Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Glia ; 72(3): 475-503, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37909340

RESUMEN

Across the globe, approximately one in 10 babies are born preterm, that is, before 37 weeks of a typical 40 weeks of gestation. Up to 50% of preterm born infants develop brain injury, encephalopathy of prematurity (EoP), that substantially increases their risk for developing lifelong defects in motor skills and domains of learning, memory, emotional regulation, and cognition. We are still severely limited in our abilities to prevent or predict preterm birth. No longer just the "support cells," we now clearly understand that during development glia are key for building a healthy brain. Glial dysfunction is a hallmark of EoP, notably, microgliosis, astrogliosis, and oligodendrocyte injury. Our knowledge of glial biology during development is exponentially expanding but hasn't developed sufficiently for development of effective neuroregenerative therapies. This review summarizes the current state of knowledge for the roles of glia in infants with EoP and its animal models, and a description of known glial-cell interactions in the context of EoP, such as the roles for border-associated macrophages. The field of perinatal medicine is relatively small but has worked passionately to improve our understanding of the etiology of EoP coupled with detailed mechanistic studies of pre-clinical and human cohorts. A primary finding from this review is that expanding our collaborations with computational biologists, working together to understand the complexity of glial subtypes, glial maturation, and the impacts of EoP in the short and long term will be key to the design of therapies that improve outcomes.


Asunto(s)
Lesiones Encefálicas , Nacimiento Prematuro , Lactante , Embarazo , Animales , Femenino , Recién Nacido , Humanos , Recien Nacido Prematuro , Neuroglía , Encéfalo
2.
Brain Behav Immun ; 119: 867-877, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750700

RESUMEN

The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.


Asunto(s)
Sistema Nervioso Entérico , Motilidad Gastrointestinal , Inflamación , Lipopolisacáridos , Macrófagos , Animales , Lipopolisacáridos/farmacología , Ratas , Motilidad Gastrointestinal/fisiología , Macrófagos/metabolismo , Inflamación/metabolismo , Inflamación/fisiopatología , Sistema Nervioso Entérico/fisiopatología , Sistema Nervioso Entérico/metabolismo , Masculino , Eje Cerebro-Intestino/fisiología , Colon/metabolismo , Tracto Gastrointestinal/metabolismo , Colitis/fisiopatología , Colitis/metabolismo , Colitis/inducido químicamente , Encéfalo/metabolismo , Ratas Sprague-Dawley , Enfermedades Gastrointestinales/fisiopatología , Enfermedades Gastrointestinales/metabolismo
3.
Int J Mol Sci ; 25(2)2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38255906

RESUMEN

Individuals with autism often experience gastrointestinal issues but the cause is unknown. Many gene mutations that modify neuronal synapse function are associated with autism and therefore may impact the enteric nervous system that regulates gastrointestinal function. A missense mutation in the Nlgn3 gene encoding the cell adhesion protein Neuroligin-3 was identified in two brothers with autism who both experienced severe gastrointestinal dysfunction. Mice expressing this mutation (Nlgn3R451C mice) are a well-studied preclinical model of autism and show autism-relevant characteristics, including impaired social interaction and communication, as well as repetitive behaviour. We previously showed colonic dysmotility in response to GABAergic inhibition and increased myenteric neuronal numbers in the small intestine in Nlgn3R451C mice bred on a mixed genetic background. Here, we show that gut dysfunction is a persistent phenotype of the Nlgn3 R451C mutation in mice backcrossed onto a C57BL/6 background. We report that Nlgn3R451C mice show a 30.9% faster gastrointestinal transit (p = 0.0004) in vivo and have 6% longer small intestines (p = 0.04) compared to wild-types due to a reduction in smooth muscle tone. In Nlgn3R451C mice, we observed a decrease in resting jejunal diameter (proximal jejunum: 10.6% decrease, p = 0.02; mid: 9.8%, p = 0.04; distal: 11.5%, p = 0.009) and neurally regulated dysmotility as well as shorter durations of contractile complexes (mid: 25.6% reduction in duration, p = 0.009; distal: 30.5%, p = 0.004) in the ileum. In Nlgn3R451C mouse colons, short contractions were inhibited to a greater extent (57.2% by the GABAA antagonist, gabazine, compared to 40.6% in wild-type mice (p = 0.007). The inhibition of nitric oxide synthesis decreased the frequency of contractile complexes in the jejunum (WT p = 0.0006, Nlgn3R451C p = 0.002), but not the ileum, in both wild-type and Nlgn3R451C mice. These findings demonstrate that changes in enteric nervous system function contribute to gastrointestinal dysmotility in mice expressing the autism-associated R451C missense mutation in the Neuroligin-3 protein.


Asunto(s)
Trastorno Autístico , Masculino , Animales , Ratones , Ratones Endogámicos C57BL , Trastorno Autístico/genética , Tránsito Gastrointestinal , Intestino Delgado , Yeyuno , Modelos Animales de Enfermedad , Cafeína , Antagonistas del GABA
4.
Infect Immun ; 91(11): e0009723, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37830823

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is a leading cause of bacterial diarrhea with the potential to cause long-term gastrointestinal (GI) dysfunction. Preventative treatments for ETEC-induced diarrhea exist, yet the effects of these treatments on GI commensals in healthy individuals are unclear. Whether administration of a prophylactic preventative treatment for ETEC-induced diarrhea causes specific shifts in gut microbial populations in controlled environments is also unknown. Here, we studied the effects of a hyperimmune bovine colostrum (IMM-124E) used in the manufacture of Travelan (AUST L 106709) on GI bacteria in healthy C57BL/6 mice. Using next-generation sequencing, we aimed to test the onset and magnitude of potential changes to the mouse gut microbiome in response to the antidiarrheagenic hyperimmune bovine colostrum product, rich in immunoglobulins against select ETEC strains (Travelan, Immuron Ltd). We show that in mice administered colostrum containing lipopolysaccharide (LPS) antibodies, there was an increased abundance of potentially gut-beneficial bacteria, such as Akkermansia and Desulfovibrio, without disrupting the underlying ecology of the GI tract. Compared to controls, there was no difference in overall weight gain, body or cecal weights, or small intestine length following LPS antibody colostrum supplementation. Overall, dietary supplementation with colostrum containing LPS antibodies produced subtle alterations in the gut bacterial composition of mice. Primarily, Travelan LPS antibody treatment decreased the ratio of Firmicutes/Bacteroidetes in gut microbial populations in unchallenged healthy mice. Further studies are required to examine the effect of Travelan LPS antibody treatment to engineer the microbiome in a diseased state and during recovery.


Asunto(s)
Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Microbioma Gastrointestinal , Femenino , Embarazo , Ratones , Animales , Bovinos , Lipopolisacáridos , Inmunoglobulina G , Calostro , Ratones Endogámicos C57BL , Factores Inmunológicos , Diarrea/microbiología , Infecciones por Escherichia coli/prevención & control
5.
Am J Physiol Gastrointest Liver Physiol ; 325(3): G230-G238, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37431584

RESUMEN

Maternal influenza A virus (IAV) infection during pregnancy can affect offspring immune programming and development. Offspring born from influenza-infected mothers are at increased risk of neurodevelopmental disorders and have impaired respiratory mucosal immunity against pathogens. The gut-associated lymphoid tissue (GALT) represents a large proportion of the immune system in the body and plays an important role in gastrointestinal (GI) homeostasis. This includes immune modulation to antigens derived from food or microbes, gut microbiota composition, and gut-brain axis signaling. Therefore, in this study, we investigated the effect of maternal IAV infection on mucosal immunity of the GI tract in the offspring. There were no major anatomical changes to the gastrointestinal tract of offspring born to influenza-infected dams. In contrast, maternal IAV did affect the mucosal immunity of offspring, showing regional differences in immune cell profiles within distinct GALT. Neutrophils, monocytes/macrophages, CD4+ and CD8+ T cells infiltration was increased in the cecal patch offspring from IAV-infected dams. In the Peyer's patches, only activated CD4+ T cells were increased in IAV offspring. IL-6 gene expression was also elevated in the cecal patch but not in the Peyer's patches of IAV offspring. These findings suggest that maternal IAV infection perturbs homeostatic mucosal immunity in the offspring gastrointestinal tract. This could have profound ramifications on the gut-brain axis and mucosal immunity in the lungs leading to increased susceptibility to respiratory infections and neurological disorders in the offspring later in life.NEW & NOTEWORTHY Influenza A virus (IAV) infection during pregnancy is associated with changes in gut-associated lymphoid tissue (GALT) in the offspring in a region-dependent manner. Neutrophils and monocytes/macrophages were elevated in the cecal patch of offspring from infected dams. This increase in innate immune cell infiltration was not observed in the Peyer's patches. T cells were also elevated in the cecal patch but not in the Peyer's patches.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Embarazo , Femenino , Ratones , Animales , Humanos , Ganglios Linfáticos Agregados , Inmunidad Mucosa , Linfocitos T CD8-positivos
6.
Dev Neurosci ; 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37717575

RESUMEN

Understanding the long-term functional implications of gut microbial communities during the perinatal period is a bourgeoning area of research. Numerous studies have revealed the existence of a "gut-brain axis" and the impact of an alteration of gut microbiota composition in brain diseases. Recent research has highlighted how gut microbiota could affect brain development and behavior. Many factors in early life such as the mode of delivery or preterm birth could lead to disturbance in the assembly and maturation of gut microbiota. Notably, global rates of cesarean sections (C-sections) have increased in recent decades and remain important when considering premature delivery. Both preterm birth and C-sections are associated with an increased risk of neurodevelopmental disorders such as autism spectrum disorders; with neuroinflammation a major risk factor. In this review, we explore links between preterm birth by C-sections, gut microbiota alteration, and neuroinflammation. We also highlight C-sections as a risk factor for developmental disorders due to alterations in the microbiome.

7.
Parasitol Res ; 122(3): 789-799, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36602586

RESUMEN

Cerebral malaria (CM) is the most severe form of malaria with the highest mortality rate and can result in life-long neurological deficits and ongoing comorbidities. Factors contributing to severity of infection and development of CM are not fully elucidated. Recent studies have indicated a key role of the gut microbiome in a range of health conditions that affect the brain, but limited microbiome research has been conducted in the context of malaria. To address this knowledge gap, the impact of CM on the gut microbiome was investigated in mice. C57BL/6J mice were infected with Plasmodium berghei ANKA (PbA) parasites and compared to non-infected controls. Microbial DNA from faecal pellets collected daily for 6-days post-infection were extracted, and microbiome comparisons conducted using 16S rRNA profiling. We identified significant differences in the composition of bacterial communities between the infected and the non-infected groups, including a higher abundance of the genera Akkermansia, Alistipes and Alloprevotella in PbA-infected mice. Furthermore, intestinal samples were collected post-cull for morphological analysis. We determined that the caecal weight was significantly lower, and the small intestine was significantly longer in PbA-infected mice than in the non-infected controls. We concluded that changes in microbial community composition were primarily driven by the infection protocol and, to a lesser extent, by the time of infection. Our findings pave the way for a new area of research and novel intervention strategies to modulate the severity of cerebral malaria disease.


Asunto(s)
Malaria Cerebral , Microbiota , Animales , Ratones , Malaria Cerebral/parasitología , ARN Ribosómico 16S/genética , Ratones Endogámicos C57BL , Intestinos/microbiología , Plasmodium berghei/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 323(5): G477-G487, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36126271

RESUMEN

Gastrointestinal motility is crucial to gut health and has been associated with different disorders such as inflammatory bowel diseases and postoperative ileus. Despite rat and mouse being the two animal models most widely used in gastrointestinal research, minimal studies in rats have investigated gastrointestinal motility. Therefore, our study provides a comparison of colonic motility in the mouse and rat to clarify species differences and assess the relative effectiveness of each animal model for colonic motility research. We describe the protocol modifications and optimization undertaken to enable video imaging of colonic motility in the rat. Apart from the broad difference in terms of gastrointestinal diameter and length, we identified differences in the fundamental histology of the proximal colon such that the rat had larger villus height-to-width and villus height-to-crypt depth ratios compared with mouse. Since gut motility is tightly regulated by the enteric nervous system (ENS), we investigated how colonic contractile activity within each rodent species responds to modulation of the ENS inhibitory neuronal network. Here we used Nω-nitro-l-arginine (l-NNA), an inhibitor of nitric oxide synthase (NOS) to assess proximal colon responses to the stimulatory effect of blocking the major inhibitory neurotransmitter, nitric oxide (NO). In rats, the frequency of proximal colonic contractions increased in the presence of l-NNA (vs. control levels) to a greater extent than in mice. This is despite a similar number of NOS-expressing neurons in the myenteric plexus across species. Given this increase in colonic contraction frequency, the rat represents another relevant animal model for investigating how gastrointestinal motility is regulated by the inhibitory neuronal network of the ENS.NEW & NOTEWORTHY Mice and rats are widely used in gastrointestinal research but have fundamental differences that make them important as different models for different questions. We found that mice have a higher villi length-to-width and villi length-to-crypt depth ratio than rat in proximal colon. Using the ex vivo video imaging technique, we observed that rat colon has more prominent response to blockade of major inhibitory neurotransmitter (nitric oxide) in myenteric plexus than mouse colon.


Asunto(s)
Sistema Nervioso Entérico , Óxido Nítrico , Ratas , Ratones , Animales , Óxido Nítrico/farmacología , Ratas Sprague-Dawley , Sistema Nervioso Entérico/fisiología , Plexo Mientérico , Motilidad Gastrointestinal/fisiología , Colon , Nitroarginina/farmacología , Óxido Nítrico Sintasa , Modelos Animales de Enfermedad
9.
FASEB J ; 34(1): 1728-1744, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914625

RESUMEN

Gestational diabetes mellitus (GDM) is a common pregnancy complication, particularly prevalent in obese women. Importantly, exercise has beneficial impacts on maternal glucose control and may prevent GDM in "at-risk" women. We aimed to determine whether a high-fat diet (HFD) exacerbates metabolic dysfunction and alters gut microbiome in GDM and whether endurance exercise prevents these changes. Uteroplacental insufficiency was induced by bilateral uterine vessel ligation (Restricted) or sham (Control) surgery on E18 in Wistar-Kyoto rats. Female offspring were fed a Chow or HFD (23% fat) from weaning (5 weeks) and at 16 weeks randomly allocated to remain Sedentary or to an exercise protocol of either Exercise prior to and during pregnancy (Exercise); or Exercise during pregnancy only (PregEx). Females were mated (20 weeks) and underwent indirect calorimetry (embryonic day 16; E16), glucose tolerance testing (E18), followed by 24-hr feces collection at E19 (n = 8-10/group). HFD consumption in female rats with GDM exacerbated the adverse metabolic adaptations to pregnancy and altered gut microbial populations. Specifically, the Firmicutes-to-Bacteroidetes ratio was increased, due to an underlying change in abundance of the orders Clostridiales and Bacteroidales. Maternal Exercise, but not PregEx, prevented the development of metabolic dysfunction, increased pancreatic ß-cell mass, and prevented the alteration of the gut microbiome in GDM females. Our findings suggest that maternal exercise and diet influence metabolic and microbiome dysfunction in females with GDM, which may impact long-term maternal and offspring health.


Asunto(s)
Diabetes Gestacional/metabolismo , Diabetes Gestacional/fisiopatología , Microbiota/fisiología , Condicionamiento Físico Animal/fisiología , Animales , Peso Corporal/fisiología , Dieta Alta en Grasa/efectos adversos , Femenino , Microbioma Gastrointestinal/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Embarazo , Ratas , Destete
10.
Curr Neurol Neurosci Rep ; 21(9): 45, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34227043

RESUMEN

PURPOSE OF REVIEW: Mild traumatic brain injury (mTBI) is a continuing healthcare concern worldwide contributing to significant cognitive and neurological impairment, consequently affecting activities of daily living. While mTBI recovery is becoming well studied, there are no interventions to reduce the known impairments of mTBI. Omega-3 fatty acids (N-3FA) are safe and beneficial for brain health; however, their potential effects in a pathophysiological environment such as that seen post-mTBI are unknown. RECENT FINDINGS: Preclinical studies using rodent models are key to understanding molecular mechanisms underlying improvements post-injury. Studies to date have shown improved outcomes in rodent models following mTBI protocols, but these data have not been quantified using a systematic review and meta-analysis approach. Our systematic review assessed 291 studies identified from the literature. Of these studies, 18 studies met inclusion criteria. We conducted a meta-analysis examining the effect of high-dose n-3FA vs placebo on neurological, cognitive and molecular changes following mTBI. Quality of studies was rated as moderate to high quality, and while mostly compliant, some areas of risk of bias were identified. Results showed that preclinical doses of 10-370 mg/kg/day of n-3FA per day in rodents (equivalent to high clinical doses) resulted in improvements in neurological and cognitive performance (pooled effect sizes ranging between 1.52 and 3.55). Similarly, improvements in molecular and inflammatory markers were observed in treated rodents vs control (pooled effect sizes: 3.73-6.55). Overall, these findings highlight the potential for high-dose n-3FA for human clinical studies following mTBI.


Asunto(s)
Conmoción Encefálica , Lesiones Encefálicas , Ácidos Grasos Omega-3 , Actividades Cotidianas , Encéfalo , Ácidos Grasos Omega-3/uso terapéutico , Humanos
11.
Clin Sci (Lond) ; 134(22): 2943-2957, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33125061

RESUMEN

Gastrointestinal (GI) dysfunction is a common comorbidity of chronic obstructive pulmonary disease (COPD) for which a major cause is cigarette smoking (CS). The underlying mechanisms and precise effects of CS on gut contractility, however, are not fully characterised. Therefore, the aim of the present study was to investigate whether CS impacts GI function and structure in a mouse model of CS-induced COPD. We also aimed to investigate GI function in the presence of ebselen, an antioxidant that has shown beneficial effects on lung inflammation resulting from CS exposure. Mice were exposed to CS for 2 or 6 months. GI structure was analysed by histology and immunofluorescence. After 2 months of CS exposure, ex vivo gut motility was analysed using video-imaging techniques to examine changes in colonic migrating motor complexes (CMMCs). CS decreased colon length in mice. Mice exposed to CS for 2 months had a higher frequency of CMMCs and a reduced resting colonic diameter but no change in enteric neuron numbers. Ten days cessation after 2 months CS reversed CMMC frequency changes but not the reduced colonic diameter phenotype. Ebselen treatment reversed the CS-induced reduction in colonic diameter. After 6 months CS, the number of myenteric nitric-oxide producing neurons was significantly reduced. This is the first evidence of colonic dysmotility in a mouse model of CS-induced COPD. Dysmotility after 2 months CS is not due to altered neuron numbers; however, prolonged CS-exposure significantly reduced enteric neuron numbers in mice. Further research is needed to assess potential therapeutic applications of ebselen in GI dysfunction in COPD.


Asunto(s)
Azoles/farmacología , Fumar Cigarrillos/efectos adversos , Tracto Gastrointestinal/fisiopatología , Compuestos de Organoselenio/farmacología , Animales , Recuento de Células , Forma de la Célula/efectos de los fármacos , Colon/efectos de los fármacos , Colon/patología , Colon/fisiopatología , Sistema Nervioso Entérico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/patología , Isoindoles , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Moco/efectos de los fármacos , Moco/metabolismo , Plexo Mientérico/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo
12.
Brain Behav Immun ; 88: 275-282, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32485290

RESUMEN

Interactions between the gut microbiome and the brain affect mood and behaviour in health and disease. Using preclinical animal models, recent discoveries begin to explain how bacteria in the gut influence our mood as well as highlighting new findings relevant to autism. Autism-associated gene mutations known to alter synapse function in the CNS also affect inflammatory response and modify the enteric nervous system resulting in abnormal gastrointestinal motility and structure. Strikingly, these mutations additionally affect the gut microbiome in mice. This review describes the changes in gut physiology and microbiota in mouse models of autism with modified synapse function. The rationale for different regions of the gastrointestinal tract having variable susceptibility to dysfunction is also discussed. To dissect underlying biological mechanisms involving gut-brain axis dysfunction in preclinical models, a range of multidisciplinary approaches are required. This research will provide insights into the role of the gut-brain axis in health and neurodevelopmental disorders including autism.


Asunto(s)
Trastorno Autístico , Sistema Nervioso Entérico , Microbioma Gastrointestinal , Animales , Trastorno Autístico/genética , Encéfalo , Ratones , Mutación
13.
Brain Behav Immun ; 79: 75-90, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31029798

RESUMEN

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.


Asunto(s)
Trastorno del Espectro Autista/inmunología , Inflamación/inmunología , Neuroinmunomodulación/inmunología , Animales , Astrocitos/inmunología , Astrocitos/fisiología , Trastorno del Espectro Autista/fisiopatología , Encéfalo/inmunología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Humanos , Inflamación/metabolismo , Microglía/inmunología , Microglía/fisiología , Neuroinmunomodulación/fisiología , Neuronas/inmunología , Neuronas/metabolismo
15.
Adv Anat Embryol Cell Biol ; 224: 159-187, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28551756

RESUMEN

Genetic factors might be largely responsible for the development of autism spectrum disorder (ASD) that alone or in combination with specific environmental risk factors trigger the pathology. Multiple mutations identified in ASD patients that impair synaptic function in the central nervous system are well studied in animal models. How these mutations might interact with other risk factors is not fully understood though. Additionally, how systems outside of the brain are altered in the context of ASD is an emerging area of research. Extracerebral influences on the physiology could begin in utero and contribute to changes in the brain and in the development of other body systems and further lead to epigenetic changes. Therefore, multiple recent studies have aimed at elucidating the role of gene-environment interactions in ASD. Here we provide an overview on the extracerebral systems that might play an important associative role in ASD and review evidence regarding the potential roles of inflammation, trace metals, metabolism, genetic susceptibility, enteric nervous system function and the microbiota of the gastrointestinal (GI) tract on the development of endophenotypes in animal models of ASD. By influencing environmental conditions, it might be possible to reduce or limit the severity of ASD pathology.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Encéfalo/fisiopatología , Animales , Trastorno del Espectro Autista/genética , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos
16.
J Physiol ; 594(15): 4325-38, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-26990461

RESUMEN

KEY POINTS: Cholera causes more than 100,000 deaths each year as a result of severe diarrhoea, vomiting and dehydration due to the actions of cholera toxin; more females than males are affected. Cholera toxin induces hypersecretion via release of mucosal serotonin and over-activation of enteric neurons, but its effects on gastrointestinal motility are not well characterized. We found that cholera toxin rapidly and reversibly reduces colonic motility in female mice in oestrus, but not in males or females in prooestrus, an effect mediated by 5-HT in the colonic mucosa and by 5-HT3 receptors. We show that the number of mucosal enterochromaffin cells containing 5-HT changes with the oestrous cycle in mice. These findings indicate that cholera toxin's effects on motility are rapid and depend on the oestrous cycle and therefore can help us better understand differences in responses in males and female patients. ABSTRACT: Extensive studies of the mechanisms responsible for the hypersecretion produced by cholera toxin (CT) have shown that this toxin produces a massive over-activation of enteric neural secretomotor circuits. The effects of CT on gastrointestinal motility, however, have not been adequately characterized. We investigated effects of luminal CT on neurally mediated motor activity in ex vivo male and female mouse full length colon preparations. We used video recording and spatiotemporal maps of contractile activity to quantify colonic migrating motor complexes (CMMCs) and resting colonic diameter. We compared effects of CT in female colon from wild-type and mice lacking tryptophan hydroxylase (TPH1KO). We also compared CMMCs in colons of female mice in oestrus with those in prooestrus. In female (but not male) colon, CT rapidly, reversibly and concentration-dependently inhibits CMMC frequency and induces a tonic constriction. These effects were blocked by granisetron (5-HT3 antagonist) and were absent from TPH1KO females. CT effects were prominent at oestrus but absent at prooestrus. The number of EC cells containing immunohistochemically demonstrable serotonin (5-HT) was 30% greater in female mice during oestrus than during prooestrus or in males. We conclude that CT inhibits CMMCs via release of mucosal 5-HT, which activates an inhibitory pathway involving 5-HT3 receptors. This effect is sex- and oestrous cycle-dependent and is probably due to an oestrous cycle-dependent change in the number of 5-HT-containing EC cells in the colonic mucosa.


Asunto(s)
Toxina del Cólera/farmacología , Colon/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Receptores de Serotonina 5-HT3/fisiología , Animales , Colon/fisiología , Células Enterocromafines/metabolismo , Estrógenos/sangre , Estro , Femenino , Granisetrón/farmacología , Técnicas In Vitro , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Serotonina/farmacología , Antagonistas de la Serotonina/farmacología , Caracteres Sexuales , Triptófano Hidroxilasa/genética
17.
J Neurophysiol ; 115(2): 790-802, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26538613

RESUMEN

Rapid transmembrane flow of sodium ions produces the depolarizing phase of action potentials (APs) in most excitable tissue through voltage-gated sodium channels (NaV). Macroscopic currents display rapid activation followed by fast inactivation (IF) within milliseconds. Slow inactivation (IS) has been subsequently observed in several preparations including neuronal tissues. IS serves important physiological functions, but the kinetic properties are incompletely characterized, especially the operative timescales. Here we present evidence for an "intermediate inactivation" (II) process in rat hippocampal CA1 neurons with time constants of the order of 100 ms. The half-inactivation potentials (V0.5) of steady-state inactivation curves were hyperpolarized by increasing conditioning pulse duration from 50 to 500 ms and could be described by a sum of Boltzmann relations. II state transitions were observed after opening as well as subthreshold potentials. Entry into II after opening was relatively insensitive to membrane potential, and recovery of II became more rapid at hyperpolarized potentials. Removal of fast inactivation with cytoplasmic papaine revealed time constants of INa decay corresponding to II and IS with long depolarizations. Dynamic clamp revealed attenuation of trains of APs over the 10(2)-ms timescale, suggesting a functional role of II in repetitive firing accommodation. These experimental findings could be reproduced with a five-state Markov model. It is likely that II affects important aspects of hippocampal neuron response and may provide a drug target for sodium channel modulation.


Asunto(s)
Potenciales de Acción , Región CA1 Hipocampal/fisiología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Región CA1 Hipocampal/efectos de los fármacos , Femenino , Masculino , Papaína/farmacología , Ratas , Ratas Wistar
18.
J Neurophysiol ; 116(4): 1924-1936, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27489371

RESUMEN

The antiepileptic drug phenytoin (PHT) is thought to reduce the excitability of neural tissue by stabilizing sodium channels (NaV) in inactivated states. It has been suggested the fast-inactivated state (IF) is the main target, although slow inactivation (IS) has also been implicated. Other studies on local anesthetics with similar effects on sodium channels have implicated the NaV voltage sensor interactions. In this study, we reexamined the effect of PHT in both equilibrium and dynamic transitions between fast and slower forms of inactivation in rat hippocampal CA1 pyramidal neurons. The effects of PHT were observed on fast and slow inactivation processes, as well as on another identified "intermediate" inactivation process. The effect of enzymatic removal of IF was also studied, as well as effects on the residual persistent sodium current (INaP). A computational model based on a gating charge interaction was derived that reproduced a range of PHT effects on NaV equilibrium and state transitions. No effect of PHT on IF was observed; rather, PHT appeared to facilitate the occupancy of other closed states, either through enhancement of slow inactivation or through formation of analogous drug-bound states. The overall significance of these observations is that our data are inconsistent with the commonly held view that the archetypal NaV channel inhibitor PHT stabilizes fast inactivation states, and we demonstrate that conventional slow activation "IS" and the more recently identified intermediate-duration inactivation process "II" are the primary functional targets of PHT. In addition, we show that the traditional explanatory frameworks based on the "modulated receptor hypothesis" can be substituted by simple, physiologically plausible interactions with voltage sensors. Additionally, INaP was not preferentially inhibited compared with peak INa at short latencies (50 ms) by PHT.


Asunto(s)
Región CA1 Hipocampal/efectos de los fármacos , Fenitoína/farmacología , Células Piramidales/efectos de los fármacos , Sodio/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Anticonvulsivantes/farmacología , Región CA1 Hipocampal/metabolismo , Simulación por Computador , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Modelos Neurológicos , Técnicas de Placa-Clamp , Células Piramidales/metabolismo , Ratas Wistar , Canales de Sodio/metabolismo , Técnicas de Cultivo de Tejidos
19.
Epilepsia ; 57(9): 1398-405, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27440235

RESUMEN

OBJECTIVE: Fracture risk is a serious comorbidity in epilepsy and may relate to the use of antiepileptic drugs (AEDs). Many AEDs inhibit ion channel function, and the expression of these channels in osteoblasts raises the question of whether altered bone signaling increases bone fragility. We aimed to confirm the expression of voltage-gated sodium (NaV ) channels in mouse osteoblasts, and to investigate the action of carbamazepine and phenytoin on NaV channels. METHODS: Immunocytochemistry was performed on primary calvarial osteoblasts extracted from neonatal C57BL/6J mice and additional RNA sequencing (RNASeq) was included to confirm expression of NaV . Whole-cell patch-clamp recordings were made to identify the native currents expressed and to assess the actions of carbamazepine (50 µm) or phenytoin (50 µm). RESULTS: NaV expression was demonstrated with immunocytochemistry, RNA sequencing, and functionally, with demonstration of robust tetrodotoxin-sensitive and voltage-activated inward currents. Application of carbamazepine or phenytoin resulted in significant inhibition of current amplitude for carbamazepine (31.6 ± 5.9%, n = 9; p < 0.001), and for phenytoin (35.5 ± 6.9%, n = 7; p < 0.001). SIGNIFICANCE: Mouse osteoblasts express NaV , and native NaV currents are blocked by carbamazepine and phenytoin, supporting our hypothesis that AEDs can directly influence osteoblast function and potentially affect bone strength.


Asunto(s)
Anticonvulsivantes/farmacología , Carbamazepina/farmacología , Osteoblastos/efectos de los fármacos , Fenitoína/farmacología , Canales de Sodio/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Regulación de la Expresión Génica/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
20.
Brain ; 137(Pt 6): 1701-15, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24747835

RESUMEN

Epileptic encephalopathies, including Dravet syndrome, are severe treatment-resistant epilepsies with developmental regression. We examined a mouse model based on a human ß1 sodium channel subunit (Scn1b) mutation. Homozygous mutant mice shared phenotypic features and pharmaco-sensitivity with Dravet syndrome. Patch-clamp analysis showed that mutant subicular and layer 2/3 pyramidal neurons had increased action potential firing rates, presumably as a consequence of their increased input resistance. These changes were not seen in L5 or CA1 pyramidal neurons. This raised the concept of a regional seizure mechanism that was supported by data showing increased spontaneous synaptic activity in the subiculum but not CA1. Importantly, no changes in firing or synaptic properties of gamma-aminobutyric acidergic interneurons from mutant mice were observed, which is in contrast with Scn1a-based models of Dravet syndrome. Morphological analysis of subicular pyramidal neurons revealed reduced dendritic arborization. The antiepileptic drug retigabine, a K+ channel opener that reduces input resistance, dampened action potential firing and protected mutant mice from thermal seizures. These results suggest a novel mechanism of disease genesis in genetic epilepsy and demonstrate an effective mechanism-based treatment of the disease.


Asunto(s)
Dendritas/genética , Epilepsias Mioclónicas/genética , Mutación/genética , Neuronas/metabolismo , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/metabolismo , Potenciales de Acción/genética , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA