- Review
- Open access
- Published:
Neuroinflammation and energy metabolism: a dual perspective on ischemic stroke
Journal of Translational Medicine volume 23, Article number: 413 (2025)
Abstract
Ischemic stroke is a prevalent form of cerebrovascular accident, with its pathogenesis involving the intricate interplay between neuroinflammation and energy metabolism. Cerebral ischemia disrupts oxygen and energy supply, triggering metabolic dysregulation and activating neuroinflammatory responses, ultimately resulting in cellular damage. This review provides an exhaustive analysis of the complex mechanisms of ischemic stroke, with a particular focus on the interaction between neuroinflammation and energy metabolism. The interruption of oxygen and energy supply due to cerebral ischemia initiates metabolic dysregulation and activates neuroinflammatory responses, including the release of inflammatory cytokines and the activation of immune cells, contributing to cellular damage and further metabolic disturbances. Studies indicate that dysregulation of energy metabolism significantly impairs neural cell function and interacts with neuroinflammation, exacerbating ischemic brain injury. Therapeutic strategies primarily concentrate on modulating energy metabolism and suppressing neuroinflammatory responses, emphasizing the importance of in-depth research into their interaction to provide a theoretical foundation for new treatment strategies for ischemic stroke. Future research should focus on how to balance anti-inflammatory treatment with energy regulation to minimize neural damage and promote recovery.
Introduction
Ischemic stroke is a primary type of stroke, characterized by obstruction of cerebral blood vessels leading to local brain tissue hypoxia and nutrient deprivation [1, 2]. This type of vascular brain injury imposes a significant health burden globally, with high incidence and mortality rates [3]. Strokes are mainly categorized into ischemic and hemorrhagic types, with ischemic stroke constituting the majority. Following acute stroke, secondary neuroinflammation is a critical pathological process that can both exacerbate brain injury leading to cell death and promote brain tissue repair and functional recovery [2, 4, 5].
In the pathological process of ischemic stroke, neuroinflammation and disruption of energy metabolism play key roles. The acute occurrence of stroke rapidly triggers a neuroinflammatory response, including the activation of microglia and macrophages, as well as leukocyte infiltration [6, 7]. These immune cells exacerbate local inflammation by releasing pro-inflammatory cytokines and chemokines. The inflammatory response has a dual role in brain injury. While it can worsen damage by releasing pro-inflammatory cytokines that cause neuronal death, it also helps clear necrotic tissue and promote repair [5, 8]. Concurrently, the interruption of oxygen and energy supply caused by cerebral ischemia triggers metabolic dysregulation, particularly mitochondrial dysfunction, leading to a reduction in adenosine triphosphate (ATP) production and subsequently affecting the stability of the intracellular environment [9, 10]. Mitochondrial dysfunction not only reduces cellular energy supply but also leads to excessive production of reactive oxygen species (ROS), which further damage cell membranes and organelles, inducing apoptosis and apoptotic and necrotic cell death. This energy metabolism imbalance severely affects neural cell function and interacts with neuroinflammation, accelerating ischemic brain injury [11, 12].
Despite progress in understanding these mechanisms, the multifactorial mechanisms of ischemic stroke, especially the interaction between neuroinflammation and energy metabolism, require further exploration. This review aims to integrate the current understanding of these mechanisms, with a particular focus on their interplay. We will analyze in detail the development of neuroinflammation following cerebral ischemia, including the release of inflammatory cytokines, activation of immune cells, and disruption of the blood–brain barrier (BBB) as key elements. Additionally, we will discuss the impact of energy metabolism dysregulation on neural cell function and how this dysregulation interacts with neuroinflammation to advance the progression of ischemic brain injury.
By exploring the interaction between neuroinflammation and energy metabolism, we aim to provide a theoretical basis for new IS treatments and guide future research. This will not only help elucidate the complex interplay between inflammation and energy metabolism but also provide a theoretical basis for the development of new therapeutic strategies and drugs.
Neuroinflammatory response in ischemic stroke
Ischemic stroke, a severe form of brain disease, involves a complex neuroinflammatory response in its pathophysiological process. Brain tissue under IS conditions suffers from hypoxia and nutrient deprivation, leading to cellular damage and death. This injury triggers the release of damage-associated molecular patterns (DAMPs), which, similar to pathogen-associated molecular patterns, can be recognized by pattern recognition receptors such as Toll-like receptors and inflammasomes, thereby activating the innate immune system and initiating a local inflammatory response. This response includes the activation of microglia and astrocytes, as well as the recruitment of peripheral immune cells. In the pathophysiology of ischemic stroke, the inflammatory response has a dual role: it may exacerbate brain injury or participate in the repair and functional recovery of brain tissue [13, 14]. Furthermore, the pathophysiological changes in ischemic stroke are a continuous and complex dynamic process involving energy impairment and ion imbalance, excitotoxicity of amino acids, oxidative stress, inflammatory responses, apoptosis and necrosis, reperfusion injury, and neuroregeneration and repair. The inflammatory response at the BBB and endothelial interface is particularly crucial, as it represents a core mechanism of ischemic tissue injury, involving the collective participation of adhesion molecules, cytokines, chemokines, and leukocytes, which is of significant importance to the pathogenesis of cerebral infarction tissue injury [15]. Neuroinflammation is triggered by immune cells like microglia, astrocytes, peripheral immune cells, and endothelial cells. These cells respond to injuries such as infection, ischemia, stress, and trauma by releasing inflammatory mediators, initiating neuroimmune responses [16] (Fig. 1).
Multidimensional interactions of pathological and physiological changes following ischemic stroke. This includes energy deficits and ionic imbalances, excitotoxicity of amino acids, oxidative stress, inflammatory responses, inducing both apoptotic and necrotic cell death, reperfusion injury, and nerve regeneration and repair. mPTP mitochondrial permeability transition pore. Glu: Glucose. ROS: reactive oxygen species
When the brain suffers ischemic injury, DAMPs signals activate local microglia and peripheral leukocytes, triggering the release of a large amount of pro-inflammatory cytokines. This activation also enhances the expression of leukocyte adhesion molecules and stimulates the production of chemokines. The permeability of the BBB is increased, facilitating the infiltration of leukocytes into the brain parenchyma to clear the substantial debris generated by cellular death [17]. Cytokines are crucial in this process. These pleiotropic polypeptides regulate cell activation, proliferation, and differentiation in the central nervous system [18]. Under normal brain conditions, cytokines are barely detectable due to their low expression levels, and their receptors are also expressed at minimal levels. Nonetheless, in the context of brain ischemic injury, these cytokines are rapidly and extensively upregulated [19], playing roles in both pro-inflammatory and anti-inflammatory mechanisms. Specifically, pro-inflammatory cytokines like interleukin- 1β(IL- 1β), tumor necrosis factor (TNF-α), and interleukin- 6 play a key role in stroke by stimulating inflammatory responses that can worsen brain injury. Anti-inflammatory cytokines, on the other hand, reduce inflammation by inhibiting the expression of these pro-inflammatory cytokines and modulating immune cell activity. However, Cytokine functions are not strictly pro-inflammatory or anti-inflammatory. Some cytokines can be neurotoxic in certain conditions, while others may have neuroprotective effects. Therefore, the balance between the beneficial and detrimental effects of cytokines depends heavily on the brain's physiological and biochemical environment [20].
It is noteworthy that the IL- 1 receptor antagonist plays a significant role in mitigating neuronal damage caused by stroke. Additionally, other cytokine blockers have also proven effective in reducing cerebral edema, shrinking infarct size, and promoting recovery after experimental stroke. These research findings further substantiate the pivotal role of cytokines in the pathophysiology of stroke. In particular, TNF-α is expressed in ischemic tissue and surrounding neuronal cells at the onset of the disease and continues to be expressed in macrophages within the infarcted tissue, potentially acting as a key mediator of toxic reactions [21]. In early ischemia, inflammatory mediators like tumor necrosis factor and interleukins rapidly increase, attracting neutrophils, monocytes, lymphocytes, and other immune cells to the ischemic area [22].
Following the occurrence of ischemic stroke, the central nervous system's primary immune cells—microglia—swiftly respond to local damage signals. As ischemia persists, these cells become activated by recognizing DAMPs and can be classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. M1 microglia release pro-inflammatory cytokines and neurotoxic mediators, worsening neuronal damage, while M2 microglia promote repair and anti-inflammatory responses [23]. Subsequently, these cells then produce more ROS and TNF-α [24], increasing C-X-C motif chemokine ligand 10 release by endothelial cells, leading to blood–brain barrier disruption and peripheral T lymphocyte infiltration that exacerbates brain injury [25]. However, during stroke recovery, microglia can promote tissue repair and functional recovery by releasing anti-inflammatory factors like transforming growth factor-beta(TGF-β) and interleukin- 10 (IL- 10), and by stimulating angiogenesis [26]. Concurrently, astrocytes actively participate in the immune response following stroke; cytokines, DAMPs, and ROS released by damaged cells stimulate astrocytes and alter their phenotype, inducing"reactive astrogliosis"[27]. Research shows that the P2Y1 receptor in astrocytes plays a key role after ischemic stroke. Damaged cells promote pro-inflammatory cytokine and chemokine production via the nuclear factor kappa B (NF-κB) pathway, mediated by p65 subunit phosphorylation. These molecular events worsen local inflammation and may also impact the brain's recovery process after infarction [28]. Furthermore, Astrocyte-derived exosomes can regulate neuronal responses by transferring microRNAs like miR- 378a- 5p, reducing neuronal damage and neuroinflammation [29].
Oligodendrocytes, crucial for maintaining myelin sheath integrity, play a significant role in inflammation and neurorestoration after ischemic brain injury [30]. Cerebral ischemia not only leads to the injury and death of oligodendrocytes but also triggers demyelination, further exacerbating neurological dysfunction [31]. During stroke recovery, the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) are essential for remyelination. These OPCs migrate to damaged areas, differentiate into mature oligodendrocytes, and promote axon remyelination [32].
Myeloid immune cells also play a key role in the inflammatory response after ischemic stroke. Macrophages and neutrophils are recruited from the blood to damaged brain tissue, guided by DAMPs and chemokines, where they actively contribute to the inflammatory response. Microglia and myeloid immune cells are closely linked. Microglia release inflammatory mediators that recruit and activate myeloid immune cells, while myeloid immune cells release mediators that affect microglia function. This complex interplay collectively drives the progression of inflammation after ischemic stroke [33], making brain tissue injury more complex and difficult to control. This type of immune cell interaction is not unique to ischemic stroke. In oncology, tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) play key roles in tumor immune evasion. TAMs can be polarized by tumor cells to promote tumor growth and immune escape, while MDSCs suppress effector immune cell function [34]. Similarly, in ischemic stroke, the interaction between microglia and myeloid immune cells significantly affects disease outcomes. Understanding these similarities can provide new perspectives for studying ischemic stroke.
After ischemic stroke, significant changes occur in TGF-β signaling in astrocytes and microglia/macrophages [35]. Research shows that during the subacute phase of stroke, inhibiting astrocyte TGF-β signaling prevents TGF-β upregulation in the peri-infarct cortex and increases innate immune cell infiltration and activation [36]. Previous studies indicate that extracellular vesicles from microglia preconditioned with oxygen–glucose deprivation (OGD) regulate the TGF-β/Smad 2/3 pathway, promoting tissue regeneration and neurorestoration in stroke mice. This mechanism provides anti-apoptotic and neuroprotective effects and may help reduce ischemic brain injury [37, 38]. This finding suggests a new therapeutic strategy: regulating TGF-β signaling to influence the immune response and neurorestoration after stroke.
Furthermore, the release of numerous cytokines and growth factors after ischemic stroke can activate the JAK/STAT3 signaling pathway [39]. Activating this pathway is crucial for regulating neuroinflammation. Resveratrol and tetrandrine can modulate the STAT3 pathway, reducing neuroinflammation [40, 41]. At the same time, the Notch signaling pathway also plays a key role in ischemic stroke, influencing astrocyte behavior and promoting neurorestoration by regulating neural stem cell proliferation and differentiation [42]. In addition, NF-κB activation is a key factor in neuronal loss after ischemic stroke. As a crucial transcription factor, NF-κB regulates many inflammation-related genes, playing a central role in the inflammatory response [43]. In recent years, increasing evidence has shown that triggering receptor expressed on myeloid cells 2 (TREM 2), an important immune receptor expressed on myeloid cells, exerts a strong neuroprotective effect in experimental ischemic stroke. TREM2, an immunoglobulin-like receptor [44], is mainly expressed on microglia in the central nervous system and protects neurons by reducing neuroinflammation caused by OGD-induced injury [45, 46]. These complex molecular signaling pathways work together to regulate neuroinflammation and repair processes after ischemic stroke.
The neuroinflammatory response following ischemic stroke is a complex and dynamic process with dual roles: it can exacerbate brain injury while also promoting repair and protection. As mentioned earlier, the protective effects of neuroinflammation are primarily observed during the recovery phase, where it aids in tissue repair and functional recovery by clearing necrotic tissue, promoting angiogenesis, and facilitating neural regeneration. For example, M2 microglia and anti-inflammatory cytokines like TGF-β and IL- 10 play a key role in later stroke stages by reducing excessive inflammation and promoting tissue repair. However, the contradiction between the protective and damaging effects of neuroinflammation persists. On one hand, early inflammation can worsen neuronal damage by releasing pro-inflammatory cytokines like IL- 1β and TNF-α, and ROS. On the other hand, moderate inflammation in the later stages may help mitigate damage by clearing dead cells and promoting repair. This contradiction may stem from the temporal dynamics, cell-type specificity, and local microenvironment of the inflammatory response [47]. Future research should focus on the temporal and spatial dynamics of neuroinflammation to clarify the specific mechanisms at different stages. For instance, shifting microglia from M1 to M2 or using anti-inflammatory drugs like IL- 1 receptor antagonists could reduce early inflammation while promoting later repair. Additionally, integrating multi-omics and single-cell sequencing technologies to investigate the roles of different cell types in the inflammatory response may provide new insights to resolve this contradiction. In summary, precise regulation of the spatiotemporal dynamics of neuroinflammation holds promise for achieving better neuroprotection and functional recovery in stroke treatment.
Energy metabolism dysregulation induced by ischemic stroke
Cerebral energy metabolism is crucial for maintaining overall body function. Despite the brain's lack of significant energy reserves, its activity relies on a delicate regulation between neurotransmission and energy metabolism. The brain meets the high energy demand during neurotransmission through a continuous supply of metabolic fuels, ensuring the efficient operation of the organism's functions. This precision in energy management guarantees that the brain can maintain efficient energy utilization and stable functional output across different activities and physiological states [48]. Based on this, brain energy metabolism exhibits a unique characteristic of high metabolic rate compared to other tissues. Further research indicates that this high metabolic rate is primarily attributed to the activity of plasma membrane ion pumps and postsynaptic receptors. Moreover, a key observation is that increased local brain activity directly correlates with higher glucose use in that brain region, showing how sensitive and adaptable brain energy metabolism is to local neural activity [49].
Glucose assumes a pivotal role in energy metabolism within the adult brain, acting as the main source of energy for the nervous system. Under specific conditions, although the brain can use other energy sources under certain conditions, glucose enters cells via GLUTs and is converted into glucose- 6-phosphate, which is then metabolized through glycolysis, the pentose phosphate pathway, and glycogen synthesis. These processes produce carbon dioxide, water, and metabolic intermediates that release energy, supporting neurotransmitter synthesis and other brain functions [50]. In this metabolic process, the connection between neurotransmitters and energy metabolism presents complex multi-level interactions. Studies have shown that acetylcholine (ACh) and its receptor, the α7 nicotinic acetylcholine receptor (α7nAChR), play a role in the arterial baroreflex and have a protective effect against stroke, possibly by affecting cell death and inflammatory responses [51]. Furthermore, plasma amino acid neurotransmitters, including glutamate, aspartate, γ-aminobutyric acid, and glycine, are associated with poor outcomes in ischemic stroke, and changes in these neurotransmitter levels may play a regulatory role in energy metabolism and neuroprotection after stroke [52]. Further research shows that the α7nAChR agonist GTS- 21 can reduce brain injury and glucose metabolic dysregulation in ischemic stroke, likely by lowering oxidative stress, inflammation, and improving mitochondrial function and energy metabolism [53]. These findings emphasize that neurotransmitters may have a direct impact on the energy metabolic process after stroke, in addition to their role in neural signal transmission.
In the context of cerebral ischemia, the equilibrium of glucose metabolism is disrupted. Local ischemia increases anaerobic glycolysis, rapidly lowering intracellular pH, leading to cellular dysfunction and swelling. To counteract the pH drop, cells expel excess H+ through faster Na+/H+ exchange, increasing Na+ influx. This ion exchange affects cell volume regulation and is linked to cellular excitability and neuroinflammatory responses, which play a key role in post-ischemic pathology [54]; in global cerebral ischemia, mitochondrial ATP synthesis is hindered, causing rapid ATP depletion. This triggers neuronal membrane depolarization, increased potassium efflux, and sodium influx, disrupting Ca2⁺-ATPase function and leading to high intracellular calcium levels [12, 55]. Under ischemic conditions, due to the lack of oxygen and nutrient supply, mitochondrial oxidative phosphorylation is impaired [56], resulting in reduced ATP production by mitochondria, affecting ATP-dependent Ca2⁺ channel function, leading to cellular and mitochondrial Ca2⁺ accumulation or overload. Additionally, during ischemia, adenine nucleotides degrade, decreasing their concentration and increasing phosphate levels, making the mitochondrial permeability transition pore (mPTP) more sensitive to Ca2⁺ [57]. Under no-flow conditions, cells rely on anaerobic respiration, leading to lactate accumulation and a decrease in cellular pH, further triggering mPTP closure [58, 59]. These changes collectively affect the metabolic state and mitochondrial function of cells under ischemic conditions.
Under ischemic conditions, reduced cellular energy can impair key functions, including Na+/K+ pump maintenance, widespread depolarization, and excessive glutamate release. In this state, the overactivation of N-methyl-D-aspartate receptor-type channels in L-glutamate (L-glu) channels mediates a large influx of Ca2⁺, leading to cellular excitotoxicity and neuronal death. A significant increase in intracellular free calcium activates a series of calcium-sensitive enzymes, such as calpains, which further activate other molecules and disrupt cellular structures, exacerbating cellular damage [50]. L-glutamate, as the dominant excitatory neurotransmitter in the brain, is widely involved in almost all activities of the nervous system. During ATP depletion, glutamate transport to presynaptic vesicles weakens. However, high extracellular glutamate or excitatory amino acid levels overstimulate glutamate receptors, causing excitotoxicity, a key pathological process in ischemic diseases [60, 61].
The energy metabolic changes in ischemic stroke underscore the brain's susceptibility to energy deprivation crises. These complex processes show the central role of energy metabolism in the brain and how ischemia disrupts intracellular stability. With in-depth research into the mechanisms of ischemic stroke, we hope to develop more effective therapeutic measures to block or reverse these pathological processes, thereby reducing brain damage and promoting the recovery of neurological functions. Future research should focus on regulating energy metabolism during ischemia and optimizing it through drugs or other interventions, offering new approaches for ischemic stroke treatment.
Energy metabolism dysregulation induced by ischemic stroke is a core mechanism of brain injury. The interruption of oxygen and energy supply due to cerebral ischemia triggers mitochondrial dysfunction, reduced ATP production, and intracellular environmental imbalance, leading to apoptosis and necrotic cell death. Disruptions in glucose metabolism, glutamate excitotoxicity, and excessive ROS production further exacerbate neuronal damage. Dysregulation of energy metabolism not only directly affects neuronal survival and function but also interacts with neuroinflammation, jointly driving the progression of ischemic brain injury. Therefore, future research should focus on how to regulate energy metabolism under ischemic conditions to mitigate brain damage and promote functional recovery.
Interplay between neuroinflammation and energy metabolism dysregulation
The interaction between neuroinflammation and energy metabolism dysregulation is complex in ischemic stroke. The disruption of oxygen and energy supply during cerebral ischemia triggers metabolic disarray [62], with mitochondrial dysfunction leading to a decrease in ATP production, affecting active transport processes within the cell, causing a decline in the function of ion pumps, and subsequently leading to an imbalance in the intracellular environment [57]; this imbalance activates energy sensors such as 5'-AMP-activated protein kinase (AMPK) [63],and influences signaling pathways like Mammalian/mechanistic target of rapamycin (mTOR) [64]. Research shows that autophagy is activated in HT22 cells treated with Oxygen Glucose Deprivation/Reperfusion (OGD/R). Rapamycin, an autophagy inducer, enhances cell viability, reduces injury, and alleviates apoptosis, suggesting that appropriately enhanced autophagy exerts a protective effect on cells [65]. Further studies have revealed that OGD/R activates the AMPK/DDiT4/mTOR axis, which increases autophagy and reduces cell damage, indicating its significant importance in neuroprotection [66]. Additionally, in a model of cerebral ischemia–reperfusion injury, Nek6 expression is upregulated. Overexpression of Nek6 activates the mTOR signaling pathway, inhibits autophagy, and reduces neuronal damage Moreover, Nek6 mRNA undergoes post-transcriptional regulation through m6 A modification, which affects its expression levels [67]. Moreover, this state of energy deprivation activates Toll-like receptor 4 (TLR4) on astrocytes and microglia, leading to the initiation of an inflammatory response. The activation of TLR4 promotes the production of inflammatory cytokines, ROS, and nitric oxide (NO), all of which contribute to neurodamage associated with stroke [68]. Simultaneously, within the ischemic penumbra, the activation of platelets and the complement system, as well as the release of inflammatory mediators, induce metabolic pathways to overcompensate for ischemic damage, leading to an excessive release of ROS [69, 70]. These ROS activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and inflammasomes involved in the inflammatory response, such as the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, resulting in the release of inflammatory mediators like the cytokine IL- 1β. This further exacerbates the neuroinflammatory response, which not only increases brain tissue damage but may also lead to long-term neurological dysfunction [71].
Following the restoration of blood supply after cerebral ischemia, glucose and oxygen levels gradually recover, while the overproduction of ROS triggers oxidative stress responses. Such responses promote the infiltration of leukocytes, the pro-inflammatory effects of neutrophils, and the activation of complement and platelets, leading to the disruption of the BBB [72]. This allows more inflammatory cells and mediators to enter brain tissue [73], activating microglia and astrocytes, which release more inflammatory mediators, creating a positive feedback loop that worsens neuroinflammation [74]. At the same time, increased ROS causes the mPTP to open, which both causes and results from mPTP opening. High levels of tumor necrosis factor TNF-α/MLR can upregulate the production of ROS [75]. Excessive ROS induce the opening of the mitochondrial outer membrane pores, releasing calcium ions, cytochrome C, and apoptosis-inducing factor AIF, leading to apoptosis [76]. These processes can disrupt nutrient and metabolic substance transport, impairing energy metabolism and affecting brain tissue recovery and function [77] (Fig. 2).
The interplay between neuroinflammation and energy metabolism dysregulation. In ischemic stroke, oxygen and energy deprivation cause mitochondrial dysfunction, reducing ATP and ion pump function, leading to cellular imbalance. This disruption activates energy sensors such as AMPK and signaling pathways like mTOR and TLR4, triggering inflammation and ROS release. Platelet and complement activation in the penumbra, coupled with the excessive release of inflammatory mediators and ROS, triggers NADPH oxidase and NLRP3 inflammasomes, exacerbating neuroinflammation and brain damage. After blood supply restoration, excessive ROS induce oxidative stress, compromise the blood–brain barrier, and facilitate the entry of inflammatory cells and mediators into the brain tissue, creating a positive feedback loop that intensifies neuroinflammation. The increase in ROS also leads to the opening of mitochondrial permeability transition pores, releasing apoptotic factors and inducing apoptosis, which affects the recovery and function of brain tissue. Dashed lines: Positive feedback."+": Promotion."X": Destruction
In addition, Vitamin B12 deficiency can disrupt immune homeostasis, increasing inflammation. For example, homocysteine, a pro-inflammatory amino acid, can promote atherosclerosis and raise the risk of ischemic stroke [78]. The CCL5/CCR5 axis is key in post-stroke neuroinflammation, and CCR5 activation may worsen neuroinflammation via the JAK2/STAT3 pathway, affecting energy metabolism [79]. Ischemic stroke also disrupts the liver's role in lipid metabolism, which is closely tied to glucose and glutathione homeostasis. Lipoproteins produced by the liver, such as very-low-density lipoprotein, may play an important role in lipid supply after cerebral ischemia, affecting cell survival, neurogenesis, and anti-inflammatory pathways [80]. It is noteworthy that PLA2G2E-mediated lipid metabolism triggers an autonomous neural repair process within the brain following cerebral ischemia. This process induces PAD4 expression in neurons via dihomo-g-linolenic acid and 15-hydroxy-eicosatrienoic acid production, promoting gene expression linked to stroke recovery. These lipid metabolic products are not only crucial for preventing additional cell death and subsequent severe inflammation but may also regulate energy metabolism by affecting mitochondrial function and oxidative phosphorylation processes [81].
This bidirectional process worsens brain tissue damage and impairs recovery and function. Accordingly, a deep understanding of this complex interplay network is crucial for the development of effective therapeutic strategies. For instance, Resolvin D1 promotes oxidative phosphorylation and inhibits glycolysis, increasing ATP production in microglia and enhancing their ability to phagocytose neutrophils. This metabolic reprogramming boosts energy production, reduces neutrophil accumulation, and prevents neutrophil extracellular trap formation, alleviating neuroinflammation [82]. By identifying and intervening in the key components of energy metabolism disorders and neuroinflammation, new strategies for the treatment of ischemic stroke can be provided. These strategies may include protecting mitochondrial function, modulating the release of inflammatory mediators, repairing the BBB, and optimizing metabolic pathways to alleviate brain damage and promote the recovery of brain function.
The interplay between neuroinflammation and energy metabolism dysregulation in ischemic stroke forms a complex pathological network. Energy supply disruption directly causes cellular dysfunction and triggers neuroinflammation by activating pathways like AMPK, mTOR, and TLR4. Excessive ROS and inflammatory mediators worsen brain injury, creating a positive feedback loop that reinforces inflammation and metabolic dysregulation. Therefore, understanding the key nodes of this interaction network, particularly how to modulate mitochondrial function, inhibit inflammatory mediator release, and repair the blood–brain barrier, is crucial for developing effective therapeutic strategies.
Nutrition and pharmacotherapy: improving neuroinflammation and dysregulation of energy metabolism
Neuroinflammation and dysregulation of energy metabolism following stroke are critical pathological processes leading to neuronal damage and functional impairment. Studies have shown that sodium butyrate and Guhong injection alleviate neuroinflammation by modulating the gut-brain axis, inhibiting the TLR4/MyD88/NF-κB signaling pathway, reducing pro-inflammatory microglia, and increasing anti-inflammatory microglia [83]. Additionally, these interventions improve energy metabolism pathways (such as glycerophospholipid metabolism, the tricarboxylic acid cycle, and glycolysis) by regulating the production of short-chain fatty acids (SCFAs), reducing oxidative stress and neuronal apoptosis, thereby promoting post-stroke neurological recovery [84]. Probiotics like Lactobacillus reuteri GMNL- 89 and Lactobacillus paracasei GMNL- 133 show potential as stroke adjunctive therapies by modulating the gut-spleen-brain axis, reducing infarct volume, improving neurological function, protecting the blood–brain and intestinal barriers, and reducing inflammation through immune regulation and gut microbiota balance [85]. Salvianolic acid C (SalC) significantly alleviates post-stroke neuroinflammation and protects neurons from ischemia–reperfusion injury by inhibiting the TLR4-TREM1-NF-κB signaling pathway. Furthermore, SalC improves post-stroke neurological function by regulating microglial inflammatory responses and reducing the release of pro-inflammatory factors, offering a novel pharmacological intervention strategy for neuroinflammation treatment [86]. Retinoic acid significantly suppresses post-stroke neuroinflammation and apoptosis by modulating the PI3 K-Akt signaling pathway and the interaction of Bcl- 2 family proteins [87]. Vitamin D has anti-inflammatory and immunomodulatory effects. While its ability to reduce cardiovascular disease and mortality is limited [88], its potential for treating post-stroke neuroinflammation and energy metabolism dysregulation deserves further study.
In terms of drug delivery systems, Polyamidoamine dendrimers encapsulating curcumin significantly reduce astrocyte reactivity, alleviate neuroinflammation, and promote functional recovery [89]. In particular, the RVG29 peptide-modified Cur@GAR NPs enhance brain targeting, effectively reduce neuroinflammation and neuronal apoptosis, and mitigate oxidative stress by scavenging free radicals, indirectly improving mitochondrial function and energy metabolism [90]. ROS generated during cerebral ischemia–reperfusion activate the AKT/SP1-NFκB pathway, increasing SURI expression and triggering cellular edema and necrosis. Antioxidants such as resveratrol (RSV) alleviate neuroinflammation and cellular damage by inhibiting ROS generation and modulating SURI expression [91]. RSV preconditioning (RPC) reduces post-ischemic cell death and mitigates NAD+/NADH level loss by downregulating PARP1 protein expression, thereby inhibiting AIF nuclear translocation and reducing neuroinflammation and apoptosis. The long-term neuroprotective effects of RPC may be achieved through energy metabolism regulation and PARP1 overactivation inhibition [92]. Additionally, polyphenolic compounds like quercetin and gallic acid in Moringa oleifera leaf extracts have anti-inflammatory and antioxidant properties, potentially inhibiting inflammatory factor release and modulating energy metabolism-related gene expression [93]. Studies on ω3 and ω6 polyunsaturated fatty acids (PUFAs) indicate that lower ω6 PUFA levels (e.g., arachidonic acid, AA) are associated with increased atrial fibrillation risk, while AA has anti-inflammatory and cardioprotective effects, potentially improving stroke outcomes by suppressing neuroinflammation and reducing atrial fibrosis [94]. Post-stroke, ceramides accumulate in astrocytes, triggering neuroinflammation and energy metabolism dysregulation by disrupting mitochondrial integrity and activating the cGAS/STING pathway. Inhibition of SPTLC2 or the use of antioxidants (e.g., SKQ1) reduces ceramide production and mitochondrial oxidative stress, thereby alleviating inflammation and brain damage [95]. These findings provide new therapeutic strategies for addressing neuroinflammation through nutritional and pharmacological interventions.
Nutritional and pharmacological interventions show great potential in improving neuroinflammation and energy metabolism dysregulation after stroke. By modulating the gut-brain axis, inhibiting inflammatory pathways like TLR4/NF-κB, enhancing mitochondrial function, and reducing oxidative stress, various interventions can effectively reduce neuroinflammation and improve energy metabolism. Probiotics, antioxidants like resveratrol, and specific fatty acid supplements not only reduce brain damage but also promote neurological recovery. These studies offer new directions for stroke treatment, and future research should explore the best timing and dosage of these interventions to maximize their effectiveness.
Conclusions and prospects
This review discusses the interplay between neuroinflammation and energy metabolism dysregulation in ischemic stroke, emphasizing their importance in the disease's pathophysiology. Neuroinflammatory responses not only exacerbate brain injury during the acute phase but also profoundly affect brain tissue repair and functional recovery during the recovery phase. At the same time, the disruption of energy metabolism significantly impacts the survival and functional stability of neural cells, further aggravating the extent of ischemic damage.
With the widespread application of advanced technologies such as single-cell RNA sequencing (scRNA-seq), we have gained a more precise understanding of the responses of different cell types following ischemic stroke. Using scRNA-seq, the dynamic changes in microglia and astrocytes post-stroke have been revealed, particularly the role of SLC7 A11 in regulating microglial polarization. Additionally, studies have found that overexpression of CD73 can alleviate astrocyte pyroptosis induced by cerebral ischemia–reperfusion through the A2B/NF-κB signaling pathway [96, 97]. These findings further confirm the critical role of neuroinflammation and energy metabolism in the pathological mechanisms of ischemic stroke and provide potential targets for future precision therapies.
Future research should focus on elucidating the underlying mechanisms of the interaction between neuroinflammation and energy metabolism dysregulation, as well as exploring new therapeutic targets. By gaining a deeper understanding of these complex biological processes and validating their therapeutic effects through clinical trials, we can develop new treatment modalities aimed at modulating neuroinflammatory responses and improving energy metabolism. These modalities may include inhibitors targeting specific inflammatory mediators, drugs that enhance mitochondrial function, and strategies to protect and repair the BBB. The common goal of these strategies is to mitigate brain injury, promote neurological recovery, and improve long-term patient outcomes. At the same time, the importance of clinical laboratory science in disease diagnosis cannot be overlooked. Establishing comprehensive academic programs to train professionals is crucial for advancing medical innovation in diseases like ischemic stroke [98, 99]. As we better understand these key pathophysiological mechanisms, we expect to provide more effective treatments for ischemic stroke patients.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ATP:
-
Adenosine triphosphate
- ROS:
-
Reactive oxygen species
- BBB:
-
Blood–brain barrier
- DAMPs:
-
Damage-associated molecular patterns
- IL- 1β:
-
Interleukin- 1β
- TNF-α:
-
Tumor necrosis factor
- TGF-β:
-
Transforming growth factor-beta
- IL- 10:
-
Interleukin- 10
- NF-κB:
-
Nuclear factor kappa B
- OPCs:
-
Oligodendrocyte precursor cells
- TAMs:
-
Tumor-associated macrophages
- MDSCs:
-
Myeloid-derived suppressor cells
- OGD:
-
Oxygen–glucose deprivation
- OGD/R:
-
Oxygen-glucose deprivation/reperfusion
- JAK/STAT 3:
-
Janus kinase/signal transducer and activator of transcription 3
- TREM 2:
-
Triggering receptor expressed on myeloid cells 2
- GLUTs:
-
Glucose transporter proteins
- ACh:
-
Acetylcholine
- α7nAChR:
-
α7 Nicotinic acetylcholine receptor
- GTS- 21:
-
α7nAChR agonist
- mPTP:
-
Mitochondrial permeability transition pore
- L-glu:
-
L-glutamate
- AMPK:
-
5'-AMP-activated protein kinase
- mTOR:
-
Mammalian/mechanistic target of rapamycin
- TLR4:
-
Toll-like receptor 4
- NO:
-
Nitric oxide
- NADPH:
-
Nicotinamide adenine dinucleotide phosphate
- NLRP3:
-
NOD-like receptor family pyrin domain containing 3
- SCFA:
-
Short-chain fatty acids
- SalC:
-
Salvianolic acid C
- RVG29:
-
Rabies virus glycoprotein
- Cur@GAR NPs:
-
Cur-loaded gelatin nanoparticles
- RSV:
-
Resveratrol
- RPC:
-
Resveratrol preconditioning
- PARP1:
-
Promotes poly-ADP-ribose polymerase 1
- PUFAs:
-
Polyunsaturated fatty acids
- AA:
-
Arachidonic acid
References
Shehjar F, Maktabi B, Rahman ZA, Bahader GA, James AW, Naqvi A, et al. Stroke: molecular mechanisms and therapies: update on recent developments. Neurochem Int. 2023;162: 105458. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuint.2022.105458.
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil extracellular traps in cerebral ischemia/reperfusion injury: friend and foe. Curr Neuropharmacol. 2023;21:2079. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1570159X21666230308090351.
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, et al. 2024 heart disease and stroke statistics: a report of US and global data from the American Heart Association. Circulation. 2024;149:e347–913. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/CIR.0000000000001209.
Kuriakose D, Xiao Z. Pathophysiology and treatment of stroke: present status and future perspectives. Int J Mol Sci. 2020;21:7609. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21207609.
Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation. 2019;16:142. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-019-1516-2.
Liu Z, Qin Q, Wang S, Kang X, Liu Y, Wei L, et al. STING activation in macrophages and microglia drives poststroke inflammation: implications for neuroinflammatory mechanisms and therapeutic interventions. CNS Neurosci Ther. 2024;30: e70106. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cns.70106.
Dabrowska S, Andrzejewska A, Lukomska B, Janowski M. Neuroinflammation as a target for treatment of stroke using mesenchymal stem cells and extracellular vesicles. J Neuroinflammation. 2019;16:178. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-019-1571-8.
Lambertsen KL, Finsen B, Clausen BH. Post-stroke inflammation—target or tool for therapy? Acta Neuropathol. 2019;137:693–714. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00401-018-1930-z.
Tian H, Chen X, Liao J, Yang T, Cheng S, Mei Z, et al. Mitochondrial quality control in stroke: from the mechanisms to therapeutic potentials. J Cell Mol Med. 2022;26:1000–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jcmm.17189.
Pham L, Arroum T, Wan J, Pavelich L, Bell J, Morse PT, et al. Regulation of mitochondrial oxidative phosphorylation through tight control of cytochrome c oxidase in health and disease—implications for ischemia/reperfusion injury, inflammatory diseases, diabetes, and cancer. Redox Biol. 2024;78:103426. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2024.103426.
Alshial EE, Abdulghaney MI, Wadan A-HS, Abdellatif MA, Ramadan NE, Suleiman AM, et al. Mitochondrial dysfunction and neurological disorders: a narrative review and treatment overview. Life Sci. 2023;334:122257. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2023.122257.
Yuan Y, Chen T, Yang Y, Han H, Xu L. E2F1/CDK5/DRP1 axis mediates microglial mitochondrial division and autophagy in the pathogenesis of cerebral ischemia-reperfusion injury. Clin Transl Med. 2025;15: e70197. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/ctm2.70197.
Doyle KP, Simon RP, Stenzel-Poore MP. Mechanisms of ischemic brain damage. Neuropharmacology. 2008;55:310–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuropharm.2008.01.005.
Castellanos-Molina A, Bretheau F, Boisvert A, Bélanger D, Lacroix S. Constitutive DAMPs in CNS injury: From preclinical insights to clinical perspectives. Brain Behav Immun. 2024;122:583–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2024.07.047.
Kvistad CE, Kråkenes T, Gavasso S, Bø L. Neural regeneration in the human central nervous system—from understanding the underlying mechanisms to developing treatments. Where do we stand today? Front Neurol. 2024;15:1398089. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fneur.2024.1398089.
Zhu H, Hu S, Li Y, Sun Y, Xiong X, Hu X, et al. Interleukins and ischemic stroke. Front Immunol. 2022;13: 828447. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.828447.
Stanzione R, Forte M, Cotugno M, Bianchi F, Marchitti S, Rubattu S. Role of DAMPs and of leukocytes infiltration in ischemic stroke: insights from animal models and translation to the human disease. Cell Mol Neurobiol. 2022;42:545–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10571-020-00966-4.
Dordoe C, Huang W, Bwalya C, Wang X, Shen B, Wang H, et al. The role of microglial activation on ischemic stroke: Modulation by fibroblast growth factors. Cytokine Growth Factor Rev. 2023;74:122–33. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cytogfr.2023.07.005.
Fan W, Chen H, Li M, Fan X, Jiang F, Xu C, et al. NRF2 activation ameliorates blood–brain barrier injury after cerebral ischemic stroke by regulating ferroptosis and inflammation. Sci Rep. 2024;14:5300. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-53836-0.
Clausen BH, Wirenfeldt M, Høgedal SS, Frich LH, Nielsen HH, Schrøder HD, et al. Characterization of the TNF and IL-1 systems in human brain and blood after ischemic stroke. Acta Neuropathol Commun. 2020;8:81. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40478-020-00957-y.
Keuters MH, Keksa-Goldsteine V, Rõlova T, Jaronen M, Kettunen P, Halkoluoto A, et al. Benserazide is neuroprotective and improves functional recovery after experimental ischemic stroke by altering the immune response. Sci Rep. 2024;14:17949. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-68986-4.
Pawluk H, Kołodziejska R, Grześk G, Kozakiewicz M, Woźniak A, Pawluk M, et al. Selected mediators of inflammation in patients with acute ischemic stroke. Int J Mol Sci. 2022;23:10614. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms231810614.
Zeng J, Bao T, Yang K, Zhu X, Wang S, Xiang W, et al. The mechanism of microglia-mediated immune inflammation in ischemic stroke and the role of natural botanical components in regulating microglia: a review. Front Immunol. 2023;13:1047550. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.1047550.
Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial cells: role of the immune response in ischemic stroke. Front Immunol. 2020;11:294. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2020.00294.
Cui P, Lu W, Wang J, Wang F, Zhang X, Hou X, et al. Microglia/macrophages require vitamin D signaling to restrain neuroinflammation and brain injury in a murine ischemic stroke model. J Neuroinflammation. 2023;20:63. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-023-02705-0.
Mihailova V, Stoyanova II, Tonchev AB. Glial populations in the human brain following ischemic injury. Biomedicines. 2023;11:2332. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines11092332.
Rahman MS, Islam R, Bhuiyan MIH. Ion transporter cascade, reactive astrogliosis and cerebrovascular diseases. Front Pharmacol. 2024;15:1374408. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2024.1374408.
Shigetomi E, Suzuki H, Hirayama YJ, Sano F, Nagai Y, Yoshihara K, et al. Disease-relevant upregulation of P2Y1 receptor in astrocytes enhances neuronal excitability via IGFBP2. Nat Commun. 2024;15:6525. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-50190-7.
Sun R, Liao W, Lang T, Qin K, Jiao K, Shao L, et al. Astrocyte-derived exosomal miR-378a-5p mitigates cerebral ischemic neuroinflammation by modulating NLRP3-mediated pyroptosis. Front Immunol. 2024;15:1454116. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2024.1454116.
Huang S, Ren C, Luo Y, Ding Y, Ji X, Li S. New insights into the roles of oligodendrocytes regulation in ischemic stroke recovery. Neurobiol Dis. 2023;184: 106200. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2023.106200.
Deng X, Hu Z, Zhou S, Wu Y, Fu M, Zhou C, et al. Perspective from single-cell sequencing: is inflammation in acute ischemic stroke beneficial or detrimental? CNS Neurosci Ther. 2024;30: e14510. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cns.14510.
Maimaiti M, Li C, Cheng M, Zhong Z, Hu J, Yang L, et al. Blocking cGAS-STING pathway promotes post-stroke functional recovery in an extended treatment window via facilitating remyelination. Med. 2024;5:622-644.e8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.medj.2024.03.018.
Aghapour SA, Torabizadeh M, Bahreiny SS, Saki N, Jalali Far MA, Yousefi-Avarvand A, et al. Investigating the dynamic interplay between cellular immunity and tumor cells in the fight against cancer: an updated comprehensive review. Iran J Blood Cancer. 2024;16:84–101. https://doiorg.publicaciones.saludcastillayleon.es/10.61186/ijbc.16.2.84.
Tang Y, Dong M-H, Pang X-W, Zhang H, Chu Y-H, Zhou L-Q, et al. Macrophage exosomal miR-30c-2-3p in atherosclerotic plaques aggravates microglial neuroinflammation during large-artery atherosclerotic stroke via TGF-β/SMAD2 pathway. J Neuroinflammation. 2024;21:292. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-024-03281-7.
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, et al. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther. 2024;9:61. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-024-01764-w.
Pan J-J, Qi L, Wang L, Liu C, Song Y, Mamtilahun M, et al. M2 microglial extracellular vesicles attenuated blood-brain barrier disruption via MiR-23a-5p in cerebral ischemic mice. Aging Dis. 2024;15:1344–56. https://doiorg.publicaciones.saludcastillayleon.es/10.14336/AD.2023.0714.
Zhang L, Wei W, Ai X, Kilic E, Hermann DM, Venkataramani V, et al. Extracellular vesicles from hypoxia-preconditioned microglia promote angiogenesis and repress apoptosis in stroke mice via the TGF-β/Smad2/3 pathway. Cell Death Dis. 2021;12:1068. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-021-04363-7.
Nath S, Martínez Santamaría JC, Chu Y-H, Choi JS, Conforti P, Lin J-D, et al. Interaction between subventricular zone microglia and neural stem cells impacts the neurogenic response in a mouse model of cortical ischemic stroke. Nat Commun. 2024;15:9095. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-024-53217-1.
Zhong Y, Yin B, Ye Y, Dekhel OYAT, Xiong X, Jian Z, et al. The bidirectional role of the JAK2/STAT3 signaling pathway and related mechanisms in cerebral ischemia-reperfusion injury. Exp Neurol. 2021;341: 113690. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.expneurol.2021.113690.
Song J, Liu Y, Guo Y, Yuan M, Zhong W, Tang J, et al. Therapeutic effects of tetrandrine in inflammatory diseases: a comprehensive review. Inflammopharmacology. 2024;32:1743–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10787-024-01452-9.
Wang P, Li Z, Song Y, Zhang B, Fan C. Resveratrol-driven macrophage polarization: unveiling mechanisms and therapeutic potential. Front Pharmacol. 2025;15:1516609. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2024.1516609.
Mohammadi A, Balduini W, Carloni S. Melatonin modulates the Notch1 signaling pathway and Sirt3 in the hippocampus of hypoxic-ischemic neonatal rats. Sci Rep. 2024;14:25069. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-76307-y.
Lu W, Chen Z, Wen J. The role of RhoA/ROCK pathway in the ischemic stroke-induced neuroinflammation. Biomed Pharmacother. 2023;165: 115141. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.115141.
Chen S, Peng J, Sherchan P, Ma Y, Xiang S, Yan F, et al. TREM2 activation attenuates neuroinflammation and neuronal apoptosis via PI3K/Akt pathway after intracerebral hemorrhage in mice. J Neuroinflammation. 2020;17:168. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-020-01853-x.
Xu J, Zhang L, Li M, He X, Luo J, Wu R, et al. TREM2 mediates physical exercise-promoted neural functional recovery in rats with ischemic stroke via microglia-promoted white matter repair. J Neuroinflammation. 2023;20:50. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-023-02741-w.
Gong Z, Guo J, Liu B, Guo Y, Cheng C, Jiang Y, et al. Mechanisms of immune response and cell death in ischemic stroke and their regulation by natural compounds. Front Immunol. 2024;14:1287857. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1287857.
Liu S, Cao X, Wu Z, Deng S, Fu H, Wang Y, et al. TREM2 improves neurological dysfunction and attenuates neuroinflammation, TLR signaling and neuronal apoptosis in the acute phase of intracerebral hemorrhage. Front Aging Neurosci. 2022;14: 967825. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnagi.2022.967825.
Bonvento G, Bolaños JP. Astrocyte-neuron metabolic cooperation shapes brain activity. Cell Metab. 2021;33:1546–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cmet.2021.07.006.
Felipe Barros L, Brown A, Swanson RA. Glia in brain energy metabolism: a perspective. Glia. 2018;66:1134–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/glia.23316.
Shichkova P, Coggan JS, Markram H, Keller D. Brain metabolism in health and neurodegeneration: the interplay among neurons and astrocytes. Cells. 2024;13:1714. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells13201714.
Liu L, Wu Z, Lu Y, Lu W, Su G, Zhou Z. Effects of phototherapy on biopterin, neopterin, tryptophan, and behavioral neuroinflammatory reaction in patients with post-stroke depression. Sci Rep. 2024;14:18368. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-68799-5.
Zhu Z, et al. Plasma amino acid neurotransmitters and ischemic stroke prognosis: a multicenter prospective study. Am J Clin Nutr. 2023;118:754–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ajcnut.2023.06.014.
Wang Y-Y, Lin S-Y, Chang C-Y, Wu C-C, Chen W-Y, Huang W-C, et al. α7 nicotinic acetylcholine receptor agonist improved brain injury and impaired glucose metabolism in a rat model of ischemic stroke. Metab Brain Dis. 2023;38:1249–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11011-023-01167-w.
Baranovicova E, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Lehotsky J. Blood and brain metabolites after cerebral ischemia. Int J Mol Sci. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms242417302.
Li Y, Zhang M, Lin J, Guo H, Zhou H, Jin Y, et al. Mitochondrial ATP synthesis and proton transport synergistically mitigate oligodendrocyte progenitor cell dysfunction following transient middle cerebral artery occlusion via the Pbx3/Dguok/Kif21b signaling pathway. Int J Med Sci. 2024;21:2189–200. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijms.100127.
Huang J, Chen L, Yao Z, Sun X, Tong X, Dong S. The role of mitochondrial dynamics in cerebral ischemia-reperfusion injury. Biomed Pharmacother. 2023;162: 114671. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopha.2023.114671.
Bernardi P, Gerle C, Halestrap AP, Jonas EA, Karch J, Mnatsakanyan N, et al. Identity, structure, and function of the mitochondrial permeability transition pore: controversies, consensus, recent advances, and future directions. Cell Death Differ. 2023;30:1869–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41418-023-01187-0.
Rabinowitz JD, Enerbäck S. Lactate: the ugly duckling of energy metabolism. Nat Metab. 2020;2:566–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42255-020-0243-4.
Yang S-H, Sun Y, Berry R, Choudhury GR, Winters A, Chaudhari K, et al. Glutamate provides cytoprotective effect for astrocytes against ischemic insult and promotes astrogliosis. Aging Dis. 2024;15:2742–51. https://doiorg.publicaciones.saludcastillayleon.es/10.14336/AD.2023.0726.
Song HY, Jin S, Lee S, Jalin AMA, Roh K-H, Kim W-K. The therapeutic effects of SP-8356, a verbenone derivative, with multimodal cytoprotective mechanisms in an ischemic stroke rat model. Int J Mol Sci. 2024;25:12769. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms252312769.
Kalia M, Meijer HGE, Van Gils SA, Van Putten MJAM, Rose CR. Ion dynamics at the energy-deprived tripartite synapse. PLoS Comput Biol. 2021;17:e1009019. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pcbi.1009019.
Zhou X, Chen H, Wang L, Lenahan C, Lian L, Ou Y, et al. Mitochondrial dynamics: a potential therapeutic target for ischemic stroke. Front Aging Neurosci. 2021;13: 721428. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnagi.2021.721428.
Jiang S, Li T, Ji T, Yi W, Yang Z, Wang S, et al. AMPK: potential therapeutic target for ischemic stroke. Theranostics. 2018;8:4535–51. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.25674.
Melanis K, Stefanou M-I, Themistoklis KM, Papasilekas T. mTOR pathway—a potential therapeutic target in stroke. Ther Adv Neurol Disord. 2023;16:17562864231187770. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/17562864231187770.
Zhang J, Wang S, Zhang H, Yang X, Ren X, Wang L, et al. Drp1 acetylation mediated by CDK5-AMPK-GCN5L1 axis promotes cerebral ischemic injury via facilitating mitochondrial fission. Mol Med. 2024;30:173. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-024-00948-y.
Zhang Y, Liu L, Hou X, Zhang Z, Zhou X, Gao W. Role of autophagy mediated by AMPK/DDiT4/mTOR axis in HT22 cells under oxygen and glucose deprivation/reoxygenation. ACS Omega. 2023;8:9221–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsomega.2c07280.
Wang Q, Liu X, Yuan J, Yang T, Ding L, Song B, et al. Nek6 regulates autophagy through the mTOR signaling pathway to alleviate cerebral ischemia-reperfusion injury. Mol Brain. 2024;17:96. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13041-024-01166-7.
Oh S-A, Seol S-I, Davaanyam D, Kim S-W, Lee J-K. Platelet-derived HMGB1 induces NETosis, exacerbating brain damage in the photothrombotic stroke model. Mol Med. 2025;31:46. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10020-025-01107-7.
Al-Mufti F, Amuluru K, Roth W, Nuoman R, El-Ghanem M, Meyers PM. Cerebral ischemic reperfusion injury following recanalization of large vessel occlusions. Neurosurgery. 2018;82:781–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuros/nyx341.
Chen M, Liu J, Wu W, Guo T, Yuan J, Wu Z, et al. SIRT1 restores mitochondrial structure and function in rats by activating SIRT3 after cerebral ischemia/reperfusion injury. Cell Biol Toxicol. 2024;40:31. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10565-024-09869-2.
Simpson DSA, Oliver PL. ROS generation in microglia: understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants (Basel). 2020;9:743. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox9080743.
Zhao X, Li S, Mo Y, Li R, Huang S, Zhang A, et al. DCA protects against oxidation injury attributed to cerebral ischemia-reperfusion by regulating glycolysis through PDK2-PDH-Nrf2 Axis. Oxid Med Cell Longev. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2021/5173035.
Gao H, Chen H, Cui G-Y, Hu J-X. Damage mechanism and therapy progress of the blood-brain barrier after ischemic stroke. Cell Biosci. 2023;13:196. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-023-01126-z.
Tirandi A, Sgura C, Carbone F, Montecucco F, Liberale L. Inflammatory biomarkers of ischemic stroke. Intern Emerg Med. 2023;18:723–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11739-023-03201-2.
Li Y, Sun J, Wu R, Bai J, Hou Y, Zeng Y, et al. Mitochondrial MPTP: a novel target of ethnomedicine for stroke treatment by apoptosis inhibition. Front Pharmacol. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2020.00352.
He Z, Ning N, Zhou Q, Khoshnam SE, Farzaneh M. Mitochondria as a therapeutic target for ischemic stroke. Free Radical Biol Med. 2020;146:45–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.freeradbiomed.2019.11.005.
Qin C, Yang S, Chu Y-H, Zhang H, Pang X-W, Chen L, et al. Signaling pathways involved in ischemic stroke: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther. 2022;7:215. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-022-01064-1.
Roth W, Mohamadzadeh M. Vitamin B12 and gut-brain homeostasis in the pathophysiology of ischemic stroke. EBioMedicine. 2021;73: 103676. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2021.103676.
Lin J, Xu Y, Guo P, Chen Y-J, Zhou J, Xia M, et al. CCL5/CCR5-mediated peripheral inflammation exacerbates blood-brain barrier disruption after intracerebral hemorrhage in mice. J Transl Med. 2023;21:196. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-023-04044-3.
Inderhees J, Schwaninger M. Liver metabolism in ischemic stroke. Neuroscience. 2024;550:62–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2023.12.013.
Nakamura A, Sakai S, Taketomi Y, Tsuyama J, Miki Y, Hara Y, et al. PLA2G2E-mediated lipid metabolism triggers brain-autonomous neural repair after ischemic stroke. Neuron. 2023;111:2995-3010.e9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuron.2023.06.024.
Li L, Cheng S-Q, Sun Y-Q, Yu J-B, Huang X-X, Dong Y-F, et al. Resolvin D1 reprograms energy metabolism to promote microglia to phagocytize neutrophils after ischemic stroke. Cell Rep. 2023;42: 112617. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2023.112617.
Sun J, Lu L, Lian Y, Xu S, Zhu Y, Wu Y, et al. Sodium butyrate attenuates microglia-mediated neuroinflammation by modulating the TLR4/MyD88/NF-κB pathway and microbiome-gut-brain axis in cardiac arrest mice. Mol Brain. 2025;18:13. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13041-025-01179-w.
Mao P, Hu J, Mai X, Li N, Liao Y, Feng L, et al. Multi-omics analysis of the gut-brain axis elucidates therapeutic mechanisms of Guhong injection in the treatment of ischemic stroke. Int J Mol Sci. 2025;26:1560. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms26041560.
Wang Y-H, Liao J-M, Jan M-S, Wang M, Su H-H, Tsai W-H, et al. Prophylactic use of probiotics as an adjunctive treatment for ischemic stroke via the gut-spleen-brain axis. Brain Behav Immun. 2025;123:784–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2024.10.026.
Guo W, Xu X, Xiao Y, Zhang J, Shen P, Lu X, et al. Salvianolic acid C attenuates cerebral ischemic injury through inhibiting neuroinflammation via the TLR4-TREM1-NF-κB pathway. Chin Med. 2024;19:46. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13020-024-00914-0.
Kang J-B, Koh P-O. Retinoic acid alleviates the reduction of Akt and Bad phosphorylation and regulates Bcl-2 family protein interactions in animal models of ischemic stroke. PLoS ONE. 2024;19: e0303213. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0303213.
Sofianopoulou E, Kaptoge SK, Afzal S, Jiang T, Gill D, Gundersen TE, et al. Estimating dose—response relationships for vitamin D with coronary heart disease, stroke, and all-cause mortality: observational and revised Mendelian randomization analyses. Lancet Diabetes Endocrinol. 2024;12:e2–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S2213-8587(23)00287-5.
Stadler J, Garmo LG, Doyle D, Cheng C-I, Richardson G, Waheed Z, et al. Curcumin encapsulated in PAMAM dendrimers for the therapeutic treatment of ischemic stroke in rats. Front Cell Dev Biol. 2025;12:1467417. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fcell.2024.1467417.
Yang Q, Li R, Hong Y, Liu H, Jian C, Zhao S. Curcumin-loaded gelatin nanoparticles cross the blood-brain barrier to treat ischemic stroke by attenuating oxidative stress and neuroinflammation. Int J Nanomed. 2024;19:11633. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/IJN.S487628.
Alquisiras-Burgos I, Hernández-Cruz A, Peralta-Arrieta I, Aguilera P. Resveratrol prevents cell swelling through inhibition of SUR1 expression in brain micro endothelial cells subjected to OGD/recovery. Mol Neurobiol. 2024;61:2099–119. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12035-023-03686-0.
Jackson CW, Xu J, Escobar I, Saul I, Fagerli E, Dave KR, et al. Resveratrol preconditioning downregulates PARP1 protein to alleviate PARP1-mediated cell death following cerebral ischemia. Transl Stroke Res. 2024;15:165–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12975-022-01119-z.
Rajabi L, Ebrahimdoost M, Mohammadi SA, Soleimani Samarkhazan H, Khamisipour G, Aghaei M. Aqueous and ethanolic extracts of Moringa oleifera leaves induce selective cytotoxicity in Raji and Jurkat cell lines by activating the P21 pathway independent of P53. Mol Biol Rep. 2025;52:102. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11033-024-10200-9.
Sato T, Okumura M, Ishikawa T, Sakuta K, Takahashi J, Tanabe M, et al. Relationship between ω3 and ω6 polyunsaturated fatty acids and atrial fibrillation in acute ischemic stroke. Clin Nutr. 2024;43:1643–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.clnu.2024.05.021.
Huang F-Q, Wang H-F, Yang T, Yang D, Liu P, Alolga RN, et al. Ceramides increase mitochondrial permeabilization to trigger mtDNA-dependent inflammation in astrocytes during brain ischemia. Metabolism. 2025;166: 156161. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.metabol.2025.156161.
Zhao S, Zhuang H, Ji W, Cheng C, Liu Y. Identification of disulfidptosis-related genes in ischemic stroke by combining single-cell sequencing, machine learning algorithms, and in vitro experiments. Neuromolecular Med. 2024;26:39. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12017-024-08804-2.
Zhuang H, Lei W, Wu Q, Zhao S, Zhao Y, Zhang S, et al. Overexpressed CD73 attenuates GSDMD-mediated astrocyte pyroptosis induced by cerebral ischemia-reperfusion injury through the A2B/NF-κB pathway. Exp Neurol. 2025;386: 115152. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.expneurol.2025.115152.
Aghaei M, Khademi R, Bahreiny SS, Saki N. The need to establish and recognize the field of clinical laboratory science (CLS) as an essential field in advancing clinical goals. Health Sci Rep. 2024;7: e70008. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hsr2.70008.
Saki N, Haybar H, Aghaei M. Subject: motivation can be suppressed, but scientific ability cannot and should not be ignored. J Transl Med. 2023;21:520. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-023-04383-1.
Acknowledgements
None.
Funding
This work was supported by General Program of Wuxi Commission of Health (M202225), General Projects of Wuxi Medical Center, Nanjing Medical University (WMCG202405).
Author information
Authors and Affiliations
Contributions
All authors contributed to the conception and design of the study. The first draft of the manuscript was written by Wen Lei, and all authors commented on previous versions of the manuscript. Wen Lei and Hao Zhuang contributed to the creation of figures and the final draft.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Lei, W., Zhuang, H., Huang, W. et al. Neuroinflammation and energy metabolism: a dual perspective on ischemic stroke. J Transl Med 23, 413 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06440-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06440-3