Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain Behav Immun ; 115: 308-318, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37914098

RESUMEN

Maternal stress during pregnancy is prevalent and associated with increased risk of neurodevelopmental disorders in the offspring. Maternal and offspring immune dysfunction has been implicated as a potential mechanism by which prenatal stress shapes offspring neurodevelopment; however, the impact of prenatal stress on the developing immune system has yet to be elucidated. Furthermore, there is evidence that the chemokine C-C motif chemokine ligand 2 (CCL2) plays a key role in mediating the behavioral sequelae of prenatal stress. Here, we use an established model of prenatal restraint stress in mice to investigate alterations in the fetal immune system, with a focus on CCL2. In the placenta, stress led to a reduction in CCL2 and Ccr2 expression with a concomitant decrease in leukocyte number. However, the fetal liver exhibited an inflammatory phenotype, with upregulation of Ccl2, Il6, and Lbp expression, along with an increase in pro-inflammatory Ly6CHi monocytes. Prenatal stress also disrupted chemokine signaling and increased the number of monocytes and microglia in the fetal brain. Furthermore, stress increased Il1b expression by fetal brain CD11b+ microglia and monocytes. Finally, intra-amniotic injections of recombinant mouse CCL2 partially recapitulated the social behavioral deficits in the adult offspring previously observed in the prenatal restraint stress model. Altogether, these data suggest that prenatal stress led to fetal inflammation, and that fetal CCL2 plays a role in shaping offspring social behavior.


Asunto(s)
Quimiocina CCL2 , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Humanos , Ratones , Embarazo , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Inflamación/metabolismo , Ligandos , Monocitos/metabolismo , Conducta Social
2.
Brain Behav Immun ; 96: 28-39, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33989741

RESUMEN

Maternal infection during pregnancy is a known risk factor for offspring mental health disorders. Animal models of maternal immune activation (MIA) have implicated specific cellular and molecular etiologies of psychiatric illness, but most rely on pathogen mimetics. Here, we developed a mouse model of live H3N2 influenza A virus (IAV) infection during pregnancy that induces a robust inflammatory response but is sublethal to both dams and offspring. We observed classic indicators of lung inflammation and severely diminished weight gain in IAV-infected dams. This was accompanied by immune cell infiltration in the placenta and partial breakdown of placental integrity. However, indications of fetal neuroinflammation were absent. Further hallmarks of mimetic-induced MIA, including enhanced circulating maternal IL-17A, were also absent. Respiratory IAV infection did result in an upregulation in intestinal expression of transcription factor RORγt, master regulator of a subset of T lymphocytes, TH17 cells, which are heavily implicated in MIA-induced etiologies. Nonetheless, subsequent augmentation in IL-17A production and concomitant overt intestinal injury was not evident. Our results suggest that mild or moderately pathogenic IAV infection during pregnancy does not inflame the developing fetal brain, and highlight the importance of live pathogen infection models for the study of MIA.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Animales , Encéfalo , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A , Ratones , Placenta , Embarazo
3.
Learn Mem ; 24(9): 385-391, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28814463

RESUMEN

Retrieval of fear memories can be state-dependent, meaning that they are best retrieved if the brain states at encoding and retrieval are similar. Such states can be induced by activating extrasynaptic γ-aminobutyric acid type A receptors (GABAAR) with the broad α-subunit activator gaboxadol. However, the circuit mechanisms and specific subunits underlying gaboxadol's effects are not well understood. Here we show that gaboxadol induces profound changes of local and network oscillatory activity, indicative of discoordinated hippocampal-cortical activity, that were accompanied by robust and long-lasting state-dependent conditioned fear. Episodic memories typically are hippocampus-dependent for a limited period after learning, but become cortex-dependent with the passage of time. In contrast, state-dependent memories continued to rely on hippocampal GABAergic mechanisms for memory retrieval. Pharmacological approaches with α-subunit-specific agonists targeting the hippocampus implicated the prototypic extrasynaptic subunits (α4) as the mediator of state-dependent conditioned fear. Together, our findings suggest that continued dependence on hippocampal rather than cortical mechanisms could be an important feature of state-dependent memories that contributes to their conditional retrieval.


Asunto(s)
Condicionamiento Clásico/fisiología , Miedo/fisiología , Recuerdo Mental/fisiología , Animales , Condicionamiento Clásico/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Reacción Cataléptica de Congelación/efectos de los fármacos , Agonistas del GABA/farmacología , Hipocampo/efectos de los fármacos , Masculino , Recuerdo Mental/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
4.
Learn Mem ; 23(11): 631-638, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27918283

RESUMEN

Understanding how episodic memories are formed and retrieved is necessary if we are to treat disorders in which they malfunction. Muscarinic acetylcholine receptors (mAChR) in the hippocampus and cortex underlie memory formation, but there is conflicting evidence regarding their role in memory retrieval. Additionally, there is no consensus on which mAChR subtypes are critical for memory processing. Using pharmacological and genetic approaches, we found that (1) encoding and retrieval of contextual memory requires mAChR in the dorsal hippocampus (DH) and retrosplenial cortex (RSC), (2) memory formation requires hippocampal M3 and cooperative activity of RSC M1 and M3, and (3) memory retrieval is more impaired by inactivation of multiple M1-M4 mAChR in DH or RSC than inactivation of individual receptor subtypes. Contrary to the view that acetylcholine supports learning but is detrimental to memory retrieval, we found that coactivation of multiple mAChR is required for retrieval of both recently and remotely acquired context memories. Manipulations with higher receptor specificity were generally less potent than manipulations targeting multiple receptor subtypes, suggesting that mAChR act in synergy to regulate memory processes. These findings provide unique insight into the development of therapies for amnestic symptoms, suggesting that broadly acting, rather than receptor-specific, mAchR agonists and positive allosteric modulators may be the most effective therapeutic approach.


Asunto(s)
Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Aprendizaje/fisiología , Memoria/fisiología , Receptores Muscarínicos/metabolismo , Animales , Catéteres de Permanencia , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Dependovirus/genética , Miedo/efectos de los fármacos , Miedo/fisiología , Técnicas de Inactivación de Genes , Vectores Genéticos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Inmunohistoquímica , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Antagonistas Muscarínicos/farmacología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Receptores Muscarínicos/genética , Escopolamina/farmacología
5.
Biol Psychiatry ; 95(4): 300-309, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38042328

RESUMEN

Stress and psychiatric disorders have been independently associated with disruption of the maternal and offspring microbiome and with increased risk of the offspring developing psychiatric disorders, both in clinical studies and in preclinical studies. However, the role of the microbiome in mediating the effect of prenatal stress on offspring behavior is unclear. While preclinical studies have identified several key mechanisms, clinical studies focusing on mechanisms are limited. In this review, we discuss 3 specific mechanisms by which the microbiome could mediate the effects of prenatal stress: 1) altered production of short-chain fatty acids; 2) disruptions in TH17 (T helper 17) cell differentiation, leading to maternal and fetal immune activation; and 3) perturbation of intestinal and microbial tryptophan metabolism and serotonergic signaling. Finally, we review the existing clinical literature focusing on these mechanisms and highlight the need for additional mechanistic clinical research to better understand the role of the microbiome in the context of prenatal stress.


Asunto(s)
Trastornos Mentales , Microbiota , Efectos Tardíos de la Exposición Prenatal , Embarazo , Femenino , Humanos , Trastornos Mentales/etiología
6.
Brain Behav Immun Health ; 36: 100730, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38323225

RESUMEN

Background: Psychosocial stress and mood-related disorders, such as depression, are prevalent and vulnerability to these conditions is heightened during pregnancy. Psychosocial stress induces consequences via several mechanisms including the gut microbiota-brain axis and associated signaling pathways. Previous preclinical work indicates that prenatal stress alters maternal gut microbial composition and impairs offspring development. Importantly, although the fecal and vaginal microenvironments undergo alterations across pregnancy, we lack consensus regarding which shifts are adaptive or maladaptive in the presence of prenatal stress and depression. Clinical studies interrogating these relationships have identified unique taxa but have been limited in study design. Methods: We conducted a prospective cohort study of pregnant individuals consisting of repeated administration of psychometrics (Perceived Stress Scale (PSS) and Center for Epidemiological Studies Depression Scale (CES-D)) and collection of fecal and vaginal microbiome samples. Fecal and vaginal microbial community composition across psychometric responses were interrogated using full-length 16S rRNA sequencing followed by α and ß-diversity metrics and taxonomic abundance. Results: Early pregnancy stress was associated with increased abundance of fecal taxa not previously identified in related studies, and stress from late pregnancy through postpartum was associated with increased abundance of typical vaginal taxa and opportunistic pathogens in the fecal microenvironment. Additionally, in late pregnancy, maternal stress and depression scores were associated with each other and with elevated maternal C-C motif chemokine ligand 2 (CCL2) concentrations. At delivery, concordant with previous literature, umbilical CCL2 concentration was negatively correlated with relative abundance of maternal fecal Lactobacilli. Lastly, participants with more severe depressive symptoms experienced steeper decreases in prenatal vaginal α-diversity. Conclusion: These findings a) underscore previous preclinical and clinical research demonstrating the effects of prenatal stress on maternal microbiome composition, b) suggest distinct biological pathways for the consequences of stress versus depression and c) extend the literature by identifying several taxa which may serve critical roles in mediating this relationship. Thus, further interrogation of the role of specific maternal microbial taxa in relation to psychosocial stress and its sequelae is warranted.

7.
Neurobiol Stress ; 21: 100480, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36532381

RESUMEN

Psychosocial stress is prevalent during pregnancy, and is associated with immune dysfunction, both for the mother and the child. The gut microbiome has been implicated as a potential mechanism by which stress during pregnancy can impact both maternal and offspring immune function; however, the complex interplay between the gut microbiome and the immune system is not well-understood. Here, we leverage a model of antimicrobial-mediated gut microbiome reduction, in combination with a well-established model of maternal restraint stress, to investigate the independent effects of and interaction between maternal stress and the gut microbiome in shaping maternal and offspring immunity. First, we confirmed that the antimicrobial treatment reduced maternal gut bacterial load and altered fecal alpha and beta diversity, with a reduction in commensal microbes and an increase in the relative abundance of rare taxa. Prenatal stress also disrupted the gut microbiome, according to measures of both alpha and beta diversity. Furthermore, prenatal stress and antimicrobials independently induced systemic and gastrointestinal immune suppression in the dam with a concomitant increase in circulating corticosterone. While stress increased neutrophils in the maternal circulation, lymphoid cells and monocytes were not impacted by either stress or antimicrobial treatment. Although the fetal immune compartment was largely spared, stress increased circulating neutrophils and CD8 T cells, and antibiotics increased neutrophils and reduced T cells in the adult offspring. Altogether, these data indicate similar, but discrete, roles for maternal stress and gut microbes in influencing maternal and offspring immune function.

8.
Behav Brain Res ; 414: 113471, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34280459

RESUMEN

The aromatic amino acid tryptophan (Trp) is a precursor for multiple metabolites that can steer proper immune and neurodevelopment as well as social behavior in later life. Dysregulation in the Trp metabolic pathways and abundance of Trp or its derivatives, including indoles, kynurenine (Kyn), and particularly serotonin, has been associated with behavioral deficits and neuropsychiatric disorders including autism spectrum disorders (ASD) and schizophrenia. Previously, we have shown that prenatal stress (PNS) alters placental Trp and serotonin, and reduces Trp-metabolizing members of the maternal colonic microbiota. Given that PNS also results in alterations in offspring neurodevelopment, behavior and immune function, we hypothesized that PNS affects Trp metabolism and transport in both the maternal and fetal compartments, and that these alterations continue into adolescence. We surmised that this is due to reductions in Trp-metabolizing microbes that would otherwise reduce the Trp pool under normal metabolic conditions. To test this, pregnant mice were exposed to a restraint stressor and gene expression of enzymes involved in Trp and serotonin metabolism were measured. Specifically, tryptophan 2,3-dioxygenase, aryl hydrocarbon receptor, and solute carrier proteins, were altered due to PNS both prenatally and postnatally. Additionally, Parasutterella and Bifidobacterium, which metabolize Trp in the gut, were reduced in both the dam and the offspring. Together, the reductions of Trp-associated microbes and concomitant dysregulation in Trp metabolic machinery in dam and offspring suggest that PNS-induced Trp metabolic dysfunction may mediate aberrant fetal neurodevelopment.


Asunto(s)
Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología , Triptófano/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo
9.
Ann Pharmacother ; 44(2): 249-56, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20103610

RESUMEN

BACKGROUND: Hyperglycemia is an important marker for clinical outcomes and mortality in hospitalized patients. New national standards have been established emphasizing the importance of improving inpatient glycemic control in individuals with diabetes or new-onset hyperglycemia. Implementation of these new standards is complex and requires a multidisciplinary team approach. A basal-bolus insulin regimen approach has been shown to improve inpatient glycemic control. Few studies have been published regarding basal-bolus insulin protocol outcomes in the non-intensive care unit (ICU) setting. OBJECTIVE: To evaluate the efficacy of a basal-bolus insulin protocol on inpatient glycemic control in a non-ICU setting, as measured by mean blood glucose and number of hypoglycemic episodes per patient admission. METHODS: A retrospective, observational, single-center study was conducted to compare blood glucose control pre- (October 2006-March 2007) and postprotocol (November 2007-January 2008) implementation. Inclusion criteria consisted of patient admission to a medical or surgical ward for at least 72 hours, with a diagnosis of diabetes, or presentation with 2 blood glucose readings greater than 180 mg/dL. Patients admitted to the ICU or those not admitted to a medical or surgical ward were excluded. RESULTS: Following protocol implementation, the mean +/- SD blood glucose level increased from 174 +/- 88 mg/dL to 188 +/- 95 mg/dL (p < 0.001) and the hypoglycemic incidents significantly decreased, from 1.11 to 0.51 events per patient admission (p < 0.0025). CONCLUSIONS: In this pilot study, implementation of a basal-bolus insulin protocol significantly reduced hypoglycemic events; however, mean blood glucose values increased. These results suggest that a basal-bolus insulin protocol can reduce hypoglycemia; however, factors such as protocol compliance, barriers in overcoming the use of the traditional sliding scale insulin regimens, staff education, and change of work-flow habits can influence the overall efficacy and impact of a basal-bolus insulin protocol on inpatient glycemic control.


Asunto(s)
Glucemia/efectos de los fármacos , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Adhesión a Directriz , Hospitales de Veteranos , Humanos , Hipoglucemia/inducido químicamente , Hipoglucemia/epidemiología , Hipoglucemiantes/efectos adversos , Insulina/efectos adversos , Persona de Mediana Edad , Proyectos Piloto , Guías de Práctica Clínica como Asunto , Estudios Retrospectivos , Resultado del Tratamiento , Estados Unidos , United States Department of Veterans Affairs
10.
Transl Psychiatry ; 10(1): 191, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546752

RESUMEN

Prenatal stress (PNS) is associated with neuropsychiatric disorders in offspring, including anxiety, depression, and autism spectrum disorders. There is mounting evidence that these behavioral phenotypes have origins in utero. Maternal microbes, inflammation, and serotonergic dysfunction have been implicated as potential mediators of the behavioral consequences of PNS; whether and how these systems interact is unclear. Here, we examine the effects of PNS in utero using late-gestation maternal restraint stress in wild-type (WT), germ-free (GF), and CCL2-/- genetic knock-out (KO) mice. In WT mice, PNS leads to placental and fetal brain inflammation, including an elevation in the chemokine CCL2. This inflammation is largely absent in GF mice, indicating the critical role of maternal microbes in mediating immune processes in utero. Furthermore, PNS in the absence of CCL2 failed to increase pro-inflammatory cytokine IL-6 in the fetal brain. PNS offspring also exhibited deficits in sociability and anxiety-like behavior that were absent in CCL2-/- PNS offspring. Tryptophan and serotonin (5-HT) were elevated in the WT PNS placenta, but not in CCL2-/- and GF animals. Altogether, these findings suggest that a complex interaction between maternal microbes, inflammation, and serotonin metabolism regulates the emergence of behavioral abnormalities following PNS.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Animales , Ansiedad , Conducta Animal , Femenino , Inflamación , Ratones , Placenta , Embarazo , Estrés Psicológico/complicaciones
11.
Sci Rep ; 10(1): 20288, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219314

RESUMEN

Maternal stress during pregnancy is widespread and is associated with poor offspring outcomes, including long-term mental health issues. Prenatal stress-induced fetal neuroinflammation is thought to underlie aberrant neurodevelopment and to derive from a disruption in intrauterine immune homeostasis, though the exact origins are incompletely defined. We aimed to identify divergent immune and microbial metagenome profiles of stressed gestating mice that may trigger detrimental inflammatory signaling at the maternal-fetal interface. In response to stress, maternal glucocorticoid circuit activation corresponded with indicators of systemic immunosuppression. At the maternal-fetal interface, density of placental mononuclear leukocytes decreased with stress, yet maternal whole blood leukocyte analysis indicated monocytosis and classical M1 phenotypic shifts. Genome-resolved microbial metagenomic analyses revealed reductions in genes, microbial strains, and metabolic pathways in stressed dams that are primarily associated with pro-inflammatory function. In particular, disrupted Parasutterella excrementihominis appears to be integral to inflammatory and metabolic dysregulation during prenatal stress. Overall, these perturbations in maternal immunological and microbial regulation during pregnancy may displace immune equilibrium at the maternal-fetal interface. Notably, the absence of and reduction in overt maternal inflammation during stress indicates that the signaling patterns driving fetal outcomes in this context are more nuanced and complex than originally anticipated.


Asunto(s)
Encéfalo/embriología , Desarrollo Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Complicaciones del Embarazo/inmunología , Estrés Psicológico/inmunología , Animales , Encéfalo/inmunología , Burkholderiales/genética , Burkholderiales/inmunología , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/genética , Glucocorticoides/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Intercambio Materno-Fetal/inmunología , Salud Mental , Metagenómica , Ratones , Neuroinmunomodulación/inmunología , Placenta/citología , Placenta/inmunología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/psicología , Efectos Tardíos de la Exposición Prenatal/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología
12.
Trends Neurosci ; 42(6): 402-413, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31053242

RESUMEN

The intrauterine environment provides a key interface between the mother and the developing fetus during pregnancy, and is a target for investigating mechanisms of fetal programming. Studies have demonstrated an association between prenatal stress and neurodevelopmental disorders. The role of the intrauterine environment in mediating this effect is still being elucidated. In this review, we discuss emerging preclinical and clinical evidence suggesting the existence of microbial communities in utero. We also outline possible mechanisms of bacterial translocation to the intrauterine environment and immune responses to the presence of microbes or microbial components. Lastly, we overview the effects of intrauterine inflammation on neurodevelopment. We hypothesize that maternal gestational stress leads to disruptions in the maternal oral, gut, and vaginal microbiome that may lead to the translocation of bacteria to the intrauterine environment, eliciting an inflammatory response and resulting in deficits in neurodevelopment.


Asunto(s)
Útero/microbiología , Animales , Femenino , Desarrollo Fetal , Humanos , Microbiota , Trastornos del Neurodesarrollo/etiología , Embarazo
13.
Nat Neurosci ; 18(9): 1265-71, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26280760

RESUMEN

Fear-inducing memories can be state dependent, meaning that they can best be retrieved if the brain states at encoding and retrieval are similar. Restricted access to such memories can present a risk for psychiatric disorders and hamper their treatment. To better understand the mechanisms underlying state-dependent fear, we used a mouse model of contextual fear conditioning. We found that heightened activity of hippocampal extrasynaptic GABAA receptors, believed to impair fear and memory, actually enabled their state-dependent encoding and retrieval. This effect required protein kinase C-ßII and was influenced by miR-33, a microRNA that regulates several GABA-related proteins. In the extended hippocampal circuit, extrasynaptic GABAA receptors promoted subcortical, but impaired cortical, activation during memory encoding of context fear. Moreover, suppression of retrosplenial cortical activity, which normally impairs retrieval, had an enhancing effect on the retrieval of state-dependent fear. These mechanisms can serve as treatment targets for managing access to state-dependent memories of stressful experiences.


Asunto(s)
Condicionamiento Psicológico/fisiología , Miedo/fisiología , Hipocampo/metabolismo , MicroARNs/fisiología , Receptores de GABA-A/metabolismo , Animales , Condicionamiento Psicológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Miedo/efectos de los fármacos , Miedo/psicología , Agonistas del GABA/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Hipocampo/efectos de los fármacos , Isoxazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA