Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurochem Int ; 142: 104919, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33242538

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease which causes a non-reversible cognitive impairment and dementia. The primary cause of late-onset AD remains unknown although its pathology was discovered over a century ago. Recently, the vascular hypothesis of AD has received backing from evidence emerging from neuroimaging studies which have revealed the presence of a significant hypoperfusion in the brain regions vulnerable to AD pathology. In fact, hypoxia can explain many of the pathological changes evident in AD pathology, e.g. the deposition of ß-amyloid plaques and chronic low-grade inflammation. Hypoxia-inducible factor-1α (HIF-1α) stimulates inflammatory responses and modulates both innate and adaptive immunity. It is known that hypoxia-induced inflammation evokes compensatory anti-inflammatory response involving tissue-resident microglia/macrophages and infiltrated immune cells. Hypoxia/HIF-1α induce immunosuppression by (i) increasing the expression of immunosuppressive genes, (ii) stimulating adenosinergic signaling, (iii) enhancing aerobic glycolysis, i.e. lactate production, and (iv) augmenting the secretion of immunosuppressive exosomes. Interestingly, it seems that these common mechanisms are also involved in the pathogenesis of AD. In AD pathology, an enhanced immunosuppression appears, e.g. as a shift in microglia/macrophage phenotypes towards the anti-inflammatory M2 phenotype and an increase in the numbers of regulatory T cells (Treg). The augmented anti-inflammatory capacity promotes the resolution of acute inflammation but persistent inflammation has crucial effects not only on immune cells but also harmful responses to the homeostasis of AD brain. I will examine in detail the mechanisms of the hypoperfusion/hypoxia-induced immunosuppressive state in general and especially, in its association with AD pathogenesis. These immunological observations support the vascular hypothesis of AD pathology.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Circulación Cerebrovascular/fisiología , Tolerancia Inmunológica/fisiología , Acoplamiento Neurovascular/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Trastornos Cerebrovasculares/inmunología , Trastornos Cerebrovasculares/metabolismo , Humanos , Red Nerviosa/irrigación sanguínea , Red Nerviosa/inmunología , Red Nerviosa/metabolismo
2.
J Neural Transm (Vienna) ; 127(4): 467-479, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31399790

RESUMEN

Pain is a necessary sensation that prevents further tissue damage, but can be debilitating and detrimental in daily life under chronic conditions. Neuronal activity strongly regulates the maturation of the somatosensory system, and aberrant sensory input caused by injury or inflammation during critical periods of early postnatal development can have prolonged, detrimental effects on pain processing. This review will outline the maturation of neuronal circuits responsible for the transmission of nociceptive signals and the generation of pain sensation-involving peripheral sensory neurons, the spinal cord dorsal horn, and brain-in addition to the influences of the neuroimmune system on somatosensation. This summary will also highlight the unique effects of neonatal tissue injury on the maturation of these systems and subsequent consequences for adult somatosensation. Ultimately, this review emphasizes the need to account for age as an independent variable in basic and clinical pain research, and importantly, to consider the distinct qualities of the pediatric population when designing novel strategies for pain management.


Asunto(s)
Vías Aferentes , Encéfalo , Enfermedades del Recién Nacido , Red Nerviosa , Plasticidad Neuronal/fisiología , Nocicepción/fisiología , Nociceptores/fisiología , Asta Dorsal de la Médula Espinal , Heridas y Lesiones , Vías Aferentes/crecimiento & desarrollo , Vías Aferentes/inmunología , Vías Aferentes/fisiopatología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/inmunología , Encéfalo/fisiopatología , Humanos , Recién Nacido , Enfermedades del Recién Nacido/tratamiento farmacológico , Enfermedades del Recién Nacido/inmunología , Enfermedades del Recién Nacido/fisiopatología , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/inmunología , Red Nerviosa/fisiopatología , Asta Dorsal de la Médula Espinal/crecimiento & desarrollo , Asta Dorsal de la Médula Espinal/inmunología , Asta Dorsal de la Médula Espinal/fisiopatología , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/inmunología , Heridas y Lesiones/fisiopatología
3.
J Ocul Pharmacol Ther ; 35(8): 447-456, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31596665

RESUMEN

The cornea and its adnexa pose a unique situation of a tightly defined set of requirements for its function. This includes: transparency, perfect built to obtain appropriate refractive power, protective barrier from microbial invaders. Moreso, the cornea also endures extreme external physical conditions (temperature, high and low humidity, winds and alike). All these functions are maintained while preserving a constant state of homogenous wetting. Toward that end the cornea is equipped with an elaborated network of sensory neural network. While enabling the blinking reflex and maintaining the physiological steady state of wetting, this neural network also makes the cornea prone to the discomfort that with or without associated changes seen on medical examination. ISOPT Clinical 2018 discussion touched upon this hypercomplex situation, addressing the role of inflammation and its resulting discomfort in dry eye conditions. The discussion also engulfed the emerging neuropathic pain syndrome that is recently gaining more attention. Another related topic was the utilization of autologous serum tears and its ability to provide amelioration to desperate patients. Finally, the panel discussed the issue of treating corneal infection, including when and how to utilize steroids in the course of therapy. We assume the reader will find interest in this discussion that directly addresses issues seen day in and day out in our busy clinics.


Asunto(s)
Enfermedades de la Córnea/tratamiento farmacológico , Síndromes de Ojo Seco/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Córnea/efectos de los fármacos , Córnea/inmunología , Córnea/inervación , Enfermedades de la Córnea/inmunología , Enfermedades de la Córnea/metabolismo , Síndromes de Ojo Seco/inmunología , Humanos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/inmunología , Soluciones Oftálmicas , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/inmunología , Lágrimas/metabolismo
4.
Mult Scler Relat Disord ; 36: 101390, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31546226

RESUMEN

This is a review paper, essentially a commentary with summary of literature that actualizes the problem of epilepsy in patients with multiple sclerosis. There is a bidirectional relation between multiple sclerosis and epilepsy. A possible associate pathophysiological pathway is considered. In multiple sclerosis, a combination of gray matter involvement and inflammation could influence epileptogenesis. Patients with multiple sclerosis have individual profiles and an inter-individual variability of epileptogenicity. No treatment guidelines have been specified for these patients. We postulate that an epileptic manifestation means a relapse or an aggravation of the inflammatory process. In this condition, over time, this symptom could integrate into the Expanded Disability Status Scale. Epileptogenesis is an active process and an interesting question is if disease-modifying therapy in multiple sclerosis can prevent, or mitigate, epilepsy. In light of the latest knowledge of the inflammatory process in epilepsy, the possibility of preventing epileptogenesis with actual treatment of MS is emphasized. We would argue that it is a strong argument for starting treatment quicker for both diseases. Over the last few years, the concepts of epilepsy have completely changed. The model of epilepsy in multiple sclerosis can currently be regarded as a network disease and this new concept can have a highly significant clinical impact.


Asunto(s)
Corteza Cerebral , Epilepsia , Esclerosis Múltiple , Red Nerviosa , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Epilepsia/tratamiento farmacológico , Epilepsia/inmunología , Epilepsia/patología , Epilepsia/fisiopatología , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Red Nerviosa/inmunología , Red Nerviosa/patología , Red Nerviosa/fisiopatología
5.
Methods Mol Biol ; 2034: 3-11, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392673

RESUMEN

Microglia are neural cells of nonneural origin; they originate from fetal macrophages that invade neural tube early in embryogenesis and undergo the most idiosyncratic metamorphosis which coverts them into elements of neural circuitry. Microglia appeared early in evolution with neural immune cells being operative in leeches and mollusks. Microglial cells acquire specific morphology characterized by small cell bodies and long motile processes which are packed with receptors sensing both physiological and pathological stimuli. Microglial cells actively sculpture neuronal networks through synaptic stripping and phagocytosis of redundant neurons; microglia also secrete neuroactive factors regulating synaptic transmission. Novel techniques emerging in recent decade allowed an in-depth understanding of physiological and pathophysiological functions of microglia.


Asunto(s)
Feto/inmunología , Macrófagos/inmunología , Microglía/inmunología , Red Nerviosa/inmunología , Fagocitosis , Sinapsis/inmunología , Animales , Humanos
6.
Front Immunol ; 10: 1779, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417557

RESUMEN

Background: Efficient personalized therapy paradigms are needed to modify the disease course and halt gray (GM) and white matter (WM) damage in patients with multiple sclerosis (MS). Presently, promising disease-modifying drugs show impressive efficiency, however, tailored markers of therapy responses are required. Here, we aimed to detect in a real-world setting patients with a more favorable brain network response and immune cell dynamics upon dimethyl fumarate (DMF) treatment. Methods: In a cohort of 78 MS patients we identified two thoroughly matched groups, based on age, disease duration, disability status and lesion volume, receiving DMF (n = 42) and NAT (n = 36) and followed them over 16 months. The rate of cortical atrophy and deep GM volumes were quantified. GM and WM network responses were characterized by brain modularization as a marker of regional and global structural alterations. In the DMF group, lymphocyte subsets were analyzed by flow cytometry and related to clinical and MRI parameters. Results: Sixty percent (25 patients) of the DMF and 36% (13 patients) of the NAT group had disease activity during the study period. The rate of cortical atrophy was higher in the DMF group (-2.4%) compared to NAT (-2.1%, p < 0.05) group. GM and WM network dynamics presented increased modularization in both groups. When dividing the DMF-treated cohort into patients free of disease activity (n = 17, DMFR) and patients with disease activity (n = 25, DMFNR) these groups differed significantly in CD8+ cell depletion counts (DMFR: 197.7 ± 97.1/µl; DMFNR: 298.4 ± 190.6/µl, p = 0.03) and also in cortical atrophy (DMFR: -1.7%; DMFNR: -3.2%, p = 0.01). DMFR presented reduced longitudinal GM and WM modularization and less atrophy as markers of preserved structural global network integrity in comparison to DMFNR and even NAT patients. Conclusions: NAT treatment contributes to a reduced rate of cortical atrophy compared to DMF therapy. However, patients under DMF treatment with a stronger CD8+ T cell depletion present a more favorable response in terms of cortical integrity and GM and WM network responses. Our findings may serve as basis for the development of personalized treatment paradigms.


Asunto(s)
Linfocitos T CD8-positivos , Corteza Cerebral , Dimetilfumarato/administración & dosificación , Factores Inmunológicos/administración & dosificación , Esclerosis Múltiple , Red Nerviosa/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Femenino , Sustancia Gris/inmunología , Sustancia Gris/patología , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Red Nerviosa/patología , Sustancia Blanca/inmunología , Sustancia Blanca/patología
7.
Neuroimage ; 200: 121-131, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31201984

RESUMEN

Recent studies have shown a critical role of the gastrointestinal microbiome in brain and behavior via the complex gut-microbiome-brain axis. However, the influence of the oral microbiome in neurological processes is much less studied, especially in response to the stimuli, such as smoking, within the oral microenvironment. Additionally, given the complex structural and functional networks in brain, our knowledge about the relationship between microbiome and brain function through specific brain circuits is still very limited. In this pilot study, we leveraged next generation sequencing for microbiome and functional neuroimaging technique to enable the delineation of microbiome-brain network links as well as their relationship to cigarette smoking. Thirty smokers and 30 age- and sex-matched nonsmokers were recruited for 16S sequencing of their oral microbial community. Among them, 56 subjects were scanned by resting-state functional magnetic resonance imaging to derive brain functional networks. Statistical analyses were performed to demonstrate the influence of smoking on the oral microbial composition, functional network connectivity, and the associations between microbial shifts and functional network connectivity alternations. Compared to nonsmokers, we found a significant decrease of beta diversity (P = 6 × 10-3) in smokers and identified several classes (Betaproteobacteria, Spirochaetia, Synergistia, and Mollicutes) with significant alterations in microbial abundance. Pathway analysis on the predicted KEGG pathways shows that the microbiota with altered abundance are mainly involved in pathways related to cell processes, DNA repair, immune system, and neurotransmitters signaling. One brain functional network connectivity component was identified to have a significant difference between smokers and nonsmokers (P = 0.032), mainly including connectivity between brain default network and other task-positive networks. This brain functional component was also significantly associated with smoking related microbiota, suggesting a correlated cross-individual pattern between smoking-induced oral microbiome dysbiosis and brain functional connectivity alternation, possibly involving immunological and neurotransmitter signaling pathways. This work is the first attempt to link oral microbiome and brain functional networks, and provides support for future work in characterizing the role of oral microbiome in mediating smoking effects on brain activity.


Asunto(s)
Corteza Cerebral/fisiopatología , Conectoma , Disbiosis/microbiología , Microbiota/fisiología , Boca/microbiología , Red Nerviosa/fisiopatología , Transducción de Señal/fisiología , Fumar/fisiopatología , Adulto , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/inmunología , Disbiosis/etiología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/inmunología , Proyectos Piloto , Saliva/microbiología , Transducción de Señal/inmunología , Fumar/efectos adversos , Adulto Joven
8.
J Neurol ; 266(7): 1809-1815, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31004212

RESUMEN

The last two decades have seen an explosion in our understanding of the clinical nature of narcolepsy and its pathogenesis, fuelling new approaches to potentially effective treatments. It is now recognised that the full narcoleptic syndrome has significant adverse effects on sleep regulation across the full 24-h period and is often associated with clinical features outside the sleep-wake domain. The discovery that most narcoleptic subjects specifically lack a hypothalamic neuropeptide (hypocretin, also called orexin) was a truly original and landmark observation in 1999, greatly furthering our understanding both of the syndrome itself and sleep biology in general. An autoimmune pathophysiology has long been suggested by the tight association with specific histocompatibility antigens and very recently partly confirmed by detailed analysis of T-cell immunological function in affected subjects. Drug treatments remain symptomatic but may soon become more focussed by restoring central hypocretin signalling with replacement therapy. Potentially disease-modifying, immunological approaches have yet to be studied systematically, although the interval between disease onset and development of the full clinical syndrome may be longer than previously appreciated, affording a realistic window of opportunity for limiting neuronal damage in this disabling condition.


Asunto(s)
Encéfalo/inmunología , Narcolepsia/inmunología , Red Nerviosa/inmunología , Orexinas/inmunología , Encéfalo/metabolismo , Emociones/fisiología , Humanos , Narcolepsia/diagnóstico , Narcolepsia/metabolismo , Red Nerviosa/metabolismo , Orexinas/metabolismo
9.
Neurochem Int ; 130: 104316, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30342960

RESUMEN

Stroke causes neuronal cell death and destruction of neuronal circuits in the brain and spinal cord. Injury to the brain tissue induces sterile inflammation triggered by the extracellular release of endogenous molecules, but cerebral inflammation after stroke is gradually resolved within several days. In this pro-resolving process, inflammatory cells adopt a pro-resolving or repairing phenotype in the injured brain, activating endogenous repairing programs. Although the mechanisms involved in the transition from inflammation to neural repair after stroke remain largely unknown to date, some of the mechanisms for inflammation and neural repair have been clarified in detail. This review focuses on the molecular or cellular mechanisms involved in sterile inflammation and neural repair after stroke. This accumulation of evidence may be helpful for speculating about the endogenous repairing mechanisms in the brain and identifying therapeutic targets for improving the functional prognoses of stroke patients.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Red Nerviosa/inmunología , Células-Madre Neurales/inmunología , Neurogénesis/fisiología , Accidente Cerebrovascular/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Red Nerviosa/metabolismo , Red Nerviosa/patología , Células-Madre Neurales/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
10.
Immun Inflamm Dis ; 6(2): 354-370, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29635889

RESUMEN

INTRODUCTION: Recently, we found abundant innervation of antigen presenting cells that were reached and enclosed by single neurites. These neurally hard-wired antigen presenting cells (wAPC) could be observed in the T-cell zone of superficial cervical lymph nodes of rats and other mammalians, including humans. METHODS: As a consequence, we investigated lymph nodes at many different anatomical positions as well as all primary and secondary lymphoid organs (SLO) in rodents for a similar morphology of innervation regarding antigen presenting cells known in those tissues. RESULTS: As a result, we confirmed wAPC in lymph nodes independent from their draining areas and anatomical positions but also in all other T-cell zones of lymphoid organs, like Peyer's patches, NALT and BALT, as well as in the thymic medulla. Other cells were innervated in a similar fashion but with seemingly missing antigen presenting capacity. Both types of innervated immune cells were observed as being also present in the dermis of the skin. Only in the spleen wAPC could not be detected. Beyond this systematic finding, we also found another regular phenomenon: a dense network of neurites that stained for neurofilament always in antigen entrance areas of lymphoid organs (subsinoidal layer of lymph nodes, subepithelial dome of Peyer's patches, subsinoidal layer of the splenic white pulp, margins of NALT and BALT). Lastly, also thymic epithelial cells (TEC) restricted to the corticomedullary junction of the thymus showed similar neurofilament staining. CONCLUSIONS: Therefore, we propose much more hard-wired and probably afferent connections between lymphoid organs and the central nervous system than is hitherto known.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Tejido Linfoide/inmunología , Red Nerviosa/inmunología , Neuritas/inmunología , Neuroinmunomodulación/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/inervación , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Animales , Neuritas/metabolismo , Neuronas Aferentes/inmunología , Neuronas Aferentes/metabolismo , Ratas , Ratas Sprague-Dawley
11.
J Neurosci ; 38(11): 2877-2886, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29487127

RESUMEN

Prenatal maternal immune activation (MIA) is associated with altered brain development and risk of psychiatric disorders in offspring. Translational human studies of MIA are few in number. Alterations of the salience network have been implicated in the pathogenesis of the same psychiatric disorders associated with MIA. If MIA is pathogenic, then associated abnormalities in the salience network should be detectable in neonates immediately after birth. We tested the hypothesis that third trimester MIA of adolescent women who are at risk for high stress and inflammation is associated with the strength of functional connectivity in the salience network of their neonate. Thirty-six women underwent blood draws to measure interleukin-6 (IL-6) and C-reactive protein (CRP) and electrocardiograms to measure fetal heart rate variability (FHRV) at 34-37 weeks gestation. Resting-state imaging data were acquired in the infants at 40-44 weeks postmenstrual age (PMA). Functional connectivity was measured from seeds placed in the anterior cingulate cortex and insula. Measures of cognitive development were obtained at 14 months PMA using the Bayley Scales of Infant and Toddler Development-Third Edition (BSID-III). Both sexes were studied. Regions in which the strength of the salience network correlated with maternal IL-6 or CRP levels included the medial prefrontal cortex, temporoparietal junction, and basal ganglia. Maternal CRP level correlated inversely with FHRV acquired at the same gestational age. Maternal CRP and IL-6 levels correlated positively with measures of cognitive development on the BSID-III. These results suggest that MIA is associated with short- and long-term influences on offspring brain and behavior.SIGNIFICANCE STATEMENT Preclinical studies in rodents and nonhuman primates and epidemiological studies in humans suggest that maternal immune activation (MIA) alters the development of brain circuitry and associated behaviors, placing offspring at risk for psychiatric illness. Consistent with preclinical findings, we show that maternal third trimester interleukin-6 and C-reactive protein levels are associated with neonatal functional connectivity and with both fetal and toddler behavior. MIA-related functional connectivity was localized to the salience, default mode, and frontoparietal networks, which have been implicated in the pathogenesis of psychiatric disorders. Our results suggest that MIA alters functional connectivity in the neonatal brain, that those alterations have consequences for cognition, and that these findings may provide pathogenetic links between preclinical and epidemiological studies associating MIA with psychiatric risk in offspring.


Asunto(s)
Desarrollo Infantil/fisiología , Conducta del Lactante/fisiología , Red Nerviosa/inmunología , Red Nerviosa/fisiología , Tercer Trimestre del Embarazo/inmunología , Adolescente , Ganglios Basales/crecimiento & desarrollo , Ganglios Basales/fisiología , Proteína C-Reactiva/análisis , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Cognición/fisiología , Electrocardiografía , Femenino , Feto/fisiología , Frecuencia Cardíaca Fetal , Humanos , Lactante , Recién Nacido , Interleucina-6/sangre , Masculino , Red Nerviosa/crecimiento & desarrollo , Pruebas Neuropsicológicas , Embarazo , Adulto Joven
12.
Science ; 359(6379): 1056-1061, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29496881

RESUMEN

The type 2 inflammatory response is induced by various environmental and infectious stimuli. Although recent studies identified group 2 innate lymphoid cells (ILC2s) as potent sources of type 2 cytokines, the molecular pathways controlling ILC2 responses are incompletely defined. Here we demonstrate that murine ILC2s express the ß2-adrenergic receptor (ß2AR) and colocalize with adrenergic neurons in the intestine. ß2AR deficiency resulted in exaggerated ILC2 responses and type 2 inflammation in intestinal and lung tissues. Conversely, ß2AR agonist treatment was associated with impaired ILC2 responses and reduced inflammation in vivo. Mechanistically, we demonstrate that the ß2AR pathway is a cell-intrinsic negative regulator of ILC2 responses through inhibition of cell proliferation and effector function. Collectively, these data provide the first evidence of a neuronal-derived regulatory circuit that limits ILC2-dependent type 2 inflammation.


Asunto(s)
Inmunidad Adaptativa , Neuronas Adrenérgicas/inmunología , Inmunidad Innata , Linfocitos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Humanos , Inflamación/inmunología , Intestinos/inmunología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/inmunología , Receptores Adrenérgicos beta 2/genética , Transducción de Señal
13.
J Neurosci ; 37(45): 10848-10854, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118214

RESUMEN

The brain processes information from the periphery and regulates visceral and immune activity to maintain internal homeostasis, optimally respond to a dynamic external environment, and integrate these functions with ongoing behavior. In addition to its relevance for survival, this integration underlies pathology as evidenced by diseases exhibiting comorbid visceral and psychiatric symptoms. Advances in neuroanatomical mapping, genetically specific neuronal manipulation, and neural network recording are overcoming the challenges of dissecting complex circuits that underlie this integration and deciphering their function. Here we focus on reciprocal communication between the brain and urological, gastrointestinal, and immune systems. These studies are revealing how autonomic activity becomes integrated into behavior as part of a social strategy, how the brain regulates innate immunity in response to stress, and how drugs impact emotion and gastrointestinal function. These examples highlight the power of the functional organization of circuits at the interface of the brain and periphery.


Asunto(s)
Sistema Nervioso Central/fisiología , Homeostasis/fisiología , Inmunidad Humoral/fisiología , Red Nerviosa/fisiología , Animales , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Humanos , Red Nerviosa/inmunología
14.
Adv Immunol ; 135: 53-79, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28826529

RESUMEN

Recent discoveries implicate the classical complement cascade in normal brain development and in disease. Complement proteins C1q, C3, and C4 participate in synapse elimination, tagging inappropriate synaptic connections between neurons for removal by phagocytic microglia that exist in a special, highly phagocytic state during the synaptic pruning period. Several neurodevelopmental disorders, such as schizophrenia and autism, are thought to be caused by an imbalance in synaptic pruning, and recent studies suggest that dysregulation of complement could promote this synaptic pruning imbalance. Moreover, in the mature brain, complement can be aberrantly activated in early stages of neurodegenerative diseases to stimulate synapse loss. Similar pathways can also be activated in response to inflammation, as in West Nile Virus infection or in lupus, where peripheral inflammation can promote microglia-mediated synapse loss. Whether synapse loss in disease is a true reactivation of developmental synaptic pruning programs remains unclear; nonetheless, complement proteins represent potential therapeutic targets for both neurodevelopmental and neurodegenerative diseases.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Red Nerviosa/inmunología , Neurogénesis/inmunología , Plasticidad Neuronal/inmunología , Sinapsis/inmunología , Animales , Trastorno Autístico/genética , Trastorno Autístico/inmunología , Trastorno Autístico/patología , Proteínas del Sistema Complemento/genética , Epilepsia/genética , Epilepsia/inmunología , Epilepsia/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Microglía/inmunología , Microglía/patología , Red Nerviosa/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neurogénesis/genética , Plasticidad Neuronal/genética , Neuronas/inmunología , Neuronas/patología , Esquizofrenia/genética , Esquizofrenia/inmunología , Esquizofrenia/patología , Sinapsis/genética , Sinapsis/patología , Transmisión Sináptica
15.
Mol Immunol ; 89: 73-83, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28601358

RESUMEN

Originally discovered as part of C1, the initiation component of the classical complement pathway, it is now appreciated that C1q regulates a variety of cellular processes independent of complement activation. C1q is a complex glycoprotein assembled from 18 polypeptide chains, with a C-terminal globular head region that mediates recognition of diverse molecular structures, and an N-terminal collagen-like tail that mediates immune effector mechanisms. C1q mediates a variety of immunoregulatory functions considered important in the prevention of autoimmunity such as the enhancement of phagocytosis, regulation of cytokine production by antigen presenting cells, and subsequent alteration in T-lymphocyte maturation. Furthermore, recent advances indicate additional roles for C1q in diverse physiologic and pathologic processes including pregnancy, tissue repair, and cancer. Finally, C1q is emerging as a critical component of neuronal network refinement and homeostatic regulation within the central nervous system. This review summarizes the classical functions of C1q and reviews novel discoveries within the field.


Asunto(s)
Sistema Nervioso Central/inmunología , Complemento C1q/inmunología , Homeostasis/inmunología , Red Nerviosa/inmunología , Inmunidad Adaptativa/inmunología , Fenómenos Fisiológicos Celulares/inmunología , Complemento C1q/química , Humanos , Inmunidad Innata/inmunología , Modelos Moleculares , Estructura Cuaternaria de Proteína
16.
J Clin Invest ; 127(1): 33-42, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045403

RESUMEN

Obesity and diabetes are associated with increased chronic low-grade inflammation and elevated plasma glucose levels. Although inflammation in the fat and liver are established features of obesity-associated insulin resistance, the intestine is emerging as a new site for immunologic changes that affect whole-body metabolism. Specifically, microbial and dietary factors incurred by diet-induced obesity influence underlying innate and adaptive responses of the intestinal immune system. These responses affect the maintenance of the intestinal barrier, systemic inflammation, and glucose metabolism. In this Review we propose that an understanding of the changes to the intestinal immune system, and how these changes influence systemic immunity and glucose metabolism in a whole-body integrative and a neuronal-dependent network, will unveil novel intestinal pathologic and therapeutic targets for diabetes and obesity.


Asunto(s)
Inmunidad Adaptativa , Diabetes Mellitus/inmunología , Inmunidad Innata , Resistencia a la Insulina/inmunología , Intestinos/inmunología , Obesidad/inmunología , Animales , Diabetes Mellitus/patología , Glucosa/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Intestinos/patología , Red Nerviosa/inmunología , Red Nerviosa/patología , Obesidad/patología
17.
Horm Behav ; 87: 25-34, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27765499

RESUMEN

Multidirectional interactions among the immune, endocrine, and nervous systems have been demonstrated in humans and non-human animal models for many decades by the biomedical community, but ecological and evolutionary perspectives are lacking. Neuroendocrine-immune interactions can be conceptualized using a series of feedback loops, which culminate into distinct neuroendocrine-immune phenotypes. Behavior can exert profound influences on these phenotypes, which can in turn reciprocally modulate behavior. For example, the behavioral aspects of reproduction, including courtship, aggression, mate selection and parental behaviors can impinge upon neuroendocrine-immune interactions. One classic example is the immunocompetence handicap hypothesis (ICHH), which proposes that steroid hormones act as mediators of traits important for female choice while suppressing the immune system. Reciprocally, neuroendocrine-immune pathways can promote the development of altered behavioral states, such as sickness behavior. Understanding the energetic signals that mediate neuroendocrine-immune crosstalk is an active area of research. Although the field of psychoneuroimmunology (PNI) has begun to explore this crosstalk from a biomedical standpoint, the neuroendocrine-immune-behavior nexus has been relatively underappreciated in comparative species. The field of ecoimmunology, while traditionally emphasizing the study of non-model systems from an ecological evolutionary perspective, often under natural conditions, has focused less on the physiological mechanisms underlying behavioral responses. This review summarizes neuroendocrine-immune interactions using a comparative framework to understand the ecological and evolutionary forces that shape these complex physiological interactions.


Asunto(s)
Sistema Inmunológico/fisiología , Red Nerviosa/fisiología , Neuroinmunomodulación/fisiología , Sistemas Neurosecretores/fisiología , Animales , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Femenino , Hormonas/farmacología , Hormonas/fisiología , Humanos , Conducta de Enfermedad/fisiología , Red Nerviosa/inmunología , Fenotipo , Psiconeuroinmunología , Reproducción/fisiología
18.
Pharmacol Ther ; 154: 21-35, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26129625

RESUMEN

Recent advanced immunological analyses have revealed that the diversity and plasticity of macrophages lead to the identification of functional polarization states (classically activated M1 type and alternatively activated M2 type) which are dependent on the extracellular environment. M1 and M2 polarization states of macrophages play an important role in controlling the balance between pro-inflammatory and anti-inflammatory conditions. Microglial cells are resident mononuclear phagocytes in the central nervous system (CNS), express several macrophage-associated markers, and appear to display functional polarization states similar to macrophages. Like M1 macrophages, M1 polarized microglia can produce pro-inflammatory cytokines and mediators such as interleukin (IL) 1ß, IL-6, tumor necrosis factor-α, CC-chemokine ligand 2, nitric oxide, and reactive oxygen species, suggesting that these molecules contribute to dysfunction of neural network in the CNS. On the other hand, M2 polarized microglia can produce anti-inflammatory cytokine, IL-10 and express several receptors that are implicated in inhibiting inflammation and restoring homeostasis. In this review, we summarize the diversity, plasticity, and immunoregulatory functions of M1 and M2 microglia in psychiatric and neurological disorders. Based on these aspects, we propose a contribution of imbalance between M1 and M2 polarization of microglia in bipolar disorder, obesity, amyotrophic lateral sclerosis, and Rett syndrome. Consequently, molecules that normalize the imbalance between M1 and M2 microglial polarization states may provide a beneficial therapeutic target for the treatment of these disorders.


Asunto(s)
Trastorno Bipolar/inmunología , Macrófagos/inmunología , Microglía/inmunología , Enfermedades del Sistema Nervioso/inmunología , Obesidad/inmunología , Adiponectina/inmunología , Encéfalo/inmunología , Citocinas/inmunología , Endocannabinoides/inmunología , Conducta Alimentaria/fisiología , Ghrelina/inmunología , Humanos , Mediadores de Inflamación/inmunología , Lisofosfolípidos/inmunología , Trastornos Mentales/inmunología , Red Nerviosa/inmunología , Conducta Social , Esfingosina/análogos & derivados , Esfingosina/inmunología
19.
Lancet Neurol ; 14(3): 318-28, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25728441

RESUMEN

The discovery of hypocretins (orexins) and their causal implication in narcolepsy is the most important advance in sleep research and sleep medicine since the discovery of rapid eye movement sleep. Narcolepsy with cataplexy is caused by hypocretin deficiency owing to destruction of most of the hypocretin-producing neurons in the hypothalamus. Ablation of hypocretin or hypocretin receptors also leads to narcolepsy phenotypes in animal models. Although the exact mechanism of hypocretin deficiency is unknown, evidence from the past 20 years strongly favours an immune-mediated or autoimmune attack, targeting specifically hypocretin neurons in genetically predisposed individuals. These neurons form an extensive network of projections throughout the brain and show activity linked to motivational behaviours. The hypothesis that a targeted immune-mediated or autoimmune attack causes the specific degeneration of hypocretin neurons arose mainly through the discovery of genetic associations, first with the HLA-DQB1*06:02 allele and then with the T-cell receptor α locus. Guided by these genetic findings and now awaiting experimental testing are models of the possible immune mechanisms by which a specific and localised brain cell population could become targeted by T-cell subsets. Great hopes for the identification of new targets for therapeutic intervention in narcolepsy also reside in the development of patient-derived induced pluripotent stem cell systems.


Asunto(s)
Encéfalo/fisiopatología , Cataplejía/fisiopatología , Péptidos y Proteínas de Señalización Intracelular , Narcolepsia/fisiopatología , Red Nerviosa/fisiopatología , Neuropéptidos , Animales , Encéfalo/inmunología , Cataplejía/genética , Cataplejía/inmunología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Narcolepsia/genética , Narcolepsia/inmunología , Red Nerviosa/inmunología , Neuropéptidos/genética , Neuropéptidos/inmunología , Orexinas
20.
Dev Cogn Neurosci ; 11: 18-30, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25081071

RESUMEN

Exposure to adversity and stress early in development yields vulnerability to mental illnesses throughout the lifespan. Growing evidence suggests that this vulnerability has mechanistic origins involving aberrant development of both neurocircuitry and neuro-immune activity. Here we review the current understanding of when and how stress exposure initiates neuroinflammatory events that interact with brain development. We first review how early life adversity has been associated with various psychopathologies, and how neuroinflammation plays a role in these pathologies. We then summarize data and resultant hypotheses describing how early life adversity may particularly alter neuro-immune development with psychiatric consequences. Finally, we review how sex differences contribute to individualistic vulnerabilities across the lifespan. We submit the importance of understanding how stress during early development might cause outright neural or glial damage, as well as experience-dependent plasticity that may insufficiently prepare an individual for sex-specific or life-stage specific challenges.


Asunto(s)
Inflamación/inmunología , Inflamación/psicología , Trastornos Mentales/etiología , Red Nerviosa/inmunología , Neuroinmunomodulación , Conducta Social , Estrés Psicológico/complicaciones , Adolescente , Adulto , Niño , Desarrollo Infantil , Preescolar , Trastornos del Desarrollo Sexual/inmunología , Trastornos del Desarrollo Sexual/psicología , Femenino , Humanos , Lactante , Inflamación/complicaciones , Inflamación/etiología , Masculino , Trastornos Mentales/inmunología , Trastornos Mentales/psicología , Red Nerviosa/patología , Red Nerviosa/fisiopatología , Caracteres Sexuales , Factores Sexuales , Estrés Psicológico/inmunología , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA