Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Ther ; 32(6): 1895-1916, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38549376

RESUMEN

Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.


Asunto(s)
Antígeno B7-H1 , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción STAT3 , Microambiente Tumoral , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Animales , Ratones , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Humanos , Microambiente Tumoral/inmunología , ARN Interferente Pequeño/genética , Inmunoterapia/métodos , Resistencia a Antineoplásicos/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/genética
2.
Nature ; 470(7335): 492-7, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21350482

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is known to broadly regulate the cellular stress response. In contrast, it is unclear if the PACAP-PAC1 receptor pathway has a role in human psychological stress responses, such as post-traumatic stress disorder (PTSD). Here we find, in heavily traumatized subjects, a sex-specific association of PACAP blood levels with fear physiology, PTSD diagnosis and symptoms in females. We examined 44 single nucleotide polymorphisms (SNPs) spanning the PACAP (encoded by ADCYAP1) and PAC1 (encoded by ADCYAP1R1) genes, demonstrating a sex-specific association with PTSD. A single SNP in a putative oestrogen response element within ADCYAP1R1, rs2267735, predicts PTSD diagnosis and symptoms in females only. This SNP also associates with fear discrimination and with ADCYAP1R1 messenger RNA expression in human brain. Methylation of ADCYAP1R1 in peripheral blood is also associated with PTSD. Complementing these human data, ADCYAP1R1 mRNA is induced with fear conditioning or oestrogen replacement in rodent models. These data suggest that perturbations in the PACAP-PAC1 pathway are involved in abnormal stress responses underlying PTSD. These sex-specific effects may occur via oestrogen regulation of ADCYAP1R1. PACAP levels and ADCYAP1R1 SNPs may serve as useful biomarkers to further our mechanistic understanding of PTSD.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/sangre , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Trastornos por Estrés Postraumático/sangre , Trastornos por Estrés Postraumático/genética , Amígdala del Cerebelo/metabolismo , Animales , Condicionamiento Clásico/fisiología , Islas de CpG/genética , Metilación de ADN , Estrógenos/metabolismo , Estrógenos/farmacología , Miedo/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Estudios de Asociación Genética , Humanos , Masculino , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/química , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Elementos de Respuesta/genética , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Caracteres Sexuales , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/psicología
3.
J Neurosci ; 32(13): 4651-9, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22457511

RESUMEN

The consolidation of conditioned fear involves upregulation of genes necessary for long-term memory formation. An important question remains as to whether this results in part from epigenetic regulation and chromatin modulation. We examined whether Homer1a, which is required for memory formation, is necessary for Pavlovian cued fear conditioning, whether it is downstream of BDNF-TrkB activation, and whether this pathway utilizes histone modifications for activity-dependent transcriptional regulation. We initially found that Homer1a knock-out mice exhibited deficits in cued fear conditioning (5 tone-shock presentations with 70 dB, 6 kHz tones and 0.5 s, 0.6 mA footshocks). We then demonstrated that: (1) Homer1a mRNA increases after fear conditioning in vivo within both amygdala and hippocampus of wild-type mice; (2) it increases after BDNF application to primary hippocampal and amygdala cultures in vitro; and (3) these increases are dependent on transcription and MAPK signaling. Furthermore, using chromatin immunoprecipitation we found that both in vitro and in vivo manipulations result in decreases in Homer1 promoter H3K9 methylation in amygdala cells but increases in Homer1 promoter H3 acetylation in hippocampal cells. However, no changes were observed in H4 acetylation or H3K27 dimethylation. Inhibition of histone deacetylation by sodium butyrate enhanced contextual but not cued fear conditioning and enhanced Homer1 H3 acetylation in the hippocampus. These data provide evidence for dynamic epigenetic regulation of Homer1a following BDNF-induced plasticity and during a BDNF-dependent learning process. Furthermore, upregulation of this gene may be regulated through distinct epigenetic modifications in the hippocampus and amygdala.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Proteínas Portadoras/biosíntesis , Condicionamiento Clásico/fisiología , Epigénesis Genética/fisiología , Miedo/fisiología , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteínas Portadoras/genética , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Miedo/psicología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Proteínas de Andamiaje Homer , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/fisiología , Cultivo Primario de Células , Regiones Promotoras Genéticas/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transcripción Genética/fisiología
4.
Cell Rep ; 35(6): 109112, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979622

RESUMEN

Receptor interacting protein kinase 1 (RIPK1) mediates cell death and inflammatory signaling and is increased in multiple sclerosis (MS) brain samples. Here, we investigate the role of glial RIPK1 kinase activity in mediating MS pathogenesis. We demonstrate RIPK1 levels correlate with MS disease progression. We find microglia are susceptible to RIPK1-mediated cell death and identify an inflammatory gene signature that may contribute to the neuroinflammatory milieu in MS patients. We uncover a distinct role for RIPK1 in astrocytes in regulating inflammatory signaling in the absence of cell death and confirm RIPK1-kinase-dependent regulation in human glia. Using a murine MS model, we show RIPK1 inhibition attenuates disease progression and suppresses deleterious signaling in astrocytes and microglia. Our results suggest RIPK1 kinase activation in microglia and astrocytes induces a detrimental neuroinflammatory program that contributes to the neurodegenerative environment in progressive MS.


Asunto(s)
Microglía/metabolismo , Esclerosis Múltiple/genética , Enfermedades Neuroinflamatorias/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Esclerosis Múltiple/patología , Transducción de Señal
5.
Cell Death Dis ; 11(10): 904, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-33097690

RESUMEN

Microglia serve as the innate immune cells of the central nervous system (CNS) by providing continuous surveillance of the CNS microenvironment and initiating defense mechanisms to protect CNS tissue. Upon injury, microglia transition into an activated state altering their transcriptional profile, transforming their morphology, and producing pro-inflammatory cytokines. These activated microglia initially serve a beneficial role, but their continued activation drives neuroinflammation and neurodegeneration. Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the CNS, and activated microglia and macrophages play a significant role in mediating disease pathophysiology and progression. Colony-stimulating factor-1 receptor (CSF1R) and its ligand CSF1 are elevated in CNS tissue derived from MS patients. We performed a large-scale RNA-sequencing experiment and identified CSF1R as a key node of disease progression in a mouse model of progressive MS. We hypothesized that modulating microglia and infiltrating macrophages through the inhibition of CSF1R will attenuate deleterious CNS inflammation and reduce subsequent demyelination and neurodegeneration. To test this hypothesis, we generated a novel potent and selective small-molecule CSF1R inhibitor (sCSF1Rinh) for preclinical testing. sCSF1Rinh blocked receptor phosphorylation and downstream signaling in both microglia and macrophages and altered cellular functions including proliferation, survival, and cytokine production. In vivo, CSF1R inhibition with sCSF1Rinh attenuated neuroinflammation and reduced microglial proliferation in a murine acute LPS model. Furthermore, the sCSF1Rinh attenuated a disease-associated microglial phenotype and blocked both axonal damage and neurological impairments in an experimental autoimmune encephalomyelitis (EAE) model of MS. While previous studies have focused on microglial depletion following CSF1R inhibition, our data clearly show that signaling downstream of this receptor can be beneficially modulated in the context of CNS injury. Together, these data suggest that CSF1R inhibition can reduce deleterious microglial proliferation and modulate microglial phenotypes during neuroinflammatory pathogenesis, particularly in progressive MS.


Asunto(s)
Inflamación/patología , Esclerosis Múltiple/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Humanos , Macrófagos/efectos de los fármacos , Ratones , Microglía/patología , Esclerosis Múltiple/patología , Transducción de Señal/efectos de los fármacos
6.
Acta Psychol (Amst) ; 127(3): 636-44, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18164272

RESUMEN

Using the think/no-think paradigm, we examined the effect of a meaningful connection between emotionally neutral cues and targets on initial learning and later recall by students in dysphoric or nondysphoric mood states. Compared to meaningfully connected cue-target pairs, unrelated pairs were less easily learned and more easily forgotten, even when initial learning was controlled. Depressive deficits were obtained in initial learning (only marginally) and final recall. When examined separately within each cuing condition, the recall deficit associated with depressed mood was restricted to the unrelated condition, but when initial learning differences were controlled this deficit was only marginally significant. Results are discussed in relation to other recent findings concerning depression-related performance in this paradigm and to findings of depression and memory more generally.


Asunto(s)
Señales (Psicología) , Trastorno Depresivo/psicología , Aprendizaje , Trastornos de la Memoria/psicología , Afecto , Femenino , Humanos , Masculino , Memoria , Recuerdo Mental , Estudiantes/psicología , Análisis y Desempeño de Tareas
7.
Endocrinology ; 148(10): 4927-36, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17595229

RESUMEN

The G protein-coupled receptor Gpr54 and its ligand metastin (derived from the Kiss1 gene product kisspeptin) are key gatekeepers of sexual maturation. Gpr54 knockout mice demonstrate hypogonadotropic hypogonadism, but until recently, the phenotype of Kiss1 knockout mice was unknown. This report describes the reproductive phenotypes of mice carrying targeted deletions of Kiss1 or Gpr54 on the same genetic background. Both Kiss1 and Gpr54 knockout mice are viable but infertile and have abnormal sexual maturation; the majority of males lack preputial separation, and females have delayed vaginal opening and absence of estrous cycling. Kiss1 and Gpr54 knockout males have significantly smaller testes compared with controls. Gpr54 knockout females have smaller ovaries and uteri than wild-type females. However, Kiss1 knockout females demonstrate two distinct phenotypes: half have markedly reduced gonadal weights similar to those of Gpr54 knockout mice, whereas half exhibit persistent vaginal cornification and have gonadal weights comparable with those of wild-type females. FSH levels in both Kiss1 and Gpr54 knockout males and females are significantly lower than in controls. When injected with mouse metastin 43-52, a Gpr54 agonist, Gpr54 knockout mice fail to increase gonadotropins, whereas Kiss1 knockout mice respond with increased gonadotropin levels. In summary, both Kiss1 and Gpr54 knockout mice have abnormal sexual maturation consistent with hypogonadotropic hypogonadism, although Kiss1 knockout mice appear to be less severely affected than their receptor counterparts. Kiss1 knockout females demonstrate a bimodal phenotypic variability, with some animals having higher gonadal weight, larger vaginal opening, and persistent vaginal cornification.


Asunto(s)
Hipogonadismo/etiología , Hipogonadismo/patología , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Animales , Femenino , Gonadotropinas/sangre , Hipogonadismo/complicaciones , Hipogonadismo/fisiopatología , Infertilidad/etiología , Péptidos y Proteínas de Señalización Intracelular , Kisspeptinas , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Ovario/patología , Fragmentos de Péptidos/farmacología , Fenotipo , Proteínas Serina-Treonina Quinasas , Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Maduración Sexual , Espermatozoides/fisiología , Testículo/patología , Testosterona/sangre
8.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(2): 482-95, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17188790

RESUMEN

We have previously shown that acute stress-induced release of norepinephrine (NE) facilitates anxiety-like behavioral responses to stress, such as reduction in open-arm exploration on the elevated-plus maze and in social behavior on the social interaction test. Since these responses represent inhibition of ongoing behavior, it is important to also address whether NE facilitates a response that represents an activation of behavior. Correspondingly, it is unknown how a chronic elevation in tonic steady-state noradrenergic (NA) neurotransmission induced by NE reuptake blockade might alter this acute modulatory function, a regulatory process that may be pertinent to the anxiolytic effects of NE reuptake blockers such as desipramine (DMI). Therefore, in this study, we investigated noradrenergic modulation of the shock-probe defensive burying response in the lateral septum (LS). In experiment 1, shock-probe exposure induced an acute 3-fold increase in NE levels measured in LS of male Sprague-Dawley rats by microdialysis. Shock-probe exposure also induced a modest rise in plasma ACTH, taken as an indicator of perceived stress, that returned to baseline more rapidly in rats that were allowed to bury the probe compared to rats prevented from burying by providing them with minimal bedding, indicating that the active defensive burying behavior is an effective coping strategy that reduces the impact of acute shock probe-induced stress. In experiment 2, blockade of either alpha(1)- or beta-adrenergic receptors in LS by local antagonist microinjection immediately before testing reduced defensive burying and increased immobility. In the next experiment, chronic DMI treatment increased basal extracellular NE levels in LS, and attenuated the acute shock probe-induced increase in NE release in LS relative to baseline. Chronic DMI treatment decreased shock-probe defensive burying behavior in a time-dependent manner, apparent only after 2 weeks or more of drug treatment. Moreover, rats treated chronically with DMI showed no significant rise of plasma ACTH in response to shock-probe exposure. Thus, acute stress-induced release of NE in LS facilitated defensive burying, an active, adaptive behavioral coping response. Chronic treatment with the NE reuptake blocker and antidepressant drug DMI attenuated acute noradrenergic facilitation of the active burying response, and also attenuated the level of perceived stress driving that response. These results suggest that long-term regulation of the acute modulatory function of NE by chronic treatment with reuptake blockers may contribute to the mechanisms by which such drugs exert their anxiolytic effects in the treatment of stress-related psychiatric conditions, including depression and anxiety.


Asunto(s)
Inhibidores de Captación Adrenérgica/administración & dosificación , Ansiedad/tratamiento farmacológico , Desipramina/administración & dosificación , Pérdida de Tono Postural/efectos de los fármacos , Norepinefrina/metabolismo , Tabique del Cerebro/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Ansiedad/sangre , Ansiedad/etiología , Conducta Animal/efectos de los fármacos , Mecanismos de Defensa , Modelos Animales de Enfermedad , Electrochoque/efectos adversos , Masculino , Ratas , Ratas Sprague-Dawley , Tabique del Cerebro/efectos de los fármacos , Factores de Tiempo
9.
Trends Neurosci ; 35(1): 24-35, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21798604

RESUMEN

Posttraumatic stress disorder (PTSD) is an anxiety disorder that can develop after a traumatic experience such as domestic violence, natural disasters or combat-related trauma. The cost of such disorders on society and the individual can be tremendous. In this article, we review how the neural circuitry implicated in PTSD in humans is related to the neural circuitry of fear. We then discuss how fear conditioning is a suitable model for studying the molecular mechanisms of the fear components that underlie PTSD, and the biology of fear conditioning with a particular focus on the brain-derived neurotrophic factor (BDNF)-tyrosine kinase B (TrkB), GABAergic and glutamatergic ligand-receptor systems. We then summarize how such approaches might help to inform our understanding of PTSD and other stress-related disorders and provide insight to new pharmacological avenues of treatment of PTSD.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Plasticidad Neuronal/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Animales , Encéfalo/anatomía & histología , Encéfalo/fisiología , Encéfalo/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Transducción de Señal/fisiología
10.
Proc Natl Acad Sci U S A ; 104(4): 1348-53, 2007 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-17229845

RESUMEN

Activin and myostatin are related members of the TGF-beta growth factor superfamily. FSTL3 (Follistatin-like 3) is an activin and myostatin antagonist whose physiological role in adults remains to be determined. We found that homozygous FSTL3 knockout adults developed a distinct group of metabolic phenotypes, including increased pancreatic islet number and size, beta cell hyperplasia, decreased visceral fat mass, improved glucose tolerance, and enhanced insulin sensitivity, changes that might benefit obese, insulin-resistant patients. The mice also developed hepatic steatosis and mild hypertension but exhibited no alteration of muscle or body weight. This combination of phenotypes appears to arise from increased activin and myostatin bioactivity in specific tissues resulting from the absence of the FSTL3 antagonist. Thus, the enlarged islets and beta cell number likely result from increased activin action. Reduced visceral fat is consistent with a role for increased myostatin action in regulating fat deposition, which, in turn, may be partly responsible for the enhanced glucose tolerance and insulin sensitivity. Our results demonstrate that FSTL3 regulation of activin and myostatin is critical for normal adult metabolic homeostasis, suggesting that pharmacological manipulation of FSTL3 activity might simultaneously reduce visceral adiposity, increase beta cell mass, and improve insulin sensitivity.


Asunto(s)
Proteínas Relacionadas con la Folistatina/fisiología , Glucosa/metabolismo , Homeostasis , Factor de Crecimiento Transformador beta/metabolismo , Animales , Composición Corporal , Peso Corporal , Femenino , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Humanos , Hipertensión/genética , Islotes Pancreáticos/metabolismo , Ligandos , Hígado/fisiología , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA