartate aminotransferase), maculopapular rash (17%), and neutropenia (17%). Responders and nonresponders had markedly different changes in serum cytokine profiles induced by duvelisib. In vitro, duvelisib potently killed 3 of 4 TCL lines with constitutive phospho-AKT (pAKT) vs 0 of 7 lines lacking pAKT (P = .024) and exceeded cell killing by the PI3K-δ-specific inhibitor idelalisib. Administration of duvelisib to mice engrafted with a PTCL patient-derived xenograft resulted in a shift among tumor-associated macrophages from the immunosuppressive M2-like phenotype to the inflammatory M1-like phenotype. In summary, duvelisib demonstrated promising clinical activity and an acceptable safety profile in relapsed/refractory TCL, as well as preclinical evidence of both tumor cell-autonomous and immune-mediated effects. This trial was registered at www.clinicaltrials.gov as #NCT01476657.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Linfoma Cutáneo de Células T/tratamiento farmacológico , Linfoma de Células T Periférico/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Purinas/administración & dosificación , Purinas/farmacocinética , Neoplasias Cutáneas/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Fosfatidilinositol 3-Quinasa Clase Ib , Femenino , Humanos , Isoquinolinas/farmacología , Linfoma Cutáneo de Células T/enzimología , Linfoma Cutáneo de Células T/patología , Linfoma de Células T Periférico/enzimología , Linfoma de Células T Periférico/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Pronóstico , Purinas/farmacología , Seguridad , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Distribución Tisular
18.
Blood ; 132(14): 1495-1506, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30089630

RESUMEN

Chimeric antigen receptor (CAR) T cells have emerged as a novel form of treatment of patients with B-cell malignancies. In particular, anti-CD19 CAR T-cell therapy has effected impressive clinical responses in B-cell acute lymphoblastic leukemia and diffuse large B-cell lymphoma. However, not all patients respond, and relapse with antigen loss has been observed in all patient subsets. Here, we report on the design and optimization of a novel CAR directed to the surface antigen CD37, which is expressed in B-cell non-Hodgkin lymphomas, in chronic lymphocytic leukemia, and in some cases of cutaneous and peripheral T-cell lymphomas. We found that CAR-37 T cells demonstrated antigen-specific activation, cytokine production, and cytotoxic activity in models of B- and T-cell lymphomas in vitro and in vivo, including patient-derived xenografts. Taken together, these results are the first showing that T cells expressing anti-CD37 CAR have substantial activity against 2 different lymphoid lineages, without evidence of significant T-cell fratricide. Furthermore, anti-CD37 CARs were readily combined with anti-CD19 CARs to generate dual-specific CAR T cells capable of recognizing CD19 and CD37 alone or in combination. Our findings indicate that CD37-CAR T cells represent a novel therapeutic agent for the treatment of patients with CD37-expressing lymphoid malignancies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Linfoma de Células T/terapia , Tetraspaninas/inmunología , Animales , Antígenos de Neoplasias/análisis , Línea Celular Tumoral , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Ratones , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/trasplante , Tetraspaninas/análisis , Tetraspaninas/antagonistas & inhibidores
19.
Nature ; 513(7519): 512-6, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25043004

RESUMEN

Mutations that deregulate Notch1 and Ras/phosphoinositide 3 kinase (PI3K)/Akt signalling are prevalent in T-cell acute lymphoblastic leukaemia (T-ALL), and often coexist. Here we show that the PI3K inhibitor GDC-0941 is active against primary T-ALLs from wild-type and Kras(G12D) mice, and addition of the MEK inhibitor PD0325901 increases its efficacy. Mice invariably relapsed after treatment with drug-resistant clones, most of which unexpectedly had reduced levels of activated Notch1 protein, downregulated many Notch1 target genes, and exhibited cross-resistance to γ-secretase inhibitors. Multiple resistant primary T-ALLs that emerged in vivo did not contain somatic Notch1 mutations present in the parental leukaemia. Importantly, resistant clones upregulated PI3K signalling. Consistent with these data, inhibiting Notch1 activated the PI3K pathway, providing a likely mechanism for selection against oncogenic Notch1 signalling. These studies validate PI3K as a therapeutic target in T-ALL and raise the unexpected possibility that dual inhibition of PI3K and Notch1 signalling could promote drug resistance in T-ALL.


Asunto(s)
Resistencia a Antineoplásicos , Indazoles/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Inhibidores de Proteínas Quinasas/farmacología , Receptor Notch1/metabolismo , Sulfonamidas/farmacología , Animales , Benzamidas/farmacología , Benzamidas/uso terapéutico , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Células Clonales/patología , Difenilamina/análogos & derivados , Difenilamina/farmacología , Difenilamina/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Genes ras/genética , Indazoles/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Notch1/química , Receptor Notch1/deficiencia , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Sulfonamidas/uso terapéutico
20.
Trans Am Clin Climatol Assoc ; 131: 147-156, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32675855

RESUMEN

Notch receptors participate is a highly conserved signaling pathway that regulates numerous facets of cellular behavior, has protean roles during development and in adult tissue homeostasis, and is frequently dysregulated in human diseases, particularly cancer. These relationships to disease and the ability to modulate Notch signaling at multiple levels have engendered attempts to target Notch therapeutically, but incomplete understanding of the outcomes of Notch activation and on-target toxicity have stymied efforts to date. Using well-controlled experimental systems, we have pursued studies that seek to understand how Notch influences the behavior of different types of cancer cells. Our work suggests that Notch effects are defined by epigenetic landscapes that are "laid out" by upstream pioneer transcription factors, which act to delineate the outcome of Notch activation. These insights define some of the "rules" that govern Notch functions and constitute one step toward bringing safe and effective targeting of Notch to fruition.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA