Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38405728

RESUMEN

In mammals, olfactory sensory neurons (OSNs) are born throughout life, presumably solely to replace neurons lost via turnover or injury. This assumption follows from the hypothesis that olfactory neurogenesis is strictly stochastic with respect to neuron subtype, as defined by the single odorant receptor allele that each neural precursor stochastically chooses out of hundreds of possibilities. This hypothesis is challenged by recent findings that the birthrates of a fraction of subtypes are selectively diminished by olfactory deprivation. These findings raise questions about how, and why, olfactory stimuli are required to promote the neurogenesis of some OSN subtypes, including whether the stimuli are generic (e.g., broadly activating odors or mechanical stimuli) or specific (e.g., discrete odorants). Based on RNA-seq and scRNA-seq analyses, we hypothesized that the neurogenic stimuli are specific odorants that selectively activate the same OSN subtypes whose birthrates are accelerated. In support of this, we have found, using subtype-specific OSN birthdating, that exposure to male and musk odors can accelerate the birthrates of responsive OSNs. Collectively, our findings reveal that certain odor experiences can selectively "amplify" specific OSN subtypes, and that persistent OSN neurogenesis may serve, in part, an adaptive function.

2.
STAR Protoc ; 4(3): 102432, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37436902

RESUMEN

Mammals typically have hundreds of distinct olfactory sensory neuron subtypes, each defined by expression of a specific odorant receptor gene, which undergo neurogenesis throughout life at rates that can depend on olfactory experience. Here, we present a protocol to quantify the birthrates of specific neuron subtypes via the simultaneous detection of corresponding receptor mRNAs and 5-ethynyl-2'-deoxyuridine. For preparation prior to beginning the protocol, we detail procedures for generating odorant receptor-specific riboprobes and experimental mouse olfactory epithelial tissue sections. For complete details on the use and execution of this protocol, please refer to van der Linden et al. (2020).1.


Asunto(s)
Neuronas Receptoras Olfatorias , Receptores Odorantes , Animales , Ratones , Mucosa Olfatoria/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Neurogénesis/genética , Odorantes , Mamíferos/metabolismo
3.
Cell Rep ; 33(1): 108210, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33027656

RESUMEN

In mammals, olfactory sensory neurons (OSNs) are born throughout life, ostensibly solely to replace damaged OSNs. During differentiation, each OSN precursor "chooses," out of hundreds of possibilities, a single odorant receptor (OR) gene, which defines the identity of the mature OSN. The relative neurogenesis rates of the hundreds of distinct OSN "subtypes" are thought to be constant, as they are determined by a stochastic process in which each OR is chosen with a fixed probability. Here, using histological, single-cell, and targeted affinity purification approaches, we show that closing one nostril in mice selectively reduces the number of newly generated OSNs of specific subtypes. Moreover, these reductions depend on an animal's age and/or environment. Stimulation-dependent changes in the number of new OSNs are not attributable to altered rates of cell survival but rather production. Our findings indicate that the relative birth rates of distinct OSN subtypes depend on olfactory experience.


Asunto(s)
Neuronas Receptoras Olfatorias/metabolismo , Receptores Odorantes/genética , Animales , Diferenciación Celular , Ratones
4.
Sci Data ; 5: 180260, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30512012

RESUMEN

Olfactory experience can alter the molecular and cellular composition of chemosensory neurons within the olfactory sensory epithelia of mice. We sought to investigate the scope of cellular and molecular changes within a mouse's olfactory system as a function of its exposure to complex and salient sets of odors: those emitted from members of the opposite sex. We housed mice either separated from members of the opposite sex (sex-separated) or together with members of the opposite sex (sex-combined) until six months of age, resulting in the generation of four cohorts of mice. From each mouse, the main olfactory epithelium (MOE), vomeronasal organ (VNO), and olfactory bulb (OB) were removed and RNA-extracted. A total of 36 RNA samples, representing three biological replicates per sex/condition/tissue combination, were analyzed for integrity and used to prepare RNA-seq libraries, which were subsequently analyzed via qPCR for the presence of tissue- or sex-specific markers. Libraries were paired-end sequenced to a depth of ~20 million fragments per replicate and the data were analyzed using the Tuxedo suite.


Asunto(s)
Perfilación de la Expresión Génica , Bulbo Olfatorio , Mucosa Olfatoria , Receptores Odorantes/genética , Caracteres Sexuales , Órgano Vomeronasal , Animales , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Odorantes , Bulbo Olfatorio/fisiología , Mucosa Olfatoria/fisiología , Órgano Vomeronasal/fisiología
5.
Nat Commun ; 9(1): 5081, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30514924

RESUMEN

Within the mammalian olfactory sensory epithelium, experience-dependent changes in the rate of neuronal turnover can alter the relative abundance of neurons expressing specific chemoreceptors. Here we investigate how the mouse olfactory sensory receptor repertoire changes as a function of exposure to odors emitted from members of the opposite sex, which are highly complex and sexually dimorphic. Upon housing mice either sex-separated or sex-combined until six months of age, we find that sex-separated mice exhibit significantly more numerous differentially expressed genes within their olfactory epithelia. A subset of these chemoreceptors exhibit altered expression frequencies following both sex-separation and olfactory deprivation. We show that several of these receptors detect either male- or female-specific odors. We conclude that the distinct odor experiences of sex-separated male and female mice induce sex-specific differences in the abundance of neurons that detect sexually dimorphic odors.


Asunto(s)
Neuronas Receptoras Olfatorias/metabolismo , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Olfato/fisiología , Animales , Conducta Animal , Femenino , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Odorantes , Mucosa Olfatoria/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo
6.
Dev Neurobiol ; 78(12): 1171-1190, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30246932

RESUMEN

As the catalytic component of γ-secretase, presenilin (PS) has long been studied in the context of Alzheimer's disease through cleaving the amyloid precursor protein. PS/γ-secretase, however, also cleaves a multitude of single-pass transmembrane proteins that are important during development, including Notch, the netrin receptor DCC, cadherins, drebrin-A, and the EphB2 receptor. Because transgenic PS-KO mice do not survive to birth, studies of this molecule during later embryonic or early postnatal stages of development have been carried out using cell cultures or conditional knock-out mice, respectively. As a result, the function of PS in synapse formation had not been well-addressed. Here, we study the role of PS in the developing Xenopus tadpole retinotectal circuit, an in-vivo model that allows for protein expression to be manipulated specifically during the peak of synapse formation between retinal ganglion cells and tectal neurons. We found that inhibiting PS in the postsynaptic tectal neurons impaired tadpole visual avoidance behavior. Whole cell recordings indicated weaker retinotectal synaptic transmission which was characterized by significant reductions in both NMDA receptor (NMDAR)- and AMPA receptor (AMPAR)-mediated currents. We also found that expression of the C-tail fragment of the EphB2 receptor, which is normally cleaved by PS/γ-secretase and which has been shown to upregulate NMDARs at the synapse, rescued the reduced NMDAR-mediated responses. Our data determine that normal PS function is important for proper formation and strengthening of retinotectal synapses through cleaving the EphB2 receptor.


Asunto(s)
Conducta Animal/fisiología , Fenómenos Electrofisiológicos/fisiología , Presenilinas/metabolismo , Receptor EphB2/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Células Ganglionares de la Retina/fisiología , Colículos Superiores/fisiología , Sinapsis/fisiología , Percepción Visual/fisiología , Animales , Larva , Colículos Superiores/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Xenopus
7.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30266714

RESUMEN

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN- population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFß signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide-dependent manner, whereas FAP+PDPN- pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.


Asunto(s)
Neoplasias de la Mama/patología , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Proliferación Celular , Endopeptidasas , Femenino , Regulación de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Pericitos/patología , Células del Estroma/patología , Linfocitos T/patología
8.
Nature ; 554(7693): 544-548, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29443960

RESUMEN

Therapeutic antibodies that block the programmed death-1 (PD-1)-programmed death-ligand 1 (PD-L1) pathway can induce robust and durable responses in patients with various cancers, including metastatic urothelial cancer. However, these responses only occur in a subset of patients. Elucidating the determinants of response and resistance is key to improving outcomes and developing new treatment strategies. Here we examined tumours from a large cohort of patients with metastatic urothelial cancer who were treated with an anti-PD-L1 agent (atezolizumab) and identified major determinants of clinical outcome. Response to treatment was associated with CD8+ T-effector cell phenotype and, to an even greater extent, high neoantigen or tumour mutation burden. Lack of response was associated with a signature of transforming growth factor ß (TGFß) signalling in fibroblasts. This occurred particularly in patients with tumours, which showed exclusion of CD8+ T cells from the tumour parenchyma that were instead found in the fibroblast- and collagen-rich peritumoural stroma; a common phenotype among patients with metastatic urothelial cancer. Using a mouse model that recapitulates this immune-excluded phenotype, we found that therapeutic co-administration of TGFß-blocking and anti-PD-L1 antibodies reduced TGFß signalling in stromal cells, facilitated T-cell penetration into the centre of tumours, and provoked vigorous anti-tumour immunity and tumour regression. Integration of these three independent biological features provides the best basis for understanding patient outcome in this setting and suggests that TGFß shapes the tumour microenvironment to restrain anti-tumour immunity by restricting T-cell infiltration.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/inmunología , Urotelio/patología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Estudios de Cohortes , Colágeno/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Inmunoterapia , Ratones , Mutación , Metástasis de la Neoplasia , Fenotipo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Neoplasias Urológicas/genética , Neoplasias Urológicas/patología , Urotelio/efectos de los fármacos , Urotelio/inmunología
9.
Cancer Immunol Immunother ; 67(2): 329-339, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29313073

RESUMEN

Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional (3D) structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a mouse tumor model that has highly endogenous mTEM1 expression in the vasculature. Our data indicate that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.

10.
Cancer Immunol Immunother ; 66(3): 367-378, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27933426

RESUMEN

Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here, we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a tumor mouse model that has highly endogenous mTEM1 expression in the vasculature. Our data indicated that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Fragmentos de Inmunoglobulinas/inmunología , Inmunotoxinas/inmunología , Nanopartículas/administración & dosificación , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Secuencia de Aminoácidos , Animales , Antígenos CD/biosíntesis , Epítopos/inmunología , Humanos , Inmunoterapia/métodos , Inmunotoxinas/farmacocinética , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Nanopartículas/metabolismo , Proteínas de Neoplasias/biosíntesis
11.
Trends Genet ; 31(9): 516-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26299477

RESUMEN

The broad diversity of cell types within vertebrates arises from a unique genetic blueprint by combining intrinsic cellular information with developmental and other extrinsic signals. Lying at the interface between cellular signals and the DNA is the chromatin, a dynamic nucleoprotein complex that helps to mediate gene regulation. The most basic subunit of chromatin, the nucleosome, consists of DNA wrapped around histones, a set of proteins that play crucial roles as scaffolding molecules and regulators of gene expression. Growing evidence indicates that canonical histones are commonly replaced by protein variants before and during cellular transitions. We highlight exciting new results suggesting that histone variants are essential players in the control of cellular plasticity during development and in the adult nervous system.


Asunto(s)
Plasticidad de la Célula/genética , Variación Genética/fisiología , Histonas/genética , Adulto , Animales , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , Humanos , Proteínas Mutantes/fisiología
12.
Elife ; 4: e07860, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-26140685

RESUMEN

The maternal and paternal genomes play different roles in mammalian brains as a result of genomic imprinting, an epigenetic regulation leading to differential expression of the parental alleles of some genes. Here we investigate genomic imprinting in the cerebellum using a newly developed Bayesian statistical model that provides unprecedented transcript-level resolution. We uncover 160 imprinted transcripts, including 41 novel and independently validated imprinted genes. Strikingly, many genes exhibit parentally biased--rather than monoallelic--expression, with different magnitudes according to age, organ, and brain region. Developmental changes in parental bias and overall gene expression are strongly correlated, suggesting combined roles in regulating gene dosage. Finally, brain-specific deletion of the paternal, but not maternal, allele of the paternally-biased Bcl-x, (Bcl2l1) results in loss of specific neuron types, supporting the functional significance of parental biases. These findings reveal the remarkable complexity of genomic imprinting, with important implications for understanding the normal and diseased brain.


Asunto(s)
Alelos , Cerebelo/fisiología , Perfilación de la Expresión Génica , Impresión Genómica , Animales , Bioestadística , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
13.
Cancer Immunol Res ; 3(1): 68-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25358763

RESUMEN

Aberrant blood vessels enable tumor growth, provide a barrier to immune infiltration, and serve as a source of protumorigenic signals. Targeting tumor blood vessels for destruction, or tumor vascular disruption therapy, can therefore provide significant therapeutic benefit. Here, we describe the ability of chimeric antigen receptor (CAR)-bearing T cells to recognize human prostate-specific membrane antigen (hPSMA) on endothelial targets in vitro as well as in vivo. CAR T cells were generated using the anti-PSMA scFv, J591, and the intracellular signaling domains: CD3ζ, CD28, and/or CD137/4-1BB. We found that all anti-hPSMA CAR T cells recognized and eliminated PSMA(+) endothelial targets in vitro, regardless of the signaling domain. T cells bearing the third-generation anti-hPSMA CAR, P28BBζ, were able to recognize and kill primary human endothelial cells isolated from gynecologic cancers. In addition, the P28BBζ CAR T cells mediated regression of hPSMA-expressing vascular neoplasms in mice. Finally, in murine models of ovarian cancers populated by murine vessels expressing hPSMA, the P28BBζ CAR T cells were able to ablate PSMA(+) vessels, cause secondary depletion of tumor cells, and reduce tumor burden. Taken together, these results provide a strong rationale for the use of CAR T cells as agents of tumor vascular disruption, specifically those targeting PSMA. Cancer Immunol Res; 3(1); 68-84. ©2014 AACR.


Asunto(s)
Antígenos de Superficie/inmunología , Células Endoteliales/inmunología , Glutamato Carboxipeptidasa II/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Anticuerpos de Cadena Única/inmunología , Neoplasias Vasculares/terapia , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Neoplasias Ováricas/patología , Transducción de Señal , Transducción Genética , Carga Tumoral
14.
Nat Med ; 20(6): 607-15, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24793239

RESUMEN

We describe a new mechanism regulating the tumor endothelial barrier and T cell infiltration into tumors. We detected selective expression of the death mediator Fas ligand (FasL, also called CD95L) in the vasculature of human and mouse solid tumors but not in normal vasculature. In these tumors, FasL expression was associated with scarce CD8(+) infiltration and a predominance of FoxP3(+) T regulatory (Treg) cells. Tumor-derived vascular endothelial growth factor A (VEGF-A), interleukin 10 (IL-10) and prostaglandin E2 (PGE2) cooperatively induced FasL expression in endothelial cells, which acquired the ability to kill effector CD8(+) T cells but not Treg cells because of higher levels of c-FLIP expression in Treg cells. In mice, genetic or pharmacologic suppression of FasL produced a substantial increase in the influx of tumor-rejecting CD8(+) over FoxP3(+) T cells. Pharmacologic inhibition of VEGF and PGE2 produced a marked increase in the influx of tumor-rejecting CD8(+) over FoxP3(+) T cells that was dependent on attenuation of FasL expression and led to CD8-dependent tumor growth suppression. Thus, tumor paracrine mechanisms establish a tumor endothelial death barrier, which has a critical role in establishing immune tolerance and determining the fate of tumors.


Asunto(s)
Endotelio Vascular/metabolismo , Proteína Ligando Fas/inmunología , Tolerancia Inmunológica/inmunología , Neoplasias/irrigación sanguínea , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Anticuerpos/administración & dosificación , Apoptosis/inmunología , Western Blotting , Proteína Ligando Fas/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Humanos , Inmunohistoquímica , Células Jurkat , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Análisis por Micromatrices , Neoplasias/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
15.
Hum Gene Ther ; 24(3): 295-305, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23297870

RESUMEN

NKG2D ligands (NKG2DLs) are widely expressed on ovarian cancers to various degrees, making them attractive targets for immunotherapy. Here, we applied a chimeric antigen receptor (CAR) approach for the targeting of NKG2DLs expressed on human ovarian cancer cells and evaluated the impact of pharmacological upregulation of NKG2DLs on immune recognition. Various NKG2DLs, including MICA/B and ULBP-1, -2, -3, and -4, were expressed at various levels on the surface of all established ovarian cancer cell lines and primary ovarian cancer samples tested. To redirect human T cells against NKG2DLs, an NKG2DL-specific CAR was generated by fusing the extracellular domain of the NKG2D receptor to the 4-1BB costimulatory and CD3-ζ chain signaling domains. In vitro expansion of chimeric NKG2D CAR T cells was delayed compared with untransduced T cells and control CAR T cells; the likely result of fratricide among activated T cells expressing NKG2DLs. However, NKG2D CAR T cells did expand and were selectively enriched during prolonged culture. In coculture, CD4(+) and CD8(+) NKG2D CAR T cells specifically recognized and killed NKG2DL-expressing ovarian cancer cell lines but not NKG2DL-negative cells. Notably, pretreatment of ovarian cancer cells expressing moderate to low levels of NKG2DLs with the histone deacetylase inhibitor sodium valproate (VPA) upregulated NKG2DL cell surface expression and consequently enhanced their immune recognition by chimeric NKG2D CAR T cells. Our results demonstrate that VPA-induced upregulation of NKG2DL expression enhances the immune recognition of ovarian cancer cells by engineered NKG2D CAR T cells, and rationalizes the use of VPA in combination with NKG2DL-targeted immunotherapy in ovarian cancer.


Asunto(s)
Histona Desacetilasas/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Línea Celular , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Orden Génico , Vectores Genéticos/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Inmunofenotipificación , Lentivirus/genética , Ligandos , Activación de Linfocitos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias Ováricas/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Ácido Valproico/farmacología
16.
Elife ; 1: e00070, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23240083

RESUMEN

We have identified a replication-independent histone variant, Hist2h2be (referred to herein as H2be), which is expressed exclusively by olfactory chemosensory neurons. Levels of H2BE are heterogeneous among olfactory neurons, but stereotyped according to the identity of the co-expressed olfactory receptor (OR). Gain- and loss-of-function experiments demonstrate that changes in H2be expression affect olfactory function and OR representation in the adult olfactory epithelium. We show that H2BE expression is reduced by sensory activity and that it promotes neuronal cell death, such that inactive olfactory neurons display higher levels of the variant and shorter life spans. Post-translational modifications (PTMs) of H2BE differ from those of the canonical H2B, consistent with a role for H2BE in altering transcription. We propose a physiological function for H2be in modulating olfactory neuron population dynamics to adapt the OR repertoire to the environment.DOI:http://dx.doi.org/10.7554/eLife.00070.001.


Asunto(s)
Histonas/genética , Mucosa Olfatoria/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Procesamiento Proteico-Postraduccional , Olfato/genética , Secuencia de Aminoácidos , Animales , Muerte Celular/genética , Expresión Génica , Células HEK293 , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Odorantes/análisis , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Mucosa Olfatoria/citología , Neuronas Receptoras Olfatorias/citología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
17.
Oncoimmunology ; 1(4): 575-577, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22754792

RESUMEN

We recently described hypoxia as one of the leading mechanism for the recruitment of regulatory T cells (Treg) through CCL28 chemokine in ovarian cancer. Treg promote progression of cancer through tumor-specific immune paralysis but also reprogramming of angiogenesis. We review these mechanisms and discuss the challenges and opportunities for therapy targeting Treg.

19.
Bioorg Med Chem ; 14(9): 3081-9, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16412655

RESUMEN

Previous reports have demonstrated that new Cre recombinase specificities can be developed for symmetrically designed lox mutants through directed evolution. The development of Cre variants that allow the recombination of true asymmetric lox mutant sites has not yet been addressed, however. In the present study, we demonstrate that a mixture of two different site-specific Cre recombinase molecules (wt Cre and a mutant Cre) catalyzes efficient recombination between two asymmetric lox sites in vitro, presumably via formation of a functionally active heterotetrameric complex. The results may broaden the application of site-specific recombination in basic and applied research, including the custom-design of recombinases for natural, asymmetric, and lox-related target sequences present in the genome. Future applications may potentially include genomic manipulations, for example, site-specific integrations, deletions or substitutions within precise regions of the genomes of mammalians and other organisms.


Asunto(s)
ADN Nucleotidiltransferasas/genética , ADN Nucleotidiltransferasas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Animales , Secuencia de Bases , Simulación por Computador , Humanos , Ratones , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica , Ratas , Especificidad por Sustrato
20.
Proc Natl Acad Sci U S A ; 101(20): 7566-71, 2004 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15138302

RESUMEN

With few exceptions the genetic codes of all known organisms encode the same 20 amino acids, yet all that is required to add a new building block are a unique tRNA/aminoacyl-tRNA synthetase pair, a source of the amino acid, and a unique codon that specifies the amino acid. For example, the amber nonsense codon, TAG, together with orthogonal Methanococcus jannaschii or Escherichia coli tRNA/synthetase pairs have been used to genetically encode a variety of unnatural amino acids in E. coli and yeast, respectively. However, the availability of noncoding triplet codons ultimately limits the number of amino acids encoded by any organism. Here, we report the design and generation of an orthogonal synthetase/tRNA pair derived from archaeal tRNA(Lys) sequences that efficiently and selectively incorporates an unnatural amino acid into proteins in response to the quadruplet codon, AGGA. Frameshift suppression with L-homoglutamine (hGln) does not significantly affect protein yields or cell growth rates and is mutually orthogonal with amber suppression, permitting the simultaneous incorporation of two unnatural amino acids, hGln and O-methyl-L-tyrosine, at distinct positions within myoglobin. This work suggests that neither the number of available triplet codons nor the translational machinery itself represents a significant barrier to further expansion of the genetic code.


Asunto(s)
Aminoácidos/metabolismo , Aminoacil-ARNt Sintetasas/metabolismo , Codón , Código Genético , Pyrococcus horikoshii/genética , Aminoácidos/genética , Aminoacil-ARNt Sintetasas/genética , Evolución Molecular , Lisina-ARNt Ligasa/metabolismo , Pyrococcus horikoshii/enzimología , ARN de Transferencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...