Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Brain Behav Immun ; 114: 287-298, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37648007

RESUMO

The circadian system is an evolutionarily adaptive system that synchronizes biological and physiological activities within the body to the 24 h oscillations on Earth. At the molecular level, circadian clock proteins are transcriptional factors that regulate the rhythmic expression of genes involved in numerous physiological processes such as sleep, cognition, mood, and immune function. Environmental and genetic disruption of the circadian clock can lead to pathology. For example, global deletion of the circadian clock gene Rev-erbα (RKO) leads to hyperlocomotion, increased anxiety-like behaviors, and cognitive impairments in male mice; however, the mechanisms underlying behavioral changes remain unclear. Here we hypothesized that RKO alters microglia function leading to neuroinflammation and altered mood and cognition, and that microglia depletion can resolve neuroinflammation and restore behavior. We show that microglia depletion (CSF1R inhibitor, PLX5622) in 8-month-old RKO mice ameliorated hyperactivity, memory impairments, and anxiety/risky-like behaviors. RKO mice exhibited striking increases in expression of pro-inflammatory cytokines (e.g., IL-1ß and IL-6). Surprisingly, these increases were only fully reversed by microglia depletion in the male but not female RKO hippocampus. In contrast, male RKO mice showed greater alterations in microglial morphology and phagocytic activity than females. In both sexes, microglia depletion reduced microglial branching and decreased CD68 production without altering astrogliosis. Taken together, we show that male and female RKO mice exhibit unique perturbations to the neuroimmune system, but microglia depletion is effective at rescuing aspects of behavioral changes in both sexes. These results demonstrate that microglia are involved in Rev-erbα-mediated changes in behavior and neuroinflammation.


Assuntos
Disfunção Cognitiva , Microglia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , Animais , Feminino , Masculino , Camundongos , Ansiedade , Ritmo Circadiano/fisiologia , Cognição , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Camundongos Knockout , Microglia/metabolismo , Doenças Neuroinflamatórias , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo
2.
Brain Behav Immun ; 114: 438-452, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37709153

RESUMO

Aging is associated with a significant shift in immune system reactivity ("inflammaging"), as basal inflammation increases but protective responses to infection are compromised. The immune system exhibits considerable sex differences, which may influence the process of inflammaging, including immune cell activation and behavioral consequences of immune signaling (i.e., impaired memory). Here, we test the hypothesis that sex differences in immune aging may mediate sex differences in cognitive decline. Aged male and female rats received peripheral immune stimulation using lipopolysaccharide (LPS), then molecular, cellular, and behavioral outcomes were assessed. We observed that LPS-treated aged male rats showed cognitive impairment and increased neuroinflammatory responses relative to adult males. In contrast, aged female rats did not display these aging-related deficits. Using transcriptomic and flow cytometry analyses, we further observed significant age- and sex- dependent changes in immune cell populations in the brain parenchyma and meninges, indicating a broad shift in the neuroinflammatory environment that may potentiate these behavioral effects. Ovariectomized aged female rats were also resistant to inflammation-induced memory deficits, indicating that ovarian hormones are not required for the attenuated neuroinflammation in aged females. Overall, our results indicate that males have amplified inflammatory priming with age, which contributes to age-associated cognitive decline. Our findings highlight sexual dimorphism in mechanisms of aging, and suggest that sex is a crucial consideration for identifying therapies for aging and neuroinflammation.


Assuntos
Disfunção Cognitiva , Microglia , Ratos , Animais , Feminino , Masculino , Caracteres Sexuais , Doenças Neuroinflamatórias , Lipopolissacarídeos/farmacologia , Inflamação
3.
Brain Behav Immun ; 103: 130-144, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35447300

RESUMO

Social status is a critical factor determining health outcomes in human and nonhuman social species. In social hierarchies with reproductive skew, individuals compete to monopolize resources and increase mating opportunities. This can come at a significant energetic cost leading to trade-offs between different physiological systems. In particular, changes in energetic investment in the immune system can have significant short and long-term effects on fitness and health. We have previously found that dominant alpha male mice living in social hierarchies have increased metabolic demands related to territorial defense. In this study, we tested the hypothesis that high-ranking male mice favor adaptive immunity, while subordinate mice show higher investment in innate immunity. We housed 12 groups of 10 outbred CD-1 male mice in a social housing system. All formed linear social hierarchies and subordinate mice had higher concentrations of plasma corticosterone (CORT) than alpha males. This difference was heightened in highly despotic hierarchies. Using flow cytometry, we found that dominant status was associated with a significant shift in immunophenotypes towards favoring adaptive versus innate immunity. Using Tag-Seq to profile hepatic and splenic transcriptomes of alpha and subordinate males, we identified genes that regulate metabolic and immune defense pathways that are associated with status and/or CORT concentration. In the liver, dominant animals showed a relatively higher expression of specific genes involved in major urinary production and catabolic processes, whereas subordinate animals showed relatively higher expression of genes promoting biosynthetic processes, wound healing, and proinflammatory responses. In spleen, subordinate mice showed relatively higher expression of genes facilitating oxidative phosphorylation and DNA repair and CORT was negatively associated with genes involved in lymphocyte proliferation and activation. Together, our findings suggest that dominant and subordinate animals adaptively shift immune profiles and peripheral gene expression to match their contextual needs.


Assuntos
Hierarquia Social , Predomínio Social , Animais , Corticosterona , Masculino , Camundongos , Transcriptoma
4.
Brain Behav Immun ; 97: 365-370, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34284114

RESUMO

Spinal cord injury (SCI) elicits chronic pain in 65% of individuals. In addition, SCI afflicts an increasing number of aged individuals, and those with SCI are predisposed to shorter lifespan. Our group previously identified that deletion of the microRNA miR-155 reduced neuroinflammation and locomotor deficits after SCI. Here, we hypothesized that aged mice would be more susceptible to pain symptoms and death soon after SCI, and that miR-155 deletion would reduce pain symptoms in adult and aged mice and improve survival. Adult (2 month-old) and aged (20 month-old) female wildtype (WT) and miR-155 knockout (KO) mice received T9 contusion SCI. Aged WT mice displayed reduced survival and increased autotomy - a symptom of spontaneous pain. In contrast, aged miR-155 KO mice after SCI were less susceptible to death or spontaneous pain. Evoked pain symptoms were tested using heat (Hargreaves test) and mechanical (von Frey) stimuli. At baseline, aged mice showed heightened heat sensitivity. After SCI, adult and aged WT and miR-155 KO mice all exhibited heat and mechanical hypersensitivity at all timepoints. miR-155 deletion in adult (but not aged) mice reduced mechanical hypersensitivity at 7 and 14 d post-SCI. Therefore, aging predisposes mice to SCI-elicited spontaneous pain and expedited mortality. miR-155 deletion in adult mice reduces evoked pain symptoms, and miR-155 deletion in aged mice reduces spontaneous pain and expedited mortality post-SCI. This study highlights the importance of studying geriatric models of SCI, and that inflammatory mediators such as miR-155 are promising targets after SCI for improving pain relief and longevity.


Assuntos
MicroRNAs , Neuralgia , Traumatismos da Medula Espinal , Envelhecimento , Animais , Modelos Animais de Doenças , Feminino , Hiperalgesia , Camundongos , Camundongos Knockout , MicroRNAs/genética , Medula Espinal , Traumatismos da Medula Espinal/complicações
5.
Brain Behav Immun ; 73: 133-148, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30009999

RESUMO

Over the course of an animal's lifespan, there is a protracted breakdown in basic homeostatic functions. Stressors (both psychological and physiological) can accelerate this process and compromise multiple homeostatic mechanisms. For example, both stress and aging can modulate neuroinflammatory function and cause a primed phenotype resulting in a heightened neuroinflammatory profile upon immune activation. Microglia, the brain's resident myeloid cell, produce "silent" immune machinery in response to stress and aging that does not cause immediate immune activation; rather, these changes prime the cell for a subsequent immune insult. Primed microglia exhibit a hyperinflammatory response upon immune activation that can exacerbate pathology. In this review, we will explore parallels between stress- and aging-induced neuroinflammatory priming. First, we will provide a background on the basic principles of neuroimmunology. Next, we will discuss evidence that neuroinflammatory responses become primed in the context of both stress and aging. We will also describe cell-specific contributions to neuroinflammatory priming with a focus on microglia. Finally, common mechanisms underlying priming in the context of stress and aging will be discussed: these mechanisms include glucocorticoid signaling; accumulation of danger signals; dis-inhibition of microglia; and breakdown of circadian rhythms. Overall, there are multifarious parallels between stress- and aging-elicited neuroinflammatory priming, suggesting that stress may promote a form of premature aging. Further unravelling mechanisms underlying priming could lead to improved treatments for buffering against stress- and aging-elicited behavioral pathologies.


Assuntos
Envelhecimento/imunologia , Neuroimunomodulação/fisiologia , Estresse Psicológico/imunologia , Envelhecimento/fisiologia , Animais , Encéfalo/metabolismo , Ritmo Circadiano/fisiologia , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Humanos , Inflamação/metabolismo , Microglia/metabolismo , Microglia/fisiologia
6.
Brain Behav Immun ; 70: 257-267, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29524458

RESUMO

Exposure to stressors can enhance neuroinflammatory responses, and both stress and neuroinflammation are predisposing factors in the development of psychiatric disorders. Females suffer disproportionately more from several psychiatric disorders, yet stress-induced changes in neuroinflammation have primarily been studied in males. Here we tested whether exposure to inescapable tail shock sensitizes or 'primes' neuroinflammatory responses in male and female rats. At 24 h post-stress, male and female rats exposed to a peripheral immune challenge enhanced neuroinflammatory responses and exacerbated anxiety- and depressive-like behaviors. These changes are likely glucocorticoid dependent, as administering exogenous CORT, caused a similar primed inflammatory response in the hippocampus of male and female rats. Further, stress disinhibited anti-inflammatory signaling mechanisms (such as CD200R) in the hippocampus of male and female rats. In males, microglia are considered the likely cellular source mediating neuroinflammatory priming; stress increased cytokine expression in ex vivo male microglia. Conversely, microglia isolated from stressed or CORT treated females did not exhibit elevated cytokine responses. Microglia isolated from both stressed male and female rats reduced phagocytic activity; however, suggesting that microglia from both sexes experience stress-induced functional impairments. Finally, an immune challenge following either stress or CORT in females, but not males, increased peripheral inflammation (serum IL-1ß). These novel data suggest that although males and females both enhance stress-induced neuroinflammatory and behavioral responses to an immune challenge, this priming may occur through distinct, sex-specific mechanisms.


Assuntos
Citocinas/imunologia , Caracteres Sexuais , Estresse Psicológico/imunologia , Animais , Citocinas/metabolismo , Feminino , Glucocorticoides/metabolismo , Hipocampo/metabolismo , Inflamação/metabolismo , Interleucina-1beta/análise , Lipopolissacarídeos/farmacologia , Masculino , Microglia/metabolismo , Ratos , Ratos Sprague-Dawley
7.
J Neurosci ; 36(32): 8516-32, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27511021

RESUMO

UNLABELLED: Axon regeneration after spinal cord injury (SCI) fails due to neuron-intrinsic mechanisms and extracellular barriers including inflammation. microRNA (miR)-155-5p is a small, noncoding RNA that negatively regulates mRNA translation. In macrophages, miR-155-5p is induced by inflammatory stimuli and elicits a response that could be toxic after SCI. miR-155 may also independently alter expression of genes that regulate axon growth in neurons. Here, we hypothesized that miR-155 deletion would simultaneously improve axon growth and reduce neuroinflammation after SCI by acting on both neurons and macrophages. New data show that miR-155 deletion attenuates inflammatory signaling in macrophages, reduces macrophage-mediated neuron toxicity, and increases macrophage-elicited axon growth by ∼40% relative to control conditions. In addition, miR-155 deletion increases spontaneous axon growth from neurons; adult miR-155 KO dorsal root ganglion (DRG) neurons extend 44% longer neurites than WT neurons. In vivo, miR-155 deletion augments conditioning lesion-induced intraneuronal expression of SPRR1A, a regeneration-associated gene; ∼50% more injured KO DRG neurons expressed SPRR1A versus WT neurons. After dorsal column SCI, miR-155 KO mouse spinal cord has reduced neuroinflammation and increased peripheral conditioning-lesion-enhanced axon regeneration beyond the epicenter. Finally, in a model of spinal contusion injury, miR-155 deletion improves locomotor function at postinjury times corresponding with the arrival and maximal appearance of activated intraspinal macrophages. In miR-155 KO mice, improved locomotor function is associated with smaller contusion lesions and decreased accumulation of inflammatory macrophages. Collectively, these data indicate that miR-155 is a novel therapeutic target capable of simultaneously overcoming neuron-intrinsic and neuron-extrinsic barriers to repair after SCI. SIGNIFICANCE STATEMENT: Axon regeneration after spinal cord injury (SCI) fails due to neuron-intrinsic mechanisms and extracellular barriers, including inflammation. Here, new data show that deleting microRNA-155 (miR-155) affects both mechanisms and improves repair and functional recovery after SCI. Macrophages lacking miR-155 have altered inflammatory capacity, which enhances neuron survival and axon growth of cocultured neurons. In addition, independent of macrophages, adult miR-155 KO neurons show enhanced spontaneous axon growth. Using either spinal cord dorsal column crush or contusion injury models, miR-155 deletion improves indices of repair and recovery. Therefore, miR-155 has a dual role in regulating spinal cord repair and may be a novel therapeutic target for SCI and other CNS pathologies.


Assuntos
MicroRNAs/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Regeneração da Medula Espinal/genética , Animais , Axônios , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Gânglios Espinais/citologia , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/genética , Proteínas dos Microfilamentos/metabolismo , Neuritos , Neurônios/fisiologia , Receptores de Superfície Celular/metabolismo , Neuropatia Ciática/genética , Medula Espinal/citologia , Fatores de Tempo , Transfecção
8.
Mol Cell Neurosci ; 64: 84-94, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25542813

RESUMO

Galectin (Gal)-1 is a small carbohydrate-binding protein and immune modulatory cytokine that is synthesized locally at the site of peripheral nerve injury. In this environment, Gal1 can promote regeneration of injured peripheral axons, in part by modifying the function of macrophages recruited to the site of injury. Unlike in injured peripheral nerves, macrophages do not promote axon regeneration in the injured central nervous system (CNS), perhaps because Gal1 levels are not regulated appropriately. Because the dynamics and cellular localization of endogenous Gal1 have not been rigorously characterized after CNS injury, we examined the spatio-temporal distribution of Gal1 in rat spinal cords subjected to a standardized contusion injury. Whereas Gal1 was not expressed in uninjured spinal cord, it was significantly upregulated after SCI, especially within the lesion core. Gal1 was expressed in ~40% of lesion-localized macrophages at 3-28 days post-injury (dpi), and in ~45% of astrocytes in the lesion border at 7-28 dpi. Most lesion-localized Gal1+ macrophages did not express the phagocytosis marker ED1, and Gal1+ cells contained less phagocytosed lipids. These data suggest that time- and location-dependent regulation of Gal1 by macrophages (and astrocytes) could be important for modulating phagocytosis, inflammation/gliosis, and axon growth after SCI.


Assuntos
Galectina 1/metabolismo , Macrófagos/metabolismo , Fagocitose , Traumatismos da Medula Espinal/metabolismo , Animais , Astrócitos/metabolismo , Feminino , Galectina 1/genética , Ratos , Ratos Sprague-Dawley , Regeneração da Medula Espinal , Regulação para Cima
9.
bioRxiv ; 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-37745393

RESUMO

Spinal cord injury (SCI) can cause long-lasting locomotor deficits, pain, and mood disorders. Anatomical and functional outcomes are exacerbated by inflammation after SCI, which causes secondary damage. One promising target after SCI is manipulating the circadian system, which optimizes biology and behavior for time of day - including neuroimmune responses and mood-related behaviors. Circadian disruption after SCI is likely worsened by a disruptive hospital environment, which typically includes dim light-at-night (dLAN). Here, we hypothesized that mice subjected to SCI, then placed in dLAN, would exhibit worsened locomotor deficits, pain-like behavior, and anxiety-depressive-like symptoms compared to mice maintained in light days with dark nights (LD). C57BL/6J mice received sham surgery or moderate T9 contusion SCI, then were placed permanently in LD or dLAN. dLAN after SCI did not worsen locomotor deficits; rather, SCI-dLAN mice showed slight improvement in open-field locomotion at the final timepoint. Although dLAN did not alter SCI-induced heat hyperalgesia, SCI-dLAN mice exhibited an increase in mechanical allodynia at 13 days post-SCI compared to SCI-LD mice. SCI-LD and SCI-dLAN mice had similar outcomes using sucrose preference (depressive-like) and open-field (anxiety-like) tests. At 21 dpo, SCI-dLAN mice had reduced preference for a novel juvenile compared to SCI-LD, implying that dLAN combined with SCI may worsen this mood-related behavior. Finally, lesion size was similar between SCI-LD and SCI-dLAN mice. Therefore, newly placing C57BL/6J mice in dLAN after SCI had modest effects on locomotor, pain-like, and mood-related behaviors. Future studies should consider whether clinically-relevant circadian disruptors, alone or in combination, could be ameliorated to enhance outcomes after SCI.

10.
Exp Neurol ; 375: 114725, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38365132

RESUMO

Spinal cord injury (SCI) can cause long-lasting locomotor deficits, pain, and mood disorders. Anatomical and functional outcomes are exacerbated by inflammation after SCI, which causes secondary damage. One promising target after SCI is manipulating the circadian system, which optimizes biology and behavior for time of day - including neuroimmune responses and mood-related behaviors. Circadian disruption after SCI is likely worsened by a disruptive hospital environment, which typically includes dim light-at-night (dLAN). Here, we hypothesized that mice subjected to SCI, then placed in dLAN, would exhibit worsened locomotor deficits, pain-like behavior, and anxiety-depressive-like symptoms compared to mice maintained in light days with dark nights (LD). C57BL/6 J mice received sham surgery or moderate T9 contusion SCI, then were placed permanently in LD or dLAN. dLAN after SCI did not worsen locomotor deficits; rather, SCI-dLAN mice showed slight improvement in open-field locomotion at the final timepoint. Although dLAN did not alter SCI-induced heat hyperalgesia, SCI-dLAN mice exhibited an increase in mechanical allodynia at 13 days post-SCI compared to SCI-LD mice. SCI-LD and SCI-dLAN mice had similar outcomes using sucrose preference (depressive-like) and open-field (anxiety-like) tests. At 21 dpo, SCI-dLAN mice had reduced preference for a novel juvenile compared to SCI-LD, implying that dLAN combined with SCI may worsen this mood-related behavior. Finally, lesion size was similar between SCI-LD and SCI-dLAN mice. Therefore, newly placing C57BL/6 J mice in dLAN after SCI had modest effects on locomotor, pain-like, and mood-related behaviors. Future studies should consider whether clinically-relevant circadian disruptors, alone or in combination, could be ameliorated to enhance outcomes after SCI.


Assuntos
Ritmo Circadiano , Traumatismos da Medula Espinal , Camundongos , Animais , Luz , Camundongos Endogâmicos C57BL , Hiperalgesia/etiologia , Dor , Traumatismos da Medula Espinal/complicações , Medula Espinal
11.
Exp Neurol ; 364: 114382, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36924982

RESUMO

Spinal cord injury (SCI) predisposes individuals to anxiety and chronic pain. Anxiety- and pain-like behavior after SCI can be tested in rodents, yet commonly used tests assess one variable and may not replicate effects of SCI or sex differences seen in humans. Thus, novel preclinical tests should be optimized to better evaluate behaviors relating to anxiety and pain. Here, we use our newly developed conflict test - the Thermal Increments Dark-Light (TIDAL) test - to explore how SCI affects anxiety- vs. pain-like behavior, and whether sex affects post-SCI behavior. The TIDAL conflict test consists of two plates connected by a walkway; one plate remains illuminated and at an isothermic temperature, whereas the other plate is dark but is heated incrementally to aversive temperatures. A control mice thermal place preference test was also performed in which both plates are illuminated. Female and male mice received moderate T9 contusion SCI or remained uninjured. At 7 days post-operative (dpo), mice with SCI increased dark plate preference throughout the TIDAL conflict test compared to uninjured mice. SCI increased dark plate preference for both sexes, although female (vs. male) mice remained on the heated-dark plate to higher temperatures. Mice with SCI that repeated TIDAL at 7 and 21 dpo showed reduced preference for the dark-heated plate at 21 dpo. Overall, in female and male mice, SCI enhances the salience of anxiety (vs. heat sensitivity). The TIDAL conflict test meets a need for preclinical anxiety- and pain-related tests that recapitulate the human condition; thus, future rodent behavioral studies should incorporate TIDAL or other conflict tests to help understand and treat neurologic disorders.


Assuntos
Dor Crônica , Traumatismos da Medula Espinal , Camundongos , Feminino , Masculino , Humanos , Animais , Temperatura Alta , Traumatismos da Medula Espinal/complicações , Ansiedade/etiologia , Transtornos de Ansiedade , Medula Espinal
12.
J Biol Rhythms ; 38(5): 419-446, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37357738

RESUMO

Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.


Assuntos
Relógios Circadianos , Doenças Neurodegenerativas , Animais , Humanos , Longevidade , Ritmo Circadiano/fisiologia , Envelhecimento , Relógios Circadianos/fisiologia , Encéfalo , Mamíferos
13.
J Neurotrauma ; 40(9-10): 833-844, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36719772

RESUMO

Spinal cord injury (SCI) in humans frequently causes intractable chronic pain. Females are susceptible to worse pain than males, and females may show higher pain prevalence after SCI. Despite this difference in the clinical prevalence of SCI pain, few pre-clinical studies have systematically studied sex differences in SCI-elicited pain-related behaviors in rodents. Here, we leverage data from a large cohort of mice to test whether contusion SCI consistently causes pain symptoms in mice, and to establish whether female (vs. male) mice display heightened hypersensitivity after SCI. Mechanical and heat sensory thresholds were assessed using the von Frey and Hargreaves tests, respectively. In an initial experiment, female mice receiving moderate 60 kDyn SCI or moderate-to-severe 75 kDyn SCI at T9 both exhibited mechanical and heat pain symptoms compared with sham controls. A 75 kDyn SCI caused excess motor deficits that confounded defining pain sensitivity at acute times; therefore, the moderate SCI force was used for subsequent experiments. Next, adult female and male C57BL6/J mice received sham surgery or T9 moderate contusion SCI. Comparing female to male mice after SCI, we reveal that mice of both sexes displayed mechanical and heat hypersensitivity compared with sham controls, from acute-to-chronic post-injury times. Females had amplified SCI-elicited hypersensitivity compared with males. Our data suggest that thoracic contusion SCI elicits consistent and persistent pain-associated symptoms, which are more intense in female than in male mice. These results have important implications for uncovering sex-specific mechanisms and therapeutic targets to ameliorate neuropathic pain after SCI.


Assuntos
Contusões , Neuralgia , Dor Intratável , Traumatismos da Medula Espinal , Humanos , Camundongos , Feminino , Masculino , Animais , Hiperalgesia/etiologia , Caracteres Sexuais , Neuralgia/etiologia , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Contusões/complicações
14.
Cell Tissue Res ; 349(1): 201-13, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22592625

RESUMO

After central nervous system (CNS) trauma, axons have a low capacity for regeneration. Regeneration failure is associated with a muted regenerative response of the neuron itself, combined with a growth-inhibitory and cytotoxic post-injury environment. After spinal cord injury (SCI), resident and infiltrating immune cells (especially microglia/macrophages) contribute significantly to the growth-refractory milieu near the lesion. By targeting both the regenerative potential of the axon and the cytotoxic phenotype of microglia/macrophages, we may be able to improve CNS repair after SCI. In this review, we discuss molecules shown to impact CNS repair by affecting both immune cells and neurons. Specifically, we provide examples of pattern recognition receptors, integrins, cytokines/chemokines, nuclear receptors and galectins that could improve CNS repair. In many cases, signaling by these molecules is complex and may have contradictory effects on recovery depending on the cell types involved or the model studied. Despite this caveat, deciphering convergent signaling pathways on immune cells (which affect axon growth indirectly) and neurons (direct effects on axon growth) could improve repair and recovery after SCI. Future studies must continue to consider how regenerative therapies targeting neurons impact other cells in the pathological CNS. By identifying molecules that simultaneously improve axon regenerative capacity and drive the protective, growth-promoting phenotype of immune cells, we may discover SCI therapies that act synergistically to improve CNS repair and functional recovery.


Assuntos
Axônios/imunologia , Axônios/fisiologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/fisiologia , Regeneração Nervosa/imunologia , Transdução de Sinais/imunologia , Animais , Humanos , Receptores de Reconhecimento de Padrão/metabolismo
15.
Curr Opin Neurobiol ; 77: 102649, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36368270

RESUMO

Aging involves progressive deterioration away from homeostasis. Whereas the healthy adult brain maintains neuroimmune cells in a surveillant and homeostatic state, aged glial cells have a hyperreactive phenotype. These age-related pro-inflammatory biases are driven in part by cell-intrinsic factors, including increased cell priming and pro-inflammatory cell states. In addition, the aged inflammatory milieu is shaped by an altered environment, such as amplified danger signals and cytokines and dysregulated glymphatic function. These cell-instrinsic and environmental factors conspire to heighten the age-related risk for neuroimmune activation and associated pathology. In this review, we discuss cellular and molecular neuroimmune shifts with "healthy" aging; how these age-related changes affect physiology and behavior; and how recent research has revealed neuroimmune pathways and targets for improving health span.


Assuntos
Encéfalo , Neuroimunomodulação , Encéfalo/metabolismo , Homeostase , Citocinas/metabolismo
16.
J Neuroinflammation ; 8: 110, 2011 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-21878126

RESUMO

In this review, we first provide a brief historical perspective, discussing how peripheral nerve injury (PNI) may have caused World War I. We then consider the initiation, progression, and resolution of the cellular inflammatory response after PNI, before comparing the PNI inflammatory response with that induced by spinal cord injury (SCI).In contrast with central nervous system (CNS) axons, those in the periphery have the remarkable ability to regenerate after injury. Nevertheless, peripheral nervous system (PNS) axon regrowth is hampered by nerve gaps created by injury. In addition, the growth-supportive milieu of PNS axons is not sustained over time, precluding long-distance regeneration. Therefore, studying PNI could be instructive for both improving PNS regeneration and recovery after CNS injury. In addition to requiring a robust regenerative response from the injured neuron itself, successful axon regeneration is dependent on the coordinated efforts of non-neuronal cells which release extracellular matrix molecules, cytokines, and growth factors that support axon regrowth. The inflammatory response is initiated by axonal disintegration in the distal nerve stump: this causes blood-nerve barrier permeabilization and activates nearby Schwann cells and resident macrophages via receptors sensitive to tissue damage. Denervated Schwann cells respond to injury by shedding myelin, proliferating, phagocytosing debris, and releasing cytokines that recruit blood-borne monocytes/macrophages. Macrophages take over the bulk of phagocytosis within days of PNI, before exiting the nerve by the circulation once remyelination has occurred. The efficacy of the PNS inflammatory response (although transient) stands in stark contrast with that of the CNS, where the response of nearby cells is associated with inhibitory scar formation, quiescence, and degeneration/apoptosis. Rather than efficiently removing debris before resolving the inflammatory response as in other tissues, macrophages infiltrating the CNS exacerbate cell death and damage by releasing toxic pro-inflammatory mediators over an extended period of time. Future research will help determine how to manipulate PNS and CNS inflammatory responses in order to improve tissue repair and functional recovery.


Assuntos
Inflamação/imunologia , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/imunologia , Traumatismos dos Nervos Periféricos/patologia , Degeneração Walleriana/imunologia , Animais , Axônios/metabolismo , Axônios/patologia , Humanos , Inflamação/patologia , Células de Schwann/citologia , Células de Schwann/metabolismo , Degeneração Walleriana/patologia
17.
J Neurotrauma ; 36(12): 1923-1934, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30501584

RESUMO

Spinal cord injury (SCI) dysregulates metabolic homeostasis. Metabolic homeostasis is optimized across the day by the circadian system. Despite the prevalence of metabolic pathologies after SCI, post-SCI circadian regulation of metabolism remains understudied. Here, we hypothesized that SCI in rats would disrupt circadian regulation of key metabolic organs, leading to metabolic dysregulation. Female and male Sprague-Dawley rats received moderate thoracic (T)-9 contusion SCI (or sham surgery). First, SCI disrupted diurnal rhythms in two metabolic behaviors: fecal production and food intake rhythms were ablated acutely. SCI also expedited whole-gut transit time. In parallel, acute SCI increased plasma glucose. Diurnal glucose storage-release cycles regulated by the liver were disrupted by SCI, which also increased liver glucose metabolism messenger RNAs (mRNAs). Further, SCI disrupted liver clock gene expression and suppressed inflammatory gene rhythms. Together, our novel data suggest that SCI disrupts typical metabolic and circadian function. Improving post-SCI metabolic function could enhance recovery of homeostasis.


Assuntos
Ritmo Circadiano/fisiologia , Defecação/fisiologia , Fígado/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Animais , Feminino , Glucose/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Vértebras Torácicas/lesões
18.
Neurotherapeutics ; 15(3): 554-577, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29728852

RESUMO

Glial cell types were classified less than 100 years ago by del Rio-Hortega. For instance, he correctly surmised that microglia in pathologic central nervous system (CNS) were "voracious monsters" that helped clean the tissue. Although these historical predictions were remarkably accurate, innovative technologies have revealed novel molecular, cellular, and dynamic physiologic aspects of CNS glia. In this review, we integrate recent findings regarding the roles of glia and glial interactions in healthy and injured spinal cord. The three major glial cell types are considered in healthy CNS and after spinal cord injury (SCI). Astrocytes, which in the healthy CNS regulate neurotransmitter and neurovascular dynamics, respond to SCI by becoming reactive and forming a glial scar that limits pathology and plasticity. Microglia, which in the healthy CNS scan for infection/damage, respond to SCI by promoting axon growth and remyelination-but also with hyperactivation and cytotoxic effects. Oligodendrocytes and their precursors, which in healthy tissue speed axon conduction and support axonal function, respond to SCI by differentiating and producing myelin, but are susceptible to death. Thus, post-SCI responses of each glial cell can simultaneously stimulate and stifle repair. Interestingly, potential therapies could also target interactions between these cells. Astrocyte-microglia cross-talk creates a feed-forward loop, so shifting the response of either cell could amplify repair. Astrocytes, microglia, and oligodendrocytes/precursors also influence post-SCI cell survival, differentiation, and remyelination, as well as axon sparing. Therefore, optimizing post-SCI responses of glial cells-and interactions between these CNS cells-could benefit neuroprotection, axon plasticity, and functional recovery.


Assuntos
Forma Celular , Neuroglia/patologia , Traumatismos da Medula Espinal , Animais , Humanos , Oligodendroglia/fisiologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
19.
Neuroscientist ; 24(3): 221-245, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28737113

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that broadly affect cellular and physiological function in all multicellular organisms. Here, the role of miRNAs in neuroinflammation is considered. miRNAs are 21- to 23-oligonucleotide RNAs that regulate translation of specific RNAs by binding to complementary regulatory RNA sequences, thereby causing mRNA degradation or sequestration. More than 5000 miRNAs likely exist in humans, and each miRNA binds an average of 200 RNAs. Specific immunomodulatory miRNAs can regulate a set of RNAs in a coordinated manner, suggesting that effective miRNA-based therapeutic manipulations for neuroinflammatory conditions may be revealed. For instance, miRNAs that preferentially inhibit translation of many cellular anti-inflammatory proteins could drive a pro-inflammatory response. Key pro-inflammatory ( miR-155, miR-27b, miR-326), anti-inflammatory ( miR-124, miR-146a, miR-21, miR-223), and mixed immunomodulatory ( let-7 family) miRNAs regulate neuroinflammation in various pathologies, including spinal cord injury, multiple sclerosis, ischemic stroke, and Alzheimer's disease. miRNAs represent a newly revealed layer of physiological complexity, the therapeutic benefits of which remain to be fully explored and exploited. In this review, we discuss the role of miRNAs in neuroinflammatory regulation and discuss how controlling miRNAs could alter cellular machinery to improve neuroinflammatory dynamics.


Assuntos
Doenças do Sistema Nervoso Central/imunologia , Inflamação/metabolismo , MicroRNAs/metabolismo , Animais , Humanos , Neuroimunomodulação/fisiologia
20.
Neurology ; 90(8): e724-e726, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29459454

RESUMO

Early multiple sclerosis is characterized by immune-associated demyelination of CNS axons. In a recent Neurology® article, Maranzano et al. evaluated MRI scans of patients with early multiple sclerosis to study the evolution of leukocortical lesions. Their novel data suggest that acute inflammation after blood-brain barrier leakage may contribute to gray matter cortical lesions in early multiple sclerosis.


Assuntos
Doenças Desmielinizantes , Esclerose Múltipla , Substância Cinzenta , Humanos , Inflamação , Imageamento por Ressonância Magnética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA