Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Alzheimers Dement ; 20(6): 4260-4289, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38687209

RESUMEN

Experimental laboratory research has an important role to play in dementia prevention. Mechanisms underlying modifiable risk factors for dementia are promising targets for dementia prevention but are difficult to investigate in human populations due to technological constraints and confounds. Therefore, controlled laboratory experiments in models such as transgenic rodents, invertebrates and in vitro cultured cells are increasingly used to investigate dementia risk factors and test strategies which target them to prevent dementia. This review provides an overview of experimental research into 15 established and putative modifiable dementia risk factors: less early-life education, hearing loss, depression, social isolation, life stress, hypertension, obesity, diabetes, physical inactivity, heavy alcohol use, smoking, air pollution, anesthetic exposure, traumatic brain injury, and disordered sleep. It explores how experimental models have been, and can be, used to address questions about modifiable dementia risk and prevention that cannot readily be addressed in human studies. HIGHLIGHTS: Modifiable dementia risk factors are promising targets for dementia prevention. Interrogation of mechanisms underlying dementia risk is difficult in human populations. Studies using diverse experimental models are revealing modifiable dementia risk mechanisms. We review experimental research into 15 modifiable dementia risk factors. Laboratory science can contribute uniquely to dementia prevention.


Asunto(s)
Demencia , Demencia/prevención & control , Humanos , Animales , Factores de Riesgo , Modelos Animales de Enfermedad
2.
Glia ; 71(4): 880-903, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36468604

RESUMEN

Traumatic brain injury (TBI) triggers neuroinflammatory cascades mediated by microglia, which promotes tissue repair in the short-term. These cascades may exacerbate TBI-induced tissue damage and symptoms in the months to years post-injury. However, the progression of the microglial function across time post-injury and whether this differs between biological sexes is not well understood. In this study, we examined the microglial proteome at 3-, 7-, or 28-days after a midline fluid percussion injury (mFPI) in male and female mice using label-free quantitative proteomics. Data are available via ProteomeXchange with identifier PXD033628. We identified a reduction in microglial proteins involved with clearance of neuronal debris via phagocytosis at 3- and 7-days post-injury. At 28 days post-injury, pro-inflammatory proteins were decreased and anti-inflammatory proteins were increased in microglia. These results indicate a reduction in microglial clearance of neuronal debris in the days post-injury with a shift to anti-inflammatory function by 28 days following TBI. The changes in the microglial proteome that occurred across time post-injury did not differ between biological sexes. However, we did identify an increase in microglial proteins related to pro-inflammation and phagocytosis as well as insulin and estrogen signaling in males compared with female mice that occurred with or without a brain injury. Although the microglial response was similar between males and females up to 28 days following TBI, biological sex differences in the microglial proteome, regardless of TBI, has implications for the efficacy of treatment strategies targeting the microglial response post-injury.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Femenino , Ratones , Masculino , Animales , Microglía/metabolismo , Proteoma/metabolismo , Proteómica , Traumatismos Difusos del Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Ratones Endogámicos C57BL
3.
Glia ; 71(8): 1847-1869, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36994950

RESUMEN

Cerebral blood flow (CBF) is important for the maintenance of brain function and its dysregulation has been implicated in Alzheimer's disease (AD). Microglia associations with capillaries suggest they may play a role in the regulation of CBF or the blood-brain-barrier (BBB). We explored the relationship between microglia and pericytes, a vessel-resident cell type that has a major role in the control of CBF and maintenance of the BBB, discovering a spatially distinct subset of microglia that closely associate with pericytes. We termed these pericyte-associated microglia (PEM). PEM are present throughout the brain and spinal cord in NG2DsRed × CX3 CR1+/GFP mice, and in the human frontal cortex. Using in vivo two-photon microscopy, we found microglia residing adjacent to pericytes at all levels of the capillary tree and found they can maintain their position for at least 28 days. PEM can associate with pericytes lacking astroglial endfeet coverage and capillary vessel width is increased beneath pericytes with or without an associated PEM, but capillary width decreases if a pericyte loses a PEM. Deletion of the microglia fractalkine receptor (CX3 CR1) did not disrupt the association between pericytes and PEM. Finally, we found the proportion of microglia that are PEM declines in the superior frontal gyrus in AD. In summary, we identify microglia that specifically associate with pericytes and find these are reduced in number in AD, which may be a novel mechanism contributing to vascular dysfunction in neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Pericitos , Ratones , Humanos , Animales , Pericitos/metabolismo , Ratones Transgénicos , Microglía , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Enfermedad de Alzheimer/metabolismo
4.
Neuroendocrinology ; 112(11): 1027-1038, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35279657

RESUMEN

Increased intake of highly processed, energy-dense foods combined with a sedentary lifestyle are helping fuel the current overweight and obesity crisis, which is more prevalent in women than in men. Although peripheral organs such as adipose tissue contribute to the physiological development of obesity, emerging work aims to understand the role of the central nervous system to whole-body energy homeostasis and development of weight gain and obesity. The present review discusses the impact of insulin, insulin resistance, free fatty acids, and inflammation on brain function and how these differ between the males and females in the context of obesity. We highlight the potential of microglia, the resident immune cells in the brain, as mediators of neuronal insulin resistance that drive reduced satiety, increased food intake, and thus, obesity.


Asunto(s)
Resistencia a la Insulina , Masculino , Femenino , Humanos , Ácidos Grasos no Esterificados , Microglía , Obesidad , Inflamación , Insulina , Encéfalo
5.
Semin Cell Dev Biol ; 94: 96-103, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30826549

RESUMEN

The striking morphology of microglia is one of their most prominent characteristics, with many studies categorising microglial function based on morphology e.g. ramified, hyper-ramified, activated, or amoeboid. Communications regarding rod microglia in neurological disease are scant, and where reported, these cells are rarely the focus of discussion. These factors make it difficult to determine how widespread these cells are not only through the brain but also across diseases. Studies in experimental diffuse brain injury are the first reports of not only significant numbers of rod microglia, but distinct arrangements of these cells, reminiscent of carriages of a train. This review summarises the available reports of rod microglia in vivo and rod-like microglia in vitro and eludes to possible functions and signalling cascades that may evoke this distinct morphology. More investigations are required to fully elucidate the function that rod microglia play in neurological diseases.


Asunto(s)
Microglía/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Animales , Humanos , Microglía/patología , Enfermedades del Sistema Nervioso/patología
6.
Acta Neuropathol ; 137(5): 731-755, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30535946

RESUMEN

This review recounts the definitions and research evidence supporting the multifaceted roles of neuroinflammation in the injured brain following trauma. We summarise the literature fluctuating from the protective and detrimental properties that cytokines, leukocytes and glial cells play in the acute and chronic stages of TBI, including the intrinsic factors that influence cytokine responses and microglial functions relative to genetics, sex, and age. We elaborate on the pros and cons that cytokines, chemokines, and microglia play in brain repair, specifically neurogenesis, and how such conflicting roles may be harnessed therapeutically to sustain the survival of new neurons. With a brief review of the clinical and experimental findings demonstrating early and chronic inflammation impacts on outcomes, we focus on the clinical conditions that may be amplified by neuroinflammation, ranging from acute seizures to chronic epilepsy, neuroendocrine dysfunction, dementia, depression, post-traumatic stress disorder and chronic traumatic encephalopathy. Finally, we provide an overview of the therapeutic agents that have been tested to reduce inflammation-driven secondary pathological cascades and speculate the future promise of alternative drugs.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Inflamación/fisiopatología , Animales , Lesiones Traumáticas del Encéfalo/epidemiología , Lesiones Traumáticas del Encéfalo/terapia , Humanos , Inflamación/epidemiología , Inflamación/terapia , Neuroinmunomodulación
7.
Brain Behav Immun ; 59: 1-7, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26975888

RESUMEN

A myriad of factors influence the developmental and aging process and impact health and life span. Mounting evidence indicates that brain injury, even moderate injury, can lead to lifetime of physical and mental health symptoms. Therefore, the purpose of this mini-review is to discuss how recovery from traumatic brain injury (TBI) depends on age-at-injury and how aging with a TBI affects long-term recovery. TBI initiates pathophysiological processes that dismantle circuits in the brain. In response, reparative and restorative processes reorganize circuits to overcome the injury-induced damage. The extent of circuit dismantling and subsequent reorganization depends as much on the initial injury parameters as other contributing factors, such as genetics and age. Age-at-injury influences the way the brain is able to repair itself, as a result of developmental status, extent of cellular senescence, and injury-induced inflammation. Moreover, endocrine dysfunction can occur with TBI. Depending on the age of the individual at the time of injury, endocrine dysfunction may disrupt growth, puberty, influence social behaviors, and possibly alter the inflammatory response. In turn, activation of microglia, the brain's immune cells, after injury may continue to fuel endocrine dysfunction. With age, the immune system develops and microglia become primed to subsequent challenges. Sustained inflammation and microglial activation can continue for weeks to months post-injury. This prolonged inflammation can influence developmental processes, behavioral performance and age-related decline. Overall, brain injury may influence the aging process and expedite glial and neuronal alterations that impact mental health.


Asunto(s)
Envejecimiento/psicología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Enfermedades del Sistema Endocrino/patología , Enfermedades del Sistema Endocrino/psicología , Microglía/patología , Anciano , Anciano de 80 o más Años , Animales , Humanos , Inflamación/patología , Inflamación/psicología
8.
Dev Neurosci ; 38(3): 195-205, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27449121

RESUMEN

Development and aging are influenced by external factors with the potential to impact health throughout the life span. Traumatic brain injury (TBI) can initiate and sustain a lifetime of physical and mental health symptoms. Over 1.7 million TBIs occur annually in the USA alone, with epidemiology suggesting a higher incidence for young age groups. Additionally, increasing life spans mean more years to age with TBI. While there is ongoing research of experimental pediatric and adult TBI, few studies to date have incorporated animal models of pediatric, adolescent, and adult TBI to understand the role of age at injury across the life span. Here, we explore repeated behavioral performance between rats exposed to diffuse TBI at five different ages. Our aim was to follow neurological morbidities across the rodent life span with respect to age at injury. A single cohort of male Sprague-Dawley rats (n = 69) was received at postnatal day (PND) 10. Subgroups of this cohort (n = 11-12/group) were subjected to a single moderate midline fluid percussion injury at age PND 17, PND 35, 2 months, 4 months, or 6 months. A control group of naïve rats (n = 12) was assembled from this cohort. The entire cohort was assessed for motor function by beam walk at 1.5, 3, 5, and 7 months of age. Anxiety-like behavior was assessed with the open field test at 8 months of age. Cognitive performance was assessed using the novel object location task at 8, 9, and 10 months of age. Depression-like behavior was assessed using the forced swim test at 10 months of age. Age at injury and time since injury differentially influenced motor, cognitive, and affective behavioral outcomes. Motor and cognitive deficits occurred in rats injured at earlier developmental time points, but not in rats injured in adulthood. In contrast, rats injured during adulthood showed increased anxiety-like behavior compared to uninjured control rats. A single diffuse TBI did not result in chronic depression-like behaviors or changes in body weight among any groups. The interplay of age at injury and aging with an injury are translationally important factors that influence behavioral performance as a quality of life metric. More complete understanding of these factors can direct rehabilitative efforts and personalized medicine for TBI survivors.


Asunto(s)
Ansiedad/fisiopatología , Traumatismos Difusos del Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Trastornos del Conocimiento/fisiopatología , Envejecimiento , Animales , Conducta Animal/fisiología , Masculino , Aprendizaje por Laberinto , Ratas Sprague-Dawley
9.
Brain Inj ; 30(2): 217-24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26646974

RESUMEN

PRIMARY OBJECTIVE: A dynamic relationship exists between diffuse traumatic brain injury and changes to the neurovascular unit. The purpose of this study was to evaluate vascular changes during the first week following diffuse TBI. It was hypothesized that pathology is associated with modification of the vasculature. METHODS: Male Sprague-Dawley rats underwent either midline fluid percussion injury or sham-injury. Brain tissue was collected 1, 2 or 7 days post-injury or sham-injury (n = 3/time point). Tissue was collected and stained by de Olmos amino-cupric silver technique to visualize neuropathology or animals were perfused with AltaBlue casting resin before high-resolution vascular imaging. The average volume, surface area, radius, branching and tortuosity of the vessels were evaluated across three regions of interest. RESULTS: In M2, average vessel volume (p < 0.01) and surface area (p < 0.05) were significantly larger at 1 day relative to 2 days, 7 days and sham. In S1BF and VPM, no significant differences in the average vessel volume or surface area at any of the post-injury time points were observed. No significant changes in average radius, branching or tortuosity were observed. CONCLUSIONS: Preliminary findings suggest gross morphological changes within the vascular network likely represent an acute response to mechanical forces of injury, rather than delayed or chronic pathological processes.


Asunto(s)
Traumatismos Difusos del Encéfalo/fisiopatología , Animales , Encéfalo/patología , Traumatismos Difusos del Encéfalo/anatomía & histología , Traumatismos Difusos del Encéfalo/lesiones , Modelos Animales de Enfermedad , Masculino , Neuropatología , Ratas , Ratas Sprague-Dawley
10.
Metab Brain Dis ; 30(2): 393-400, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24733573

RESUMEN

The brain is comprised of neurons and its support system including astrocytes, glial cells and microglia, thereby forming neurovascular units. Neurons require support from glial cells to establish and maintain functional circuits, but microglia are often overlooked. Microglia function as the immune cell of the central nervous system, acting to monitor the microenvironment for changes in signaling, pathogens and injury. More recently, other functional roles for microglia within the healthy brain have been identified, including regulating synapse formation, elimination and function. This review aims to highlight and discuss these alternate microglial roles in the healthy and in contrast, diseased brain with a focus on two acute neurological diseases, traumatic brain injury and epilepsy. In these conditions, microglial roles in synaptic stripping and stabilization as part of neuronal:glial interactions may position them as mediators of the transition between injury-induced circuit dismantling and subsequent reorganization. Increased understanding of microglia roles could identify therapeutic targets to mitigate the consequences of neurological disease.


Asunto(s)
Microglía/fisiología , Enfermedades del Sistema Nervioso/patología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/patología , Traumatismos del Sistema Nervioso/patología , Animales , Humanos , Sinapsis
11.
Curr Protoc ; 3(5): e791, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37222240

RESUMEN

In recent decades, mounting evidence has shown that microglia play a vital role in maintaining synapses throughout life. This maintenance is done via numerous microglial processes, which are long, thin, and highly motile protrusions from the cell body that monitor their environment. However, due to the brevity of the contacts and the potentially transient nature of synaptic structures, establishing the underlying dynamics of this relationship has proven difficult. This article describes a method of using rapidly acquired multiphoton microscopy images to track microglial dynamics and microglia:synapse interactions and the fate of the synaptic structures following those interactions. First, we detail a method for capturing multiphoton images at 1-min intervals for approximately 1 hr and how that process can be done at multiple time points. We then discuss how best to prevent and account for any drifting of the region of interest that can occur during the imaging session and how to remove excessive background noise from those images. Finally, we detail the annotation process for dendritic spines and microglial processes using plugins in MATLAB and Fiji, respectively. These semi-automated plugins allow tracking of individual cell structures, even if both microglia and neurons are imaged in the same fluorescent channel. This protocol presents a method of tracking both microglial dynamics and synaptic structures, in the same animal, at multiple time points, giving the user information on process speed, branching, tip size, location, and dwell time, as well as any dendritic spine gains, losses, and size changes. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Rapid multiphoton image capture Basic Protocol 2: Image preparation using MATLAB and Fiji Basic Protocol 3: Dendritic spine and microglial processes annotation using ScanImage and TrackMate.


Asunto(s)
Espinas Dendríticas , Microglía , Animales , Microscopía , Cuerpo Celular , Colorantes
12.
J Neuroinflammation ; 9: 247, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23111107

RESUMEN

BACKGROUND: Since their discovery, the morphology of microglia has been interpreted to mirror their function, with ramified microglia constantly surveying the micro-environment and rapidly activating when changes occur. In 1899, Franz Nissl discovered what we now recognize as a distinct microglial activation state, microglial rod cells (Stäbchenzellen), which he observed adjacent to neurons. These rod-shaped microglia are typically found in human autopsy cases of paralysis of the insane, a disease of the pre-penicillin era, and best known today from HIV-1-infected brains. Microglial rod cells have been implicated in cortical 'synaptic stripping' but their exact role has remained unclear. This is due at least in part to a scarcity of experimental models. Now we have noted these rod microglia after experimental diffuse brain injury in brain regions that have an associated sensory sensitivity. Here, we describe the time course, location, and surrounding architecture associated with rod microglia following experimental diffuse traumatic brain injury (TBI). METHODS: Rats were subjected to a moderate midline fluid percussion injury (mFPI), which resulted in transient suppression of their righting reflex (6 to 10 min). Multiple immunohistochemistry protocols targeting microglia with Iba1 and other known microglia markers were undertaken to identify the morphological activation of microglia. Additionally, labeling with Iba1 and cell markers for neurons and astrocytes identified the architecture that surrounds these rod cells. RESULTS: We identified an abundance of Iba1-positive microglia with rod morphology in the primary sensory barrel fields (S1BF). Although present for at least 4 weeks post mFPI, they developed over the first week, peaking at 7 days post-injury. In the absence of contusion, Iba1-positive microglia appear to elongate with their processes extending from the apical and basal ends. These cells then abut one another and lay adjacent to cytoarchitecture of dendrites and axons, with no alignment with astrocytes and oligodendrocytes. Iba1-positive rod microglial cells differentially express other known markers for reactive microglia including OX-6 and CD68. CONCLUSION: Diffuse traumatic brain injury induces a distinct rod microglia morphology, unique phenotype, and novel association between cells; these observations entice further investigation for impact on neurological outcome.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Microglía/fisiología , Corteza Somatosensorial/patología , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Análisis de Fourier , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Microglía/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Ratas , Ratas Sprague-Dawley
13.
Neurobiol Aging ; 105: 340-348, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34174592

RESUMEN

Tauopathies are a group of neurodegenerative diseases that involve pathological changes to the tau protein. Neuroinflammation is a commonly reported feature of tauopathies that has been demonstrated to exacerbate tau pathology and, hence, neurodegeneration. Microglia can mediate the inflammatory response in order to maintain brain homeostasis. In the aged brain, microglia are reported to undergo morphological and functional changes, adopting a pro-inflammatory profile and loss of homeostatic functions. Dystrophic and dysfunctional microglia are associated with tau pathology in the healthy and diseased brain which is proposed to contribute to disease development and progression. Microglia have also been recently demonstrated to possess sexually dimorphic roles in the developing, adult and aged brain. The sex differences in microglial functionality suggest that microglia may contribute to tauopathies which may differ between sexes. This review highlights the detrimental loop between age-related microglial changes and tau pathology with implications for microglial sexual dichotomy.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Microglía/patología , Microglía/fisiología , Caracteres Sexuales , Tauopatías/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Progresión de la Enfermedad , Femenino , Homeostasis , Humanos , Inflamación , Masculino , Ratones
14.
Front Neurol ; 12: 722526, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566867

RESUMEN

Traumatic brain injury (TBI) can occur at any age, from youth to the elderly, and its contribution to age-related neuropathology remains unknown. Few studies have investigated the relationship between age-at-injury and pathophysiology at a discrete biological age. In this study, we report the immunohistochemical analysis of naïve rat brains compared to those subjected to diffuse TBI by midline fluid percussion injury (mFPI) at post-natal day (PND) 17, PND35, 2-, 4-, or 6-months of age. All brains were collected when rats were 10-months of age (n = 6-7/group). Generalized linear mixed models were fitted to analyze binomial proportion and count data with R Studio. Amyloid precursor protein (APP) and neurofilament (SMI34, SMI32) neuronal pathology were counted in the corpus callosum (CC) and primary sensory barrel field (S1BF). Phosphorylated TAR DNA-binding protein 43 (pTDP-43) neuropathology was counted in the S1BF and hippocampus. There was a significantly greater extent of APP and SMI34 axonal pathology and pTDP-43 neuropathology following a TBI compared with naïves regardless of brain region or age-at-injury. However, age-at-injury did determine the extent of dendritic neurofilament (SMI32) pathology in the CC and S1BF where all brain-injured rats exhibited a greater extent of pathology compared with naïve. No significant differences were detected in the extent of astrocyte activation between brain-injured and naïve rats. Microglia counts were conducted in the S1BF, hippocampus, ventral posteromedial (VPM) nucleus, zona incerta, and posterior hypothalamic nucleus. There was a significantly greater proportion of deramified microglia, regardless of whether the TBI was recent or remote, but this only occurred in the S1BF and hippocampus. The proportion of microglia with colocalized CD68 and TREM2 in the S1BF was greater in all brain-injured rats compared with naïve, regardless of whether the TBI was recent or remote. Only rats with recent TBI exhibited a greater proportion of CD68-positive microglia compared with naive in the hippocampus and posterior hypothalamic nucleus. Whilst, only rats with a remote brain-injury displayed a greater proportion of microglia colocalized with TREM2 in the hippocampus. Thus, chronic alterations in neuronal and microglial characteristics are evident in the injured brain despite the recency of a diffuse brain injury.

15.
Neurobiol Dis ; 40(2): 394-403, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20621186

RESUMEN

The contribution of infiltrated neutrophils to secondary damage following traumatic brain injury remains controversial. Chemokines that regulate neutrophil migration by signaling through the CXCR2 receptor are markedly elevated by brain injury and are associated with the propagation of secondary damage. This study thus investigated the function of CXCR2 in posttraumatic inflammation and secondary degeneration by examining Cxcr2-deficient (Cxcr2(-/-)) mice over 14 days following closed head injury (CHI). We demonstrate a significant attenuation of neutrophil infiltration in Cxcr2(-/-) mice at 12 hours and 7 days after CHI, despite increased levels of CXC neutrophil-attracting chemokines in the lesioned cortex. This coincides with reduced tissue damage, neuronal loss, and cell death in Cxcr2(-/-) mice compared to wild-type controls, with heterozygotes showing intermediate responses. In contrast, blood-brain barrier permeability and functional recovery did not appear to be affected by Cxcr2 deletion. This study highlights the deleterious contribution of neutrophils to posttraumatic neurodegeneration and demonstrates the importance of CXC chemokine signaling in this process. Therefore, CXCR2 antagonistic therapeutics currently in development for other inflammatory conditions may also be of benefit in posttraumatic neuroinflammation.


Asunto(s)
Corteza Cerebral/inmunología , Traumatismos Cerrados de la Cabeza/inmunología , Infiltración Neutrófila/inmunología , Receptores de Interleucina-8B/deficiencia , Factores de Edad , Animales , Barrera Hematoencefálica/patología , Muerte Celular , Corteza Cerebral/patología , Quimiocinas/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Traumatismos Cerrados de la Cabeza/patología , Heterocigoto , Homocigoto , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Interleucina-8B/genética , Recuperación de la Función
16.
Molecules ; 15(9): 6598-618, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20877247

RESUMEN

Injury to the central nervous system initiates complex physiological, cellular and molecular processes that can result in neuronal cell death. Of interest to this review is the activation of the kinin family of neuropeptides, in particular bradykinin and substance P. These neuropeptides are known to have a potent pro-inflammatory role and can initiate neurogenic inflammation resulting in vasodilation, plasma extravasation and the subsequent development of edema. As inflammation and edema play an integral role in the progressive secondary injury that causes neurological deficits, this review critically examines kinin receptor antagonists as a potential neuroprotective intervention for acute brain injury, and more specifically, traumatic brain and spinal cord injury and stroke.


Asunto(s)
Antagonistas de los Receptores de Bradiquinina , Antagonistas del Receptor de Neuroquinina-1 , Fármacos Neuroprotectores , Sistema Nervioso Central/lesiones , Sistema Nervioso Central/patología , Edema/tratamiento farmacológico , Humanos , Inflamación/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología
17.
J Alzheimers Dis ; 77(4): 1765-1781, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32894243

RESUMEN

BACKGROUND: Microglia are traditionally described as the immune cells of the brain and have an inflammatory role in Alzheimer's disease (AD). Microglial morphological and phenotypic shifts in AD have not been fully characterized; however, microglia are often described as either pro- or anti-inflammatory. OBJECTIVE: To determine microglial if microglial morphology and phenotype changes with disease status. METHODS: This study observed morphology through Iba1 immunohistochemistry on tissue sections encompassing the primary motor cortex and somatosensory barrel fields. Immunohistochemistry for pro-inflammatory markers: CD14 and CD40; and anti-inflammatory markers: CD16 and TREM2, was performed at 3, 6, and 12 months of age which correlated with pre-plaque, onset, and significant plaque load in APP/PS1 brains (n = 6) and compared to age-matched littermate controls (n = 6). RESULTS: Microglia demonstrated a defined morphological shift with time. Deramified morphologies increased in the APP/PS1, at both 6 months (p < 0.0001) and 12 months (p < 0.0001). At 12 months, there were significantly lower numbers of ramified microglia (p < 0.001). Results indicated that microglia have a heterogenic marker immunoreactivity as CD16, TREM2, and CD40 were associated with an activated morphology at the same time points. All inflammatory markers were significantly upregulated at 12 months in the APP/PS1 mice (TREM2 (F (2,30) â€Š= 10.75, p = 0.0003), CD40 (F (2,30) â€Š= 15.86, p < 0.0001), CD14 (F (2,30) â€Š= 6.84, p = 0.0036), and CD16 (F (2,30) â€Š= 3.026, p = 0.0635)). CONCLUSION: Our data indicate that pro- and anti-inflammatory factors of microglia occur in APP/PS1 mice.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Presenilina-1/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Masculino , Ratones , Ratones Transgénicos , Microglía/patología
18.
J Neurotrauma ; 37(5): 782-791, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32046575

RESUMEN

Traumatic brain injury (TBI) can cause persistent cognitive changes and ongoing neurodegeneration in the brain. Accumulating epidemiological and pathological evidence implicates TBI in the development of Alzheimer's disease, the most common cause of dementia. Further, the TBI-induced form of dementia, called chronic traumatic encephalopathy, shares many pathological hallmarks present in multiple different diseases which cause dementia. The inflammatory and neuritic responses to TBI and dementia overlap, indicating that they may share common pathological mechanisms and that TBI may ultimately cause a pathological cascade culminating in the development of dementia. This review explores Australian pre-clinical research investigating the pathological links between TBI and dementia.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Encéfalo/patología , Demencia/patología , Animales , Australia , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Demencia/etiología , Demencia/metabolismo , Humanos , Microglía/metabolismo , Microglía/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteínas tau/metabolismo
19.
Neuroscience ; 408: 282-292, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30999032

RESUMEN

Environmental enrichment (EE) has been consistently reported to enhance cognitive function in mouse models of neuropathology. Microglia, implicated in Alzheimer's disease pathology, may mediate this effect. The aim of the present study was to investigate the effect of EE on cognitive function and microglia in mouse models of aging and amyloidosis. Male wild-type (Wt) and APP/PS1 mice were randomly assigned to standard housing (SH) or EE from 12 to 18 months of age. Spatial memory testing was performed using the Y and Barnes maze. Immunohistochemical analysis of Aß load, Iba1 and CD-68-labeled (phagocytic-type) microglia was examined between conditions. EE from 12 months of age was associated with improved short-term memory performance in APP/PS1 mice, despite no reductions to Aß load. APP/PS1 mice in SH had significantly increased microglia occupying the neocortex and hippocampus (p = 0.02; p = 0.004, respectively) relative to Wt animals. Microglia labeling was not statistically different between EE-exposed APP/PS1 compared to Wt mice, indicating that EE may attenuate the increased microglial load in aging APP/PS1 mice. APP/PS1 mice from EE had significantly (p = 0.01) higher colocalization of Iba1 and CD-68 labeling, indicative of increased phagocytic microglia compared to mice from SH. The findings of the present study suggest that EE after substantial brain amyloidosis, has the potential to preserve domains of cognitive function, while having no effect on Aß deposition. The current study demonstrates that EE may attenuate microglia in aging APP/PS1 mice, and may promote alterations in cellular phenotype.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Ambiente , Vivienda para Animales , Memoria a Corto Plazo/fisiología , Microglía/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
20.
Neuroscience ; 359: 209-223, 2017 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-28736137

RESUMEN

Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.


Asunto(s)
Traumatismos Difusos del Encéfalo/metabolismo , Encefalitis/metabolismo , Microglía/metabolismo , Receptor Nogo 1/metabolismo , Animales , Modelos Animales de Enfermedad , Encefalitis/complicaciones , Masculino , Receptor Nogo 1/antagonistas & inhibidores , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA