microglial response
Recently Published Documents


TOTAL DOCUMENTS

163
(FIVE YEARS 50)

H-INDEX

38
(FIVE YEARS 5)

2021 ◽  
Author(s):  
Rida Rehman ◽  
Michael Miller ◽  
Sruthi Sankari Krishnamurthy ◽  
Jacob Kjell ◽  
Lobna Elsayed ◽  
...  

AbstractThe complexity of the signaling events, cellular responses unfolding in neuronal, glial and immune cells upon Traumatic brain injury (TBI) constitutes an obstacle in elucidating pathophysiological links and targets for intervention. We used array phosphoproteomics in a murine mild blunt TBI to reconstruct the temporal dynamics of tyrosine-kinase signaling in TBI and then to scrutinize the large-scale effects of the perturbation of cMet/HGFR, VEGFR1 and Btk signaling by small molecules. cMet/HGFR emerged as a selective modifier of the early microglial response, and cMet/HGFR blockade prevented the induction of microglial inflammatory mediators, of reactive microglia morphology and of TBI-associated responses in neurons, vessels and brain extracellular matrix. Acute or prolonged cMet/HGFR inhibition ameliorated neuronal survival and motor recovery. Early elevation of HGF itself in the CSF of TBI patients suggest that this mechanism has translational value in human subjects. Our findings identify cMet/HGFR as a modulator of early neuroinflammation in TBI with translational potential and indicate several RTK families as possible additional targets for TBI treatment.SummaryControlling neuroinflammation in neurotrauma is an important but unachieved goal. This study exploits a moderate TBI model and array-based proteomics to identify cMet as a new inducer of reactive microglia. A small-molecule inhibitor of cMet contains microglial reactivity, reduces neuronal and vascular alterations, limits behavioural disturbances and accelerates recovery.HighlightsMet is activated in microglia upon TBI and drives microglial reactivity.A Met inhibitor reduces motor dysfunction upon TBI and promotes recovery.Blockade of MET prevents the appearance of a reactive microglia.The cMET inhibitor reduces the sub-acute neuronal loss after TBI.


2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. 994-995
Author(s):  
Rodney Ritzel ◽  
Yun Li ◽  
Jordan Carter ◽  
Niaz Khan ◽  
Junyun He ◽  
...  

Abstract Older patients with traumatic brain injury (TBI) have higher mortality and poorer long-term outlook compared to younger individuals. This may contribute to the assumption that aggressive management of geriatric TBI is futile. The present study examined the long-term recovery potential and underlying mechanisms associated with advanced age in male C57BL/6 mice using a controlled cortical impact model of TBI. Older (18 mos) mice had higher mortality compared to younger (10 wks) mice at 12 weeks post-injury. While aging alone had a profound impact on behavioral ability, the recovery slope in some, but not all, neurobehavioral tests was relatively similar between young and old injured mice. NanoString analysis identified several age- and injury-specific genes that were differentially expressed, including those involved with the complement, phagocytosis, and autophagy pathways. Flow cytometry demonstrated dysregulation of autophagic function in microglia with normal aging which was exacerbated after TBI. Given the critical role for autophagy in promoting the cellular degradation of cytoplasmic materials, we reasoned that treatment with the autophagic inducer, trehalose, may be a viable therapeutic strategy. Trehalose was administered in the drinking water (3%) starting at d1 post-injury up to 8 weeks. Older TBI mice treated with trehalose exhibited either delayed deficits or enhanced recovery in cognitive and motor tasks. Trehalose modified expression of autophagy markers and reprogrammed the microglial response to TBI. Our data indicate that microglia undergo chronic changes in autophagic regulation that are associated with poor outcome. Boosting autophagy may be a promising therapeutic strategy for older TBI patients.


2021 ◽  
Vol 17 (S1) ◽  
Author(s):  
Gloria Biechele ◽  
Boris‐Stephan Rauchmann ◽  
Daniel Janowitz ◽  
Katharina Buerger ◽  
Nicolai Franzmeier ◽  
...  

2021 ◽  
Author(s):  
Corinne Benakis ◽  
Alba Simats ◽  
Sophie Tritschler ◽  
Steffanie Heindl ◽  
Simon Besson-Girard ◽  
...  

Neuroinflammation after stroke is characterized by the activation of resident microglia and the invasion of circulating leukocytes into the brain. Although lymphocytes infiltrate the brain in small number, they have been consistently demonstrated to be the most potent leukocyte subpopulation contributing to secondary inflammatory brain injury. However, the exact mechanism how this minimal number of lymphocytes can profoundly affect stroke outcome is still largely elusive. Here, using a mouse model for ischemic stroke, we demonstrated that early activation of microglia in response to stroke is differentially regulated by distinct T cell subpopulations. Acute treatment with engineered T cells overexpressing IL-10 administered into the cisterna magna after stroke induces a switch of microglial gene expression to a profile associated with pro-regenerative functions. These findings substantiate the role of T cells in stroke with large impact on the cerebral inflammatory milieu by polarizing the microglial phenotype. Targeting T cell-microglia interactions can have direct translational relevance for further development of immune-targeted therapies for stroke and other neuroinflammatory conditions.


Cells ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 2381
Author(s):  
Alexandra Boyd ◽  
Sarah Byrne ◽  
Ryan J. Middleton ◽  
Richard B. Banati ◽  
Guo-Jun Liu

Microglia, the innate immune cells of the central nervous system, play a pivotal role in the modulation of neuroinflammation. Neuroinflammation has been implicated in many diseases of the CNS, including Alzheimer’s disease and Parkinson’s disease. It is well documented that microglial activation, initiated by a variety of stressors, can trigger a potentially destructive neuroinflammatory response via the release of pro-inflammatory molecules, and reactive oxygen and nitrogen species. However, the potential anti-inflammatory and neuroprotective effects that microglia are also thought to exhibit have been under-investigated. The application of ionising radiation at different doses and dose schedules may reveal novel methods for the control of microglial response to stressors, potentially highlighting avenues for treatment of neuroinflammation associated CNS disorders, such as Alzheimer’s disease and Parkinson’s disease. There remains a need to characterise the response of microglia to radiation, particularly low dose ionising radiation.


Antioxidants ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 1340
Author(s):  
Andrea Lopez-Lopez ◽  
Begoña Villar-Cheda ◽  
Aloia Quijano ◽  
Pablo Garrido-Gil ◽  
María Garcia-Garrote ◽  
...  

Dysregulation of the tissue renin–angiotensin system (RAS) is involved in tissue oxidative and inflammatory responses. Among RAS components, renin, its precursor (pro)renin and its specific receptor (PRR) have been less investigated, particularly in the brain. We previously showed the presence of PRR in neurons and glial cells in the nigrostriatal system of rodents and primates, including humans. Now, we used rat and mouse models and cultures of BV2 and primary microglial cells to study the role of PRR in microglial pro-inflammatory responses. PRR was upregulated in the nigral region, particularly in microglia during the neuroinflammatory response. In the presence of the angiotensin type-1 receptor blocker losartan, to exclude angiotensin-related effects, treatment of microglial cells with (pro)renin induces the expression of microglial pro-inflammatory markers, which is mediated by upregulation of NADPH-oxidase and Rho-kinase activities, downregulation of autophagy and upregulation of inflammasome activity. Conditioned medium from (pro)renin-treated microglia increased dopaminergic cell death relative to medium from non-treated microglia. However, these effects were blocked by pre-treatment of microglia with the Rho-kinase inhibitor fasudil. Activation of microglial PRR enhances the microglial pro-inflammatory response and deleterious effects of microglia on dopaminergic cells, and microglial NADPH-oxidase, Rho-Kinase and autophagy are involved in this process.


2021 ◽  
Vol 2 ◽  
Author(s):  
Rita Silva ◽  
Marzia Malcangio

Injuries to the nervous system can result in a debilitating neuropathic pain state that is often resistant to treatment with available analgesics, which are commonly associated with several side-effects. Growing pre-clinical and clinical evidence over the last two decades indicates that immune cell-mediated mechanisms both in the periphery and in the Central Nervous System (CNS) play significant roles in the establishment and maintenance of neuropathic pain. Specifically, following peripheral nerve injury, microglia, which are CNS resident immune cells, respond to the activity of the first pain synapse in the dorsal horn of spinal cord and also to neuronal activity in higher centres in the brain. This microglial response leads to the production and release of several proinflammatory mediators which contribute to neuronal sensitisation under neuropathic pain states. In this review, we collect evidence demonstrating the critical role played by the Fractalkine/CX3CR1 signalling pathway in neuron-to-microglia communication in neuropathic pain states and explore how strategies that include components of this pathway offer opportunities for innovative targets for neuropathic pain.


2021 ◽  
Vol 218 (9) ◽  
Author(s):  
Yun Chen ◽  
Marco Colonna

Alzheimer’s disease (AD) is characterized by extracellular aggregates of amyloid β peptides, intraneuronal tau aggregates, and neuronal death. This pathology triggers activation of microglia. Because variants of genes expressed in microglia correlate with AD risk, microglial response to pathology plausibly impacts disease course. In mouse AD models, single-cell RNA sequencing (scRNA-seq) analyses delineated this response as progressive conversion of homeostatic microglia into disease-associated microglia (DAM); additional reactive microglial populations have been reported in other models of neurodegeneration and neuroinflammation. We review all of these microglial signatures, highlighting four fundamental patterns: DAM, IFN–microglia, MHC-II microglia, and proliferating microglia. We propose that all reported microglia populations are either just one or a combination, depending on the clustering strategy applied and the disease model. We further review single-nucleus RNA sequencing (snRNA-seq) data from human AD specimens and discuss reasons for parallels and discrepancies between human and mouse transcriptional profiles. Finally, we outline future directions for delineating the microglial impact in AD pathogenesis.


2021 ◽  
Author(s):  
Akshita Jade Kumar ◽  
Supinder Singh Bedi ◽  
Naama Toledano-Furman ◽  
Louis Carrillo ◽  
Fanni Cardenas ◽  
...  

Abstract Background: Traumatic brain injury (TBI) is a systemic injury that disrupts a complex arrangement of interacting cells in the brain and in the gastrointestinal tract (GI). Disruption in the brain results in neuroinflammation, in which microglia are a central component along with cytokines and other soluble factors [pro and anti-inflammatory microglia (M1:M2)]. Disruption in the GI due to TBI results in a systemic inflammation which is dependent upon the gut microbiome (GM). Gut microbiome can influence microglia in the brain via the gut-brain axis. In order to determine if the microbiome-microglia connections via the gut-brain axis can be modulated, we used probiotics and antibiotics in a rodent TBI model to evaluate the microbiome-microglial connections in acute and chronic experiments.Methods: The temporal effects of treatment (probiotics or antibiotics) were used to evaluate the gut-associated lymphoid tissue (GALT) influence on the microglial response at 72 hours or 21 days after a cortical contusion injury (CCI), a rodent model of TBI. Injured animals received daily probiotics, antibiotics, or no treatment. Sham-injured animals (controls) did not receive any treatment.Results: Twenty-one days of probiotic treatment attenuated the pro-inflammatory response of microglia (M1:M2) after CCI. The post-injury inflammatory response was heightened in the GALT with antibiotic-induced dysbiosis which resulted in amplification of the pro-inflammatory microglial response. Conclusions: Probiotic treatment after TBI is a potential therapeutic in attenuating microglial activation through anti-inflammatory signaling.


Sign in / Sign up

Export Citation Format

Share Document