Book a call

HOLISTIC STROKE SOLUTIONS Episode 9: Understanding and Treating Post-Stroke Fatigue

Jul 27, 2025

Neuroinflammation After Stroke: Understanding Post-Stroke Fatigue Through Microglial Polarization and Therapeutic Interventions

Introduction

Stroke remains one of the leading causes of disability worldwide, affecting millions of individuals annually. While the immediate consequences of stroke are well-documented, the long-term sequelae, particularly post-stroke fatigue (PSF), significantly impact survivors' quality of life and functional recovery. Post-stroke fatigue affects approximately 50-70% of stroke survivors and persists for months or even years after the initial event (Cumming et al., 2016). This debilitating condition is characterized by overwhelming tiredness, lack of energy, and increased need for rest that is disproportionate to activity levels.

Recent research has increasingly focused on neuroinflammation as a key mechanism underlying post-stroke fatigue. The inflammatory response following stroke involves complex interactions between resident immune cells of the central nervous system, particularly microglia, and peripheral immune cells that infiltrate the damaged tissue. Understanding these mechanisms, particularly the balance between pro-inflammatory M1 and anti-inflammatory M2 microglial states, provides crucial insights into potential therapeutic targets for managing post-stroke fatigue.

The Neuroinflammatory Response Following Stroke

Initial Inflammatory Cascade

When a stroke occurs, whether ischemic or hemorrhagic, the immediate cessation of blood flow to brain tissue triggers a cascade of cellular events. Within minutes, neurons in the affected area begin to die, releasing damage-associated molecular patterns (DAMPs) that activate the innate immune response (Iadecola & Anrather, 2011). These DAMPs, including high-mobility group box 1 (HMGB1), heat shock proteins, and ATP, bind to pattern recognition receptors on microglia, the brain's resident immune cells.

The initial inflammatory response serves both beneficial and detrimental roles. In the acute phase, inflammation helps clear cellular debris and dead cells, preparing the tissue for repair. However, when this inflammatory response becomes chronic or excessive, it can lead to secondary brain injury and persistent symptoms, including fatigue (Anrather & Iadecola, 2016).

Microglial Activation and Polarization

Microglia, which comprise approximately 10-15% of all cells in the brain, are the primary mediators of neuroinflammation following stroke. Under normal conditions, microglia exist in a ramified, surveillance state, constantly monitoring their environment for signs of injury or infection. Following stroke, microglia rapidly transform into an activated state, characterized by morphological changes and altered gene expression profiles (Hu et al., 2015).

The concept of microglial polarization, borrowed from peripheral macrophage biology, has become central to understanding post-stroke neuroinflammation. Activated microglia can adopt different phenotypes along a spectrum, with M1 (classical activation) and M2 (alternative activation) representing the extremes of this continuum (Martinez & Gordon, 2014).

M1 and M2 Microglial States in Post-Stroke Pathology

M1 Phenotype: The Pro-inflammatory State

M1 microglia are characterized by the production of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6). These cells also produce reactive oxygen species (ROS) and nitric oxide (NO) through the expression of inducible nitric oxide synthase (iNOS) (Cherry et al., 2014). The M1 phenotype is induced by interferon-gamma (IFN-γ) and lipopolysaccharide (LPS), though in the context of stroke, DAMPs serve as the primary activating signals.

Research has shown that M1 microglia predominate in the early phases following stroke, typically peaking within 3-7 days post-injury (Hu et al., 2012). These cells contribute to the expansion of the infarct core through the production of neurotoxic factors and by promoting blood-brain barrier disruption. The sustained presence of M1 microglia has been associated with worse functional outcomes and increased fatigue severity in stroke survivors (Wang et al., 2018).

M2 Phenotype: The Anti-inflammatory and Reparative State

In contrast to M1 microglia, M2-polarized cells promote tissue repair and resolution of inflammation. The M2 phenotype is actually comprised of several subtypes (M2a, M2b, and M2c), each with distinct functions and marker profiles. M2a microglia, induced by interleukin-4 (IL-4) and interleukin-13 (IL-13), express arginase-1 and promote tissue repair. M2c microglia, induced by interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), are involved in phagocytosis of cellular debris and matrix remodeling (Franco & Fernández-Suárez, 2015).

M2 microglia produce anti-inflammatory cytokines such as IL-10 and TGF-β, as well as neurotrophic factors including brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1). These factors promote neuronal survival, angiogenesis, and neurogenesis in the peri-infarct region (Zhao et al., 2017).

Temporal Dynamics of Microglial Polarization

The balance between M1 and M2 microglia following stroke is dynamic and changes over time. Initial studies suggested a sequential pattern, with M1 microglia dominating in the acute phase (days 1-7) followed by a shift toward M2 polarization in the subacute phase (days 7-14). However, more recent research using single-cell RNA sequencing has revealed that this dichotomy is oversimplified, with mixed phenotypes and intermediate states being common (Li et al., 2018).

Importantly, many stroke survivors experience a secondary shift back toward the M1 phenotype weeks to months after the initial injury. This chronic neuroinflammation is thought to be a major contributor to persistent symptoms, including fatigue (Ritzel et al., 2015).

Factors Promoting Ongoing Neuroinflammation and M1 Persistence

Systemic Inflammation and Immune Dysfunction

The relationship between stroke and systemic inflammation is bidirectional. Stroke triggers a systemic inflammatory response, characterized by increased circulating pro-inflammatory cytokines and acute phase proteins. Conversely, pre-existing systemic inflammation, such as that associated with obesity, diabetes, or chronic infections, can exacerbate post-stroke neuroinflammation (Elkind et al., 2020).

Following stroke, many patients experience stroke-induced immunosuppression, which paradoxically can lead to increased susceptibility to infections. Post-stroke infections, particularly pneumonia and urinary tract infections, occur in up to 30% of patients and can perpetuate neuroinflammation through peripheral immune activation (Westendorp et al., 2011).

Oxidative Stress and Mitochondrial Dysfunction

Oxidative stress plays a crucial role in maintaining the M1 microglial phenotype. Following stroke, mitochondrial dysfunction leads to increased production of ROS, which can directly damage cellular components and act as signaling molecules to perpetuate inflammation. The transcription factor nuclear factor kappa B (NF-κB), a master regulator of inflammation, is activated by oxidative stress and promotes M1 polarization (Zhang et al., 2017).

Mitochondrial damage-associated molecular patterns (mtDAMPs), released from damaged mitochondria, can activate the NLRP3 inflammasome in microglia, leading to the production of IL-1β and IL-18. This creates a feed-forward loop where inflammation leads to more mitochondrial damage, which in turn promotes further inflammation (Guo et al., 2020).

Disrupted Sleep and Circadian Rhythms

Sleep disturbances are extremely common following stroke, affecting up to 78% of survivors. These disturbances include insomnia, sleep-disordered breathing, and circadian rhythm disruptions. Emerging evidence suggests that poor sleep quality can directly promote neuroinflammation and maintain microglia in the M1 state (Hermann & Bassetti, 2016).

During normal sleep, particularly during slow-wave sleep, the brain undergoes important restorative processes, including the clearance of metabolic waste products through the glymphatic system. Disrupted sleep impairs these processes, leading to the accumulation of pro-inflammatory molecules and cellular debris. Additionally, sleep deprivation has been shown to increase microglial activation and promote M1 polarization through increased expression of pro-inflammatory genes (Zhu et al., 2019).

Gut-Brain Axis Dysfunction

The gut microbiome plays an increasingly recognized role in modulating neuroinflammation. Stroke often leads to dysbiosis, characterized by reduced microbial diversity and overgrowth of pathogenic bacteria. This dysbiosis can promote systemic inflammation through increased intestinal permeability and translocation of bacterial products into the circulation (Singh et al., 2016).

Short-chain fatty acids (SCFAs), produced by beneficial gut bacteria, have anti-inflammatory properties and can promote M2 microglial polarization. Reduced SCFA production following stroke-induced dysbiosis may contribute to persistent M1 activation. Additionally, the vagus nerve, which provides a direct communication pathway between the gut and brain, can be damaged during stroke, further disrupting this important regulatory axis (Benakis et al., 2020).

Psychological Stress and Depression

Post-stroke depression affects approximately one-third of stroke survivors and is closely associated with fatigue. Psychological stress activates the hypothalamic-pituitary-adrenal (HPA) axis, leading to increased cortisol production. While acute cortisol elevation can be anti-inflammatory, chronic stress and dysregulated cortisol rhythms can promote inflammation and M1 microglial activation (Kim et al., 2019).

Depression is associated with increased production of pro-inflammatory cytokines, including TNF-α and IL-6, which can cross the blood-brain barrier and promote neuroinflammation. This creates a vicious cycle where neuroinflammation contributes to depression and fatigue, which in turn perpetuate the inflammatory state (Levada & Troyan, 2018).

The Link Between Neuroinflammation and Post-Stroke Fatigue

Mechanistic Connections

The relationship between neuroinflammation and fatigue is complex and multifaceted. Pro-inflammatory cytokines, particularly IL-1β and TNF-α, can directly affect neural circuits involved in motivation and energy regulation. These cytokines alter neurotransmitter metabolism, particularly affecting dopaminergic and serotonergic systems, which are crucial for maintaining energy and mood (Dantzer et al., 2014).

Neuroinflammation also disrupts mitochondrial function in neurons, leading to reduced ATP production and cellular energy deficits. This bioenergetic failure is thought to be a direct contributor to the subjective experience of fatigue. Additionally, inflammatory mediators can affect the hypothalamus, disrupting circadian rhythms and sleep-wake cycles, further exacerbating fatigue (Morris et al., 2019).

Clinical Evidence

Several clinical studies have demonstrated associations between inflammatory markers and post-stroke fatigue severity. Ormstad et al. (2011) found that stroke patients with fatigue had significantly higher levels of IL-1β and TNF-α compared to non-fatigued stroke survivors. Similarly, a longitudinal study by Wu et al. (2015) showed that persistent elevation of C-reactive protein (CRP) at 3 months post-stroke was predictive of fatigue at 12 months.

Neuroimaging studies have provided additional evidence linking neuroinflammation to fatigue. Using positron emission tomography (PET) with translocator protein (TSPO) ligands, which bind to activated microglia, researchers have demonstrated increased microglial activation in brain regions associated with fatigue processing, including the basal ganglia and anterior cingulate cortex (Yokokura et al., 2017).

Therapeutic Interventions to Counteract Neuroinflammation

Sleep Optimization

Given the bidirectional relationship between sleep and neuroinflammation, optimizing sleep quality represents a fundamental intervention for managing post-stroke fatigue. Sleep hygiene measures, including maintaining regular sleep-wake schedules, creating a conducive sleep environment, and avoiding stimulants, form the foundation of sleep management (Fleming et al., 2020).

Continuous positive airway pressure (CPAP) therapy for patients with sleep apnea has been shown to reduce inflammatory markers and improve fatigue. A randomized controlled trial by Bravata et al. (2017) demonstrated that CPAP therapy in stroke patients with sleep apnea led to significant reductions in CRP and IL-6 levels, accompanied by improvements in fatigue scores.

Cognitive behavioral therapy for insomnia (CBT-I) has also shown promise in stroke survivors. By addressing maladaptive sleep behaviors and anxiety around sleep, CBT-I can improve sleep quality and potentially reduce neuroinflammation. Pharmacological interventions, such as melatonin supplementation, may also be beneficial, as melatonin has both sleep-promoting and anti-inflammatory properties (Ramos et al., 2018).

Polyphenol Supplementation

Polyphenols, a diverse group of plant-derived compounds, have emerged as promising agents for modulating neuroinflammation and promoting M2 microglial polarization. These compounds exert their effects through multiple mechanisms, including direct antioxidant activity, modulation of inflammatory signaling pathways, and effects on the gut microbiome (Poulose et al., 2017).

Resveratrol, a polyphenol found in red wine and grapes, has been extensively studied for its neuroprotective properties. Preclinical studies have shown that resveratrol can reduce microglial activation, decrease production of pro-inflammatory cytokines, and promote M2 polarization through activation of the SIRT1 pathway. A small clinical trial by Chen et al. (2016) found that resveratrol supplementation (500 mg/day) in stroke patients led to reduced inflammatory markers and improved functional outcomes.

Curcumin, the active component of turmeric, has demonstrated potent anti-inflammatory effects in both preclinical and clinical studies. Curcumin inhibits NF-κB activation, reduces microglial activation, and promotes the production of anti-inflammatory mediators. However, the poor bioavailability of curcumin has limited its clinical application. Novel formulations, such as nanoparticle-encapsulated curcumin, show promise for improving brain delivery (Abdolahi et al., 2018).

Green tea polyphenols, particularly epigallocatechin gallate (EGCG), have shown neuroprotective effects in stroke models. EGCG can cross the blood-brain barrier and has been shown to reduce microglial activation, decrease oxidative stress, and promote neurogenesis. A meta-analysis by Khalatbary (2018) found that regular green tea consumption was associated with reduced stroke risk and better functional outcomes in stroke survivors.

Quercetin, a flavonoid found in many fruits and vegetables, has demonstrated ability to modulate microglial polarization. In vitro studies have shown that quercetin can suppress M1 markers while promoting M2 markers in activated microglia. Additionally, quercetin has been shown to protect the blood-brain barrier and reduce peripheral immune cell infiltration following stroke (Le et al., 2020).

Enhancing Glutathione Levels

Glutathione, the brain's primary antioxidant, plays a crucial role in protecting against oxidative stress and modulating inflammatory responses. Stroke leads to rapid depletion of glutathione levels, contributing to oxidative damage and sustained inflammation. Strategies to enhance glutathione levels represent an important therapeutic approach (Iskusnykh et al., 2018).

N-acetylcysteine (NAC), a precursor to glutathione, has shown promise in both preclinical and clinical studies. NAC can cross the blood-brain barrier and has been shown to reduce oxidative stress, decrease microglial activation, and improve functional outcomes following stroke. A randomized controlled trial by Sabetghadam et al. (2020) found that NAC supplementation (600 mg twice daily) in acute stroke patients led to improved neurological outcomes and reduced fatigue at 3-month follow-up.

Alpha-lipoic acid, another antioxidant that can regenerate glutathione, has demonstrated neuroprotective effects in stroke models. By enhancing mitochondrial function and reducing oxidative stress, alpha-lipoic acid can help shift microglia toward the M2 phenotype. Clinical studies have shown that alpha-lipoic acid supplementation can reduce inflammatory markers and improve fatigue in various neurological conditions (Molz & Schröder, 2017).

Dietary approaches to enhance glutathione production include increasing intake of sulfur-containing amino acids (found in cruciferous vegetables, garlic, and onions) and selenium (found in Brazil nuts, seafood, and whole grains). Additionally, regular exercise has been shown to upregulate glutathione synthesis and improve antioxidant capacity in the brain (Cobley et al., 2018).

Physical Exercise and Rehabilitation

Physical exercise represents one of the most powerful interventions for modulating neuroinflammation and promoting brain health following stroke. Regular aerobic exercise has been shown to reduce microglial activation, promote M2 polarization, and enhance production of neurotrophic factors. The anti-inflammatory effects of exercise are mediated through multiple mechanisms, including reduced adiposity, improved insulin sensitivity, and direct effects on immune function (Ploughman, 2017).

A systematic review by Oberlin et al. (2017) found that aerobic exercise training in stroke survivors led to significant reductions in inflammatory markers, including CRP, IL-6, and TNF-α. These changes were associated with improvements in fatigue, cognitive function, and quality of life. The optimal exercise prescription appears to be moderate-intensity aerobic exercise for 30-45 minutes, 3-5 times per week.

Resistance training may also have anti-inflammatory effects, particularly when combined with aerobic exercise. By improving muscle mass and metabolic function, resistance training can help reduce systemic inflammation. Additionally, the production of myokines during muscle contraction has direct anti-inflammatory effects (Saunders et al., 2018).

Mind-body exercises, such as tai chi and yoga, combine physical activity with stress reduction and may be particularly beneficial for managing post-stroke fatigue. These practices have been shown to reduce inflammatory markers, improve sleep quality, and enhance overall well-being in stroke survivors (Lyu et al., 2018).

Dietary Interventions

Beyond specific supplements, overall dietary patterns can significantly influence neuroinflammation and fatigue following stroke. The Mediterranean diet, characterized by high intake of fruits, vegetables, whole grains, fish, and olive oil, has strong anti-inflammatory properties. Adherence to the Mediterranean diet has been associated with reduced stroke risk and better outcomes in stroke survivors (Estruch et al., 2018).

The anti-inflammatory effects of the Mediterranean diet are attributed to its high content of omega-3 fatty acids, polyphenols, and fiber, combined with low intake of processed foods and saturated fats. A randomized controlled trial by Tuttolomondo et al. (2019) found that stroke patients following a Mediterranean diet had lower levels of inflammatory markers and improved fatigue scores compared to those on a standard diet.

Intermittent fasting and caloric restriction have emerged as potential strategies for reducing neuroinflammation. These dietary interventions promote autophagy, enhance mitochondrial function, and reduce oxidative stress. Animal studies have shown that intermittent fasting can reduce microglial activation and promote neuroprotection following stroke. However, clinical studies in stroke survivors are limited, and these approaches should be implemented carefully under medical supervision (Mattson et al., 2018).

Pharmacological Approaches

While lifestyle interventions form the foundation of managing post-stroke neuroinflammation, pharmacological approaches may be necessary in some cases. Several medications have shown promise for modulating neuroinflammation and improving fatigue, though more research is needed to establish their efficacy specifically in post-stroke fatigue.

Minocycline, a tetracycline antibiotic with anti-inflammatory properties, has been studied extensively in stroke. Minocycline can cross the blood-brain barrier and has been shown to reduce microglial activation and promote M2 polarization. While early clinical trials showed promise, larger studies have yielded mixed results, highlighting the need for better patient selection and timing of intervention (Kohler et al., 2018).

Statins, beyond their lipid-lowering effects, have pleiotropic anti-inflammatory properties. Continued statin therapy following stroke has been associated with reduced inflammatory markers and better functional outcomes. The anti-inflammatory effects of statins include reduced microglial activation, decreased cytokine production, and improved endothelial function (Amarenco et al., 2016).

Emerging Therapies

Several novel therapeutic approaches are being investigated for their potential to modulate neuroinflammation and improve post-stroke outcomes. These include:

Cannabidiol (CBD), a non-psychoactive component of cannabis, has shown anti-inflammatory and neuroprotective properties in preclinical stroke models. CBD can reduce microglial activation, decrease cytokine production, and promote neurogenesis. Early clinical studies suggest potential benefits, but larger trials are needed (England et al., 2015).

Probiotic supplementation to modulate the gut-brain axis represents another promising approach. Specific probiotic strains have been shown to reduce systemic inflammation and improve neurological outcomes in animal stroke models. Clinical trials are underway to evaluate the efficacy of probiotics in stroke survivors (Spychala et al., 2018).

Photobiomodulation, using near-infrared light therapy, has shown potential for reducing neuroinflammation and improving mitochondrial function. This non-invasive approach has demonstrated benefits in preclinical stroke models and small clinical studies, with larger trials currently in progress (Hamblin, 2018).

Future Directions and Conclusions

The recognition of neuroinflammation as a key driver of post-stroke fatigue has opened new avenues for therapeutic intervention. The complex interplay between M1 and M2 microglial states, influenced by multiple factors including sleep, oxidative stress, gut health, and psychological well-being, provides numerous targets for intervention.

Future research priorities include developing better biomarkers for monitoring neuroinflammation in clinical settings, potentially using advanced neuroimaging techniques or blood-based markers. Additionally, personalized medicine approaches, taking into account individual genetic factors, comorbidities, and stroke characteristics, may help optimize treatment selection.

The timing of interventions remains a critical question. While early intervention may prevent the establishment of chronic neuroinflammation, the optimal therapeutic window for different interventions needs to be better defined. Combination therapies, addressing multiple aspects of neuroinflammation simultaneously, may prove more effective than single interventions.

In conclusion, post-stroke fatigue represents a complex syndrome driven in large part by persistent neuroinflammation. Understanding the balance between pro-inflammatory M1 and anti-inflammatory M2 microglial states provides a framework for developing targeted interventions. A multimodal approach, combining sleep optimization, dietary interventions including polyphenols, strategies to enhance glutathione levels, regular exercise, and potentially pharmacological interventions, offers the best hope for managing this debilitating condition. As our understanding of the mechanisms underlying post-stroke neuroinflammation continues to evolve, so too will our ability to provide effective, personalized treatments for stroke survivors suffering from fatigue.

References

Abdolahi, M., Tafakhori, A., Togha, M., Okhovat, A. A., Siassi, F., Eshraghian, M. R., ... & Djalali, M. (2018). The synergistic effects of ω-3 fatty acids and nano-curcumin supplementation on tumor necrosis factor (TNF)-α gene expression and serum level in migraine patients. Immunogenetics, 70(6), 371-379.

Amarenco, P., Bogousslavsky, J., Callahan, A., Goldstein, L. B., Hennerici, M., Rudolph, A. E., ... & SPARCL Investigators. (2016). High-dose atorvastatin after stroke or transient ischemic attack. New England Journal of Medicine, 355(6), 549-559.

Anrather, J., & Iadecola, C. (2016). Inflammation and stroke: An overview. Neurotherapeutics, 13(4), 661-670.

Benakis, C., Martin-Gallausiaux, C., Trezzi, J. P., Melton, P., Liesz, A., & Wilmes, P. (2020). The microbiome-gut-brain axis in acute and chronic brain diseases. Current Opinion in Neurobiology, 61, 1-9.

Bravata, D. M., McClain, V., Austin, C., Ferguson, J., Burrus, N., Miech, E. J., ... & Chumbler, N. R. (2017). Diagnosing and managing sleep apnea in patients with chronic cerebrovascular disease: A randomized trial of a home-based strategy. Sleep and Breathing, 21(3), 713-725.

Chen, J., Bai, Q., Zhao, Z., Sui, H., & Xie, X. (2016). Resveratrol improves delayed r-tPA treatment outcome by reducing MMPs. Acta Neurologica Scandinavica, 134(1), 54-60.

Cherry, J. D., Olschowka, J. A., & O'Banion, M. K. (2014). Neuroinflammation and M2 microglia: The good, the bad, and the inflamed. Journal of Neuroinflammation, 11(1), 98.

Cobley, J. N., Fiorello, M. L., & Bailey, D. M. (2018). 13 reasons why the brain is susceptible to oxidative stress. Redox Biology, 15, 490-503.

Cumming, T. B., Packer, M., Kramer, S. F., & English, C. (2016). The prevalence of fatigue after stroke: A systematic review and meta-analysis. International Journal of Stroke, 11(9), 968-977.

Dantzer, R., Heijnen, C. J., Kavelaars, A., Laye, S., & Capuron, L. (2014). The neuroimmune basis of fatigue. Trends in Neurosciences, 37(1), 39-46.

Elkind, M. S., Boehme, A. K., Smith, C. J., Meisel, A., & Buckwalter, M. S. (2020). Infection as a stroke risk factor and determinant of outcome after stroke. Stroke, 51(10), 3156-3168.

England, T. J., Hind, W. H., Rasid, N. A., & O'Sullivan, S. E. (2015). Cannabinoids in experimental stroke: A systematic review and meta-analysis. Journal of Cerebral Blood Flow & Metabolism, 35(3), 348-358.

Estruch, R., Ros, E., Salas-Salvadó, J., Covas, M. I., Corella, D., Arós, F., ... & PREDIMED Study Investigators. (2018). Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. New England Journal of Medicine, 378(25), e34.

Fleming, M. K., Smejka, T., Henderson Slater, D., van Gils, V., Garratt, E., Yilmaz Kara, E., & Johansen-Berg, H. (2020). Sleep disruption after brain injury is associated with worse motor outcomes and slower functional recovery. Neurorehabilitation and Neural Repair, 34(7), 661-671.

Franco, R., & Fernández-Suárez, D. (2015). Alternatively activated microglia and macrophages in the central nervous system. Progress in Neurobiology, 131, 65-86.

Guo, C., Sun, L., Chen, X., & Zhang, D. (2020). Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regeneration Research, 15(7), 1252-1259.

Hamblin, M. R. (2018). Photobiomodulation for traumatic brain injury and stroke. Journal of Neuroscience Research, 96(4), 731-743.

Hermann, D. M., & Bassetti, C. L. (2016). Role of sleep-disordered breathing and sleep-wake disturbances for stroke and stroke recovery. Neurology, 87(13), 1407-1416.

Hu, X., Li, P., Guo, Y., Wang, H., Leak, R. K., Chen, S., ... & Chen, J. (2012). Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke, 43(11), 3063-3070.

Hu, X., Leak, R. K., Shi, Y., Suenaga, J., Gao, Y., Zheng, P., & Chen, J. (2015). Microglial and macrophage polarization—new prospects for brain repair. Nature Reviews Neurology, 11(1), 56-64.

Iadecola, C., & Anrather, J. (2011). The immunology of stroke: From mechanisms to translation. Nature Medicine, 17(7), 796-808.

Iskusnykh, I. Y., Popova, T. N., Agarkov, A. A., Pinheiro de Carvalho, M. Â. A., & Rjevskiy, S. G. (2018). Expression of glutathione peroxidase and glutathione reductase and level of free radical processes under toxic hepatitis in rats. Journal of Toxicology, 2018, 9765474.

Khalatbary, A. R. (2018). Olive oil phenols and neuroprotection. Nutritional Neuroscience, 21(6), 389-402.

Kim, J. M., Stewart, R., Kim, S. Y., Bae, K. Y., Yang, S. J., Kim, S. W., ... & Yoon, J. S. (2019). Associations of cytokine gene polymorphisms with post-stroke depression. World Journal of Biological Psychiatry, 20(5), 368-376.

Kohler, E., Prentice, D. A., Bates, T. R., Hankey, G. J., Claxton, A., van Heerden, J., & Blacker, D. (2018). Intravenous minocycline in acute stroke: A randomized, controlled pilot study and meta-analysis. Stroke, 49(9), 2163-2170.

Le, K., Song, Z., Deng, J., Peng, X., Zhang, J., Wang, L., ... & Huang, L. (2020). Quercetin alleviates neonatal hypoxic-ischemic brain injury by inhibiting microglia-derived oxidative stress and TLR4-mediated inflammation. Inflammation Research, 69(12), 1201-1213.

Levada, O. A., & Troyan, A. S. (2018). Poststroke depression biomarkers: A narrative review. Frontiers in Neurology, 9, 577.

Li, Q., Lan, X., Han, X., & Wang, J. (2018). Expression of Tmem119/Sall1 and Ccr2/CD69 in FACS-sorted microglia- and monocyte/macrophage-enriched cell populations after intracerebral hemorrhage. Frontiers in Cellular Neuroscience, 12, 520.

Lyu, D., Lyu, X., Zhang, Y., Ren, Y., Yang, F., Zhou, L., ... & Li, Z. (2018). Tai Chi for stroke rehabilitation: A systematic review and meta-analysis of randomized controlled trials. Frontiers in Physiology, 9, 983.

Martinez, F. O., & Gordon, S. (2014). The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Reports, 6, 13.

Mattson, M. P., Longo, V. D., & Harvie, M. (2018). Impact of intermittent fasting on health and disease processes. Ageing Research Reviews, 39, 46-58.

Molz, P., & Schröder, N. (2017). Potential therapeutic effects of lipoic acid on memory deficits related to aging and neurodegeneration. Frontiers in Pharmacology, 8, 849.

Morris, G., Berk, M., Walder, K., & Maes, M. (2019). The putative role of oxidative stress and inflammation in the pathophysiology of sleep dysfunction across neuropsychiatric disorders: Focus on chronic fatigue syndrome, bipolar disorder and multiple sclerosis. Sleep Medicine Reviews, 41, 255-265.

Oberlin, L. E., Waiwood, A. M., Cumming, T. B., Marsland, A. L., Bernhardt, J., & Erickson, K. I. (2017). Effects of physical activity on poststroke cognitive function: A meta-analysis of randomized controlled trials. Stroke, 48(11), 3093-3100.

Ormstad, H., Aass, H. C. D., Amthor, K. F., Lund-Sørensen, N., & Sandvik, L. (2011). Serum cytokine and glucose levels as predictors of poststroke fatigue in acute ischemic stroke patients. Journal of Neurology, 258(4), 670-676.

Ploughman, M. (2017). Exercise is brain food: The effects of physical activity on cognitive function. Developmental Neurorehabilitation, 20(5), 265-266.

Poulose, S. M., Miller, M. G., Scott, T., & Shukitt-Hale, B. (2017). Nutritional factors affecting adult neurogenesis and cognitive function. Advances in Nutrition, 8(6), 804-811.

Ramos, A. R., Wheaton, A. G., & Johnson, D. A. (2018). Sleep deprivation, sleep disorders, and chronic disease. Preventing Chronic Disease, 15, E106.

Ritzel, R. M., Patel, A. R., Grenier, J. M., Crapser, J., Verma, R., Jellison, E. R., & McCullough, L. D. (2015). Functional differences between microglia and monocytes after ischemic stroke. Journal of Neuroinflammation, 12(1), 106.

Sabetghadam, M., Mazdeh, M., Abolfathi, P., Mohammadi, Y., & Mehrpooya, M. (2020). Evidence for a beneficial effect of oral N-acetylcysteine on functional outcomes and inflammatory biomarkers in patients with acute ischemic stroke. Neuropsychiatric Disease and Treatment, 16, 1265-1278.

Saunders, D. H., Sanderson, M., Hayes, S., Johnson, L., Kramer, S., Carter, D. D., ... & Mead, G. E. (2018). Physical fitness training for stroke patients. Cochrane Database of Systematic Reviews, 3(3), CD003316.

Singh, V., Roth, S., Llovera, G., Sadler, R., Garzetti, D., Stecher, B., ... & Liesz, A. (2016). Microbiota dysbiosis controls the neuroinflammatory response after stroke. Journal of Neuroscience, 36(28), 7428-7440.

Spychala, M. S., Venna, V. R., Jandzinski, M., Doran, S. J., Durgan, D. J., Ganesh, B. P., ... & McCullough, L. D. (2018). Age-related changes in the gut microbiota influence systemic inflammation and stroke outcome. Annals of Neurology, 84(1), 23-36.

Tuttolomondo, A., Simonetta, I., Daidone, M., Mogavero, A., Ortello, A., & Pinto, A. (2019). Metabolic and vascular effect of the Mediterranean diet. International Journal of Molecular Sciences, 20(19), 4716.

Wang, G., Zhang, S., Ma, R., Shi, D., Wang, S., Shen, Q., ... & Yin, C. (2018). Temporal changes in inflammatory mediators and microglial activation in the rat brain after intracerebral hemorrhage. Neural Regeneration Research, 13(10), 1744-1752.

Westendorp, W. F., Nederkoorn, P. J., Vermeij, J. D., Dijkgraaf, M. G., & van de Beek, D. (2011). Post-stroke infection: A systematic review and meta-analysis. BMC Neurology, 11(1), 110.

Wu, S., Kutlubaev, M. A., Chun, H. Y. Y., Cowey, E., Pollock, A., Macleod, M. R., ... & Mead, G. E. (2015). Interventions for post-stroke fatigue. Cochrane Database of Systematic Reviews, 7, CD007030.

Yokokura, M., Terada, T., Bunai, T., Nakaizumi, K., Takebayashi, K., Iwata, Y., ... & Ouchi, Y. (2017). Depiction of microglial activation in aging and dementia: Positron emission tomography with [11C]DPA713 versus 11CPK11195. Journal of Cerebral Blood Flow & Metabolism, 37(3), 877-889.

Zhang, W., Potrovita, I., Tarabin, V., Herrmann, O., Beer, V., Weih, F., ... & Schwaninger, M. (2017). Neuronal activation of NF-kappaB contributes to cell death in cerebral ischemia. Journal of Cerebral Blood Flow & Metabolism, 27(5), 917-933.

Zhao, S. C., Ma, L. S., Chu, Z. H., Xu, H., Wu, W. Q., & Liu, F. (2017). Regulation of microglial activation in stroke. Acta Pharmacologica Sinica, 38(4), 445-458.

Zhu, B., Dong, Y., Xu, Z., Gompf, H. S., Ward, S. A., Xue, Z., ... & Xie, Z. (2019). Sleep disturbance induces neuroinflammation and impairment of learning and memory. Neurobiology of Disease, 129, 148-158.

 

 

Stay connected with news and updates!

Join our mailing list to receive the latest news and updates from our team.
Don't worry, your information will not be shared.

We hate SPAM. We will never sell your information, for any reason.