
Mechanisms and interventions in aneurysmal subarachnoid hemorrhage: Unraveling the role of inflammatory responses and cell death in early brain injury (Review)
- Authors:
- Published online on: July 14, 2025 https://doi.org/10.3892/mmr.2025.13621
- Article Number: 256
-
Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
In the aging population, stroke has become a global concern (1). Subarachnoid hemorrhage (SAH) is the third most common subtype of stroke (2). The global mortality rate 25–35% (3). There are severe long-term neurological sequelae as cognitive ability may decline by >20% 1 year post-injury (4,5). Aneurysmal (a)SAH affects 6–9/100,000 individuals/year (6). According to the World Health Organization, the fatality rates 1, 2 and 7 days after the onset of SAH are 37, 60 and 75%, respectively (7). The cumulative mortality at 28 days in Australia is 26.7% (8) and 20–38% in Europe (9–11). The incidence rate of SAH in China is 2/100,000 individuals/year (12); additionally, it is estimated that the cumulative mortality at 28 days of aSAH in China is 16.9% (13). Considering the high mortality rate, it is important to pay attention to the complications following the SAH that contribute to mortality. Early case fatality are high across the world, which may increase by 50% in 2050 (1).
The pathophysiological mechanism of SAH is complex and multifactorial. Early brain injury (EBI) and delayed cerebral ischemia are key pathological processes following aSAH (14). Numerous clinical trials (14) have been conducted to improve outcomes for patients with aSAH, but there are challenges in aSAH prevention and the development of lower-risk treatments, these challenges persist in translating promising preclinical findings into effective low-risk treatments, partly due to issues with drug delivery across the blood-brain barrier and the multifactorial nature of early brain injury. The present aimed to summarize the pathogenesis and treatment for aSAH.
Pathology in the development of aSAH
SAH is primarily divided into two stages: The pathophysiological changes with the first 72 h after SAH are called ‘EBI’ and the second pathological process that occurs 72 h after EBI involves delayed cerebral ischemia (DCI) which typically occurs days after the initial hemorrhage (15,16). EBI is the key factor affecting the prognosis of SAH (2,17). EBI primarily describes the pathological changes 72 h after aneurysm rupture: Blood leaks into the subarachnoid space following aneurysm rupture, followed by the rapid increase in intracranial pressure, acute vasospasm, decreased cerebral blood flow, disruption of brain autoregulation and brain swelling (15–17). It is triggered by primary disturbances such as hemorrhage, increased intracranial pressure, vasospasm, and decreased cerebral blood flow. These factors activate downstream pathological mechanisms, including oxidative stress, apoptosis, autophagy, and immune inflammation (14). These mechanisms lead to the formation of pro-inflammatory signals and metabolic disturbances in the brain, which are focal points for early neuroprotective strategies (18,19). EBI manifests via various secondary pathological cascades triggered by the primary insults of hemorrhage including microvascular dysfunction, blood-brain barrier (BBB) disruption, cerebral edema, neuroinflammation, oxidative stress, and neuronal death, leading to acute neurological deficit (17,18).
For example, the breakdown of heme releases toxic substrates that catalyze reactive oxygen species (ROS) production and activate neuroinflammation, further contributing to neuronal death (12). Additionally, blood in the subarachnoid space activates toll-like receptor 4 (TLR4) via the TLR4/NF-κB signaling pathway, which mediates neuroinflammation (20).
The upregulation of inflammatory cytokines enhances the expression of matrix metalloproteinase-9 (21,22), an enzyme that degrades tight junction proteins such as zonula occludens-1 (23). This degradation compromises the integrity of tight junctions, accelerating BBB disruption (24). Compromised BBB further promotes neuroinflammation, creating a cycle that exacerbates BI. Understanding these pathological mechanisms is essential for developing targeted therapeutic strategies to mitigate the impact of EBI and improve patient outcomes following aSAH.
EBI-associated inflammation responses
TLR4 serves a key role in recognizing danger-associated molecular patterns (DAMPs), which are released following an aneurysm rupture. DAMPs markedly contribute to the increased permeability of the damaged BBB, leading to white blood cell infiltration, tissue edema and exacerbated BI (25). The activation of TLR4 initiates a series of signaling cascades that result in leukocyte activation and proliferation (26), further enhancing the expression of pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-8 and IL-12 (27).
Hemoglobin (Hb) degradation pathway, or the Hb-heme-iron axis, contributes to EBI. Hb degrades into heme, which breaks down into bilirubin and free iron (28). Free iron catalyzes ROS (29,30) production, and ROS-induced NLRP3 inflammasome activation triggers inflammatory responses (31). Excessive ROS production, coupled with decreased antioxidant defenses, results in cellular damage. Yue et al (30) revealed that in an intravascular perforation mouse model of SAH, ROS induces pyroptosis of neural stem cells (NSCs) by activating the NLRP3/gasdermin D (GSDMD) pathway. These findings indicate that Hb-induced NSCs may hinder nerve regeneration following SAH (32–35). Chang et al (36) used a mouse model of SAH to reveal that triiodothyronine (T3) treatment decreases mitochondrial ROS release, inhibiting neuronal apoptosis. These findings suggest that ROS is a potential therapeutic target for treating EBI following SAH (34,35). Studies (31,32) indicate that antioxidant treatment is an effective approach to mitigate EBI (Fig. 1).
Inflammation signaling pathways in EBI: The TLR4/NF-κB signaling pathway
TLR4 is the most well studied TLR and is widely expressed in the central nervous system (37–40). TLR4 serves out an important role in stroke-related inflammation (41). It is activated by the extravasated blood components in myeloid differentiation primary response-88/Toll/interleukin-1 receptor-domain-containing adapter-inducing interferon-β (MyD88/TRIF)-dependent pathway (42) after SAH. Transcription factors initiated by the activation of TLR4, such as NF-κB, mitogen-activated protein kinase and interferon regulatory factor that regulate the expression of proinflammatory cytokine genes cause brain damage after SAH (43,44). These factors collectively regulate the expression of pro-inflammatory cytokine genes, which contribute to brain damage post-SAH (45–47).
Moreover, NF-κB is a key driver of inflammation, which increases the expression of inflammatory markers and matrix metalloproteinases (48) and contributes to the pathogenesis of intracranial aneurysm (IA) (49). In addition, NF-κB activation can lead to endothelial dysfunction (50,51). Furthermore, TLR4-mediated inflammation fosters smooth muscle cell phenotype switching (52–54) and promotes the infiltration of inflammatory cells in arterial walls, potentially leading to the occurrence and progression of IAs, which may result in rupture (55). Therefore, targeting the TLR4/NF-κB pathway presents a promising therapeutic strategy to mitigate EBI and delay BI associated with neuroinflammation following SAH, ultimately improving patient prognosis (47,49).
Pyroptosis signaling pathways in EBI: The NLRP3/GSDMD signaling pathway
NLRP3-dependent signaling pathway serve a role in almost every mechanism of cell death, including pyroptosis (24,56–58). The NLRP3 inflammasome serves a role in the progression of injury following SAH (59,60). NLRs are a family of intracellular sensors of microbial motifs and ‘danger signals’ that serve as key components of innate immune responses and inflammation (61). Inflammasomes are multiprotein complexes that activate caspase-1. They process proinflammatory cytokines such as IL-1β. For a functional NLRP3 inflammasome, key components include a sensor protein (like an NLR), the adaptor protein ASC, and pro-caspase-1. ASC contains a CARD domain, which is important for recruiting pro-caspase-1 (62–64) and caspase-1 (65). GSDMD, a 53 kDa protein, is an inactive prerequisite protein in the cytoplasm, primarily composed of two domain groups, the C-terminal domain (CT-GSMD) and the T-terminal domain (NT-GSMD), which are connected by a flexible interdomain linker (66). After the aneurysm ruptures, heme groups of unstable extracellular Hb spontaneously oxidize to ferric methemoglobin and release superoxide in the reaction (67), which contributes to the production of ROS (68). The SAH model suggests that NLRP3 is activated by a common pathway of ROS (69,70). Activated NLRP3 inflammasomes recruit ASC and pro-caspase-1, further decompose and convert pro-caspase-1 into caspase-1 (71). Studies have demonstrated that caspase-1 can cleave the active linker (62,65,72), leading to the activation of the GSDMD protein and stimulating the secretion of the pro-inflammatory cytokines IL-1β and IL-18. The GSDMD protein also leads to the secretion of the pro-inflammatory cytokines IL-1β and IL-18 (71), as well as apoptotic and pyroptotic cell death (73). In addition, GSDMD is essential for both canonical and non-canonical inflammasome pathways (74) (Table I).
Treatments for aSAH
Current drugs targeting EBI
Salvianolic acid B (SalB) is a polyphenolic compound extracted from the Chinese herb Salvia miltiorrhiza (75). It possesses antioxidant and neuroprotective properties, demonstrating effectiveness in decreasing oxidative damage (76) and neuronal apoptosis post-SAH (77). SalB operates via the Nrf2 pathway (78), enhancing the expression of antioxidant proteins and improving neurological functions. Experimental studies (78–80) have demonstrated that the knockout of Nrf2 negates the protective effects of SalB, indicating its key role in the mechanism of action of SalB. SalB also activates Sirtuin 1 (SIRT1), a protein that modulates the Nrf2 signaling pathway (78). By enhancing SIRT1 activity, SalB indirectly promotes Nrf2 signaling, amplifying its neuroprotective effects. SIRT1 is known for its role in cellular stress resistance and metabolic regulation, making it a key component in neuroprotection (77–79). Furthermore, SalB has been shown (79) to exert anti-inflammatory effects in microglia, the immune cells of the brain. By regulating microglial activation and decreasing the release of pro-inflammatory cytokines (such as TNF-α and IL-1β), SalB helps to alleviate neuroinflammation, which is a contributor to EBI following SAH (80).
Therapeutic drugs targeting oxidative stress
Pterostilbene treatment reduces neuronal apoptosis by inhibiting NLRP3 inflammasome and Nox2-associated oxidative stress (81). This intervention not only mitigates neuronal death but also addresses the inflammatory response that often exacerbates BI after SAH.
Inhibitors of caspase family enzymes
Studies (82–86) have demonstrated that caspase family enzymes are involved in both neuronal and endothelial cell apoptosis in the primary stage following SAH. Activation of caspase post-SAH is complex, involving intrinsic and extrinsic pathways of apoptosis, endoplasmic reticulum stress-induced apoptosis and necroptosis (82). The intrinsic pathway is triggered by internal cellular stress signals, such as oxidative stress and mitochondrial dysfunction (83). It leads to the release of cytochrome c from the mitochondria, which activates initiator caspases such as caspase-9 (84). By contrast, the extrinsic pathway is activated by external signals, primarily through death receptors on the cell membrane, such as TNF receptors. Activation of these receptors leads to the formation of the death-inducing signaling complex, which activates initiator caspases such as caspase-8 (85). This activates downstream effector caspases (such as caspase-3), culminating in cell death (86). Caspase inhibitors, such as x-linked inhibitor of apoptosis protein (XIAP) (83), Z-VAD-FMK[carbobenzoxy-valyl-alanyl-aspartyl-(O-methyl)-fluoromethylketone] (84) and VX-765 (also known as Belnacasan), a potent caspase-1 and caspase-4 inhibitor (85), decrease the impacts of EBI by decreasing apoptosis and inflammation associated with caspase activation (86).
Luteolin (LUT) exerts biological functions beneficial to cerebrovascular diseases (87,88). In SAH rats (89), LUT markedly inhibits neuroinflammation via the Nrf2-dependent pathway. LUT decreases microglial activation (90), neutrophil infiltration and the release of pro-inflammatory cytokines, while also ameliorating oxidative damage and restoring the endogenous antioxidant system. Furthermore, LUT markedly ameliorates SAH-induced oxidative damage and restores the endogenous antioxidant system. Fluoxetine decreases neuroinflammation in EBI after SAH by regulating the TLR4/MyD88/NF-κB signaling pathway (91,92). This regulation helps to modulate inflammatory responses, providing a protective effect against neuronal damage.
The neuroprotective effect of necrostatin-1 (93) in SAG rats may stem from its ability to prevent BBB disruption by inhibiting the RIP3/MLKL signaling pathway (94). Administration of necrostatin-1 improves albumin leakage and tight junction protein degradation (95).
Electro-acupuncture (EA) therapy
EA has been revealed to regulate the balance between pro-apoptotic and anti-apoptotic proteins (96–98), decreasing levels of cleaved caspase-3 and inflammatory cytokines such as TNF-α, IL-1β and IL-6. Additionally, EA decreases the M1 polarization of activated microglia, suggesting an anti-inflammatory effect that improves outcomes in EBI. In summary, EA is a potential therapy for the treatment of SAH (Table II).
The aforementioned therapies illustrate a multi-faceted approach to mitigating EBI following SAH. By targeting oxidative stress, inflammatory pathways and apoptotic processes, these drugs aim to enhance neuronal survival and improve clinical outcomes. Understanding the mechanisms of these treatments may aid in developing more effective therapeutic strategies for patients with SAH.
Conclusion
aSAH is a severe cerebrovascular event characterized by complex and multifaceted pathophysiological mechanisms, leading to high rates of morbidity and mortality. The present review summarized understanding of pathophysiology of aSAH and therapeutic strategies targeting the molecular pathways involved. Further research is required to translate these findings into clinical practice and improve outcomes for patients affected by aSAH.
Acknowledgements
Not applicable.
Funding
The present study was supported by Cerebrovascular Disease Interventional Therapy Remote Network Consultation Project (grant no. RKX202101007).
Availability of data and materials
Not applicable.
Authors' contributions
RL conceived the study and wrote and edited the manuscript. SG conducted the literature review and wrote and edited the manuscript. JW and MH performed the literature review and wrote the manuscript. MF substantial intellectual contributions to the analysis and interpretation of the existing literature, critically shaping the review's key arguments and conclusions, conceptualized and designed the illustrative figures and comprehensive tables and provided language polishing assistance. JXW and NZ wrote and revised the manuscript. XZ revised the manuscript. JL contributed significantly to the intellectual content of the review and participated in the drafting and finalization of the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Feigin VL and Owolabi MO; World Stroke Organization-Lancet Neurology Commission Stroke Collaboration Group, : Pragmatic solutions to reduce the global burden of stroke: A world stroke organization-lancet neurology commission. Lancet Neurol. 22:1160–1206. 2023. View Article : Google Scholar : PubMed/NCBI | |
Claassen J and Park S: Spontaneous subarachnoid haemorrhage. Lancet. 400:846–862. 2022. View Article : Google Scholar : PubMed/NCBI | |
Etminan N, Chang HS, Hackenberg K, de Rooij NK, Vergouwen MDI, Rinkel GJE and Algra A: Worldwide Incidence of Aneurysmal Subarachnoid Hemorrhage According to Region, Time period, blood pressure, and smoking prevalence in the population: A systematic review and meta-analysis. JAMA Neurol. 76:588–597. 2019. View Article : Google Scholar : PubMed/NCBI | |
Springer MV, Schmidt JM, Wartenberg KE, Frontera JA, Badjatia N and Mayer SA: Predictors of global cognitive impairment 1 year after subarachnoid hemorrhage. Neurosurgery. 65:1043–1051. 2009. View Article : Google Scholar : PubMed/NCBI | |
Chai CZ, Ho UC and Kuo LT: Systemic inflammation after aneurysmal subarachnoid hemorrhage. Int J Mol Sci. 24:109432023. View Article : Google Scholar : PubMed/NCBI | |
Neifert SN, Chapman EK, Martini ML, Shuman WH, Schupper AJ, Oermann EK, Mocco J and Macdonald RL: Aneurysmal subarachnoid hemorrhage: The last decade. Transl Stroke Res. 12:428–446. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kuo LT and Huang AP: The pathogenesis of hydrocephalus following aneurysmal subarachnoid hemorrhage. Int J Mol Sci. 22:50502021. View Article : Google Scholar : PubMed/NCBI | |
Huang H and Lai LT: Incidence and case-fatality of aneurysmal subarachnoid hemorrhage in Australia, 2008–2018. World Neurosurg. 144:e438–e446. 2020. View Article : Google Scholar : PubMed/NCBI | |
Fischer T, Johnsen SP, Pedersen L, Gaist D, Sørensen HT and Rothman KJ: Seasonal variation in hospitalization and case fatality of subarachnoid hemorrhage-a nationwide danish study on 9,367 patients. Neuroepidemiology. 24:32–37. 2005. View Article : Google Scholar : PubMed/NCBI | |
Biotti D, Jacquin A, Boutarbouch M, Bousquet O, Durier J, Ben Salem D, Ricolfi F, Beaurain J, Osseby GV, Moreau T, et al: Trends in case-fatality rates in hospitalized nontraumatic subarachnoid hemorrhage: Results of a population-based study in Dijon, France, From 1985 to 2006. Neurosurgery. 66:1039–1043. 2010. View Article : Google Scholar : PubMed/NCBI | |
Vadikolias K, Tsivgoulis G, Heliopoulos I, Papaioakim M, Aggelopoulou C, Serdari A, Birbilis T and Piperidou C: Incidence and case fatality of subarachnoid haemorrhage in Northern Greece: The evros registry of subarachnoid haemorrhage. Int J Stroke. 4:322–327. 2009. View Article : Google Scholar : PubMed/NCBI | |
Pan P, Xu L, Zhang H, Liu Y, Lu X, Chen G, Tang H and Wu J: A review of hematoma components clearance mechanism after subarachnoid hemorrhage. Front Neurosci. 14:6852020. View Article : Google Scholar : PubMed/NCBI | |
Bian LH, Liu YF, Nichols LT, Wang CX, Wang YL, Liu GF, Wang WJ and Zhao XQ: Epidemiology of subarachnoid hemorrhage, patterns of management, and outcomes in China: A hospital-based multicenter prospective study. CNS Neurosci Ther. 18:895–902. 2012. View Article : Google Scholar : PubMed/NCBI | |
Macdonald RL and Schweizer TA: Spontaneous subarachnoid haemorrhage. Lancet. 389:655–666. 2017. View Article : Google Scholar : PubMed/NCBI | |
van Lieshout JH, Dibué-Adjei M, Cornelius JF, Slotty PJ, Schneider T, Restin T, Boogaarts HD, Steiger HJ, Petridis AK and Kamp MA: An introduction to the pathophysiology of aneurysmal subarachnoid hemorrhage. Neurosurg Rev. 41:917–930. 2018. View Article : Google Scholar : PubMed/NCBI | |
Foreman B: The pathophysiology of delayed cerebral ischemia. J Clin Neurophysiol. 33:174–182. 2016. View Article : Google Scholar : PubMed/NCBI | |
Lauzier DC, Jayaraman K, Yuan JY, Diwan D, Vellimana AK, Osbun JW, Chatterjee AR, Athiraman U, Dhar R and Zipfel GJ: Early brain injury after subarachnoid hemorrhage: Incidence and mechanisms. Stroke. 54:1426–1440. 2023. View Article : Google Scholar : PubMed/NCBI | |
Weiland J, Beez A, Westermaier T, Kunze E, Sirén AL and Lilla N: Neuroprotective strategies in aneurysmal subarachnoid hemorrhage (aSAH). Int J Mol Sci. 22:54422021. View Article : Google Scholar : PubMed/NCBI | |
Guo Y, Liu X, Liu D, Li K, Wang C, Liu Y, He B and Shi P: Inhibition of BECN1 suppresses lipid peroxidation by increasing system Xc− activity in early brain injury after subarachnoid hemorrhage. J Mol Neurosci. 67:622–631. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chang S, Li X, Zheng Y, Shi H, Zhang D, Jing B, Chen Z, Qian G and Zhao G: Kaempferol exerts a neuroprotective effect to reduce neuropathic pain through TLR4/NF-ĸB signaling pathway. Phytother Res. 36:1678–1691. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Li Q, Tang J, Feng H and Zhang JH: The evolving roles of pericyte in early brain injury after subarachnoid hemorrhage. Brain Res. 1623:110–122. 2015. View Article : Google Scholar : PubMed/NCBI | |
Shao AW, Wu HJ, Chen S, Ammar AB, Zhang JM and Hong Y: Resveratrol attenuates early brain injury after subarachnoid hemorrhage through inhibition of NF-κB-dependent inflammatory/MMP-9 pathway. CNS Neurosci Ther. 20:182–185. 2014. View Article : Google Scholar : PubMed/NCBI | |
Fang X and Xu RS: Protective effect of simvastatin on impaired intestine tight junction protein ZO-1 in a mouse model of Parkinson's disease. J Huazhong Univ Sci Technolog Med Sci. 35:880–884. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xu P, Hong Y, Xie Y, Yuan K, Li J, Sun R, Zhang X, Shi X, Li R, Wu J, et al: TREM-1 exacerbates neuroinflammatory injury via NLRP3 inflammasome-mediated pyroptosis in experimental subarachnoid hemorrhage. Transl Stroke Res. 12:643–659. 2021. View Article : Google Scholar : PubMed/NCBI | |
Stanzione R, Forte M, Cotugno M, Bianchi F, Marchitti S and 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. 42:545–556. 2022. View Article : Google Scholar : PubMed/NCBI | |
Pradilla G, Chaichana KL, Hoang S, Huang J and Tamargo RJ: Inflammation and cerebral vasospasm after subarachnoid hemorrhage. Neurosurg Clin N Am. 21:365–379. 2010. View Article : Google Scholar : PubMed/NCBI | |
Kwon MS, Woo SK, Kurland DB, Yoon SH, Palmer AF, Banerjee U, Iqbal S, Ivanova S, Gerzanich V and Simard JM: Methemoglobin is an endogenous toll-like receptor 4 ligand-relevance to subarachnoid hemorrhage. Int J Mol Sci. 16:5028–5046. 2015. View Article : Google Scholar : PubMed/NCBI | |
Babadjouni RM, Radwanski RE, Walcott BP, Patel A, Durazo R, Hodis DM, Emanuel BA and Mack WJ: Neuroprotective strategies following intraparenchymal hemorrhage. J Neurointerv Surg. 9:1202–1207. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhu F, Zi L, Yang P, Wei Y, Zhong R, Wang Y, You C, Li Y, Tian M and Gu Z: Efficient iron and ROS nanoscavengers for brain protection after intracerebral hemorrhage. ACS Appl Mater Interfaces. 13:9729–9738. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yue T, Li X, Chen X, Zhu T, Li W, Wang B and Hang C: Hemoglobin derived from subarachnoid hemorrhage-induced pyroptosis of neural stem cells via ROS/NLRP3/GSDMD pathway. Oxid Med Cell Longev. 2023:43833322023. View Article : Google Scholar : PubMed/NCBI | |
Thilak S, Brown P, Whitehouse T, Gautam N, Lawrence E, Ahmed Z and Veenith T: Diagnosis and management of subarachnoid haemorrhage. Nat Commun. 15:18502024. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Zhang J, Lu K, Zhang Y, Xu X, Deng J, Zhang X, Zhang H, Zhao Y and Wang X: ChemR23 signaling ameliorates brain injury via inhibiting NLRP3 inflammasome-mediated neuronal pyroptosis in ischemic stroke. J Transl Med. 22:232024. View Article : Google Scholar : PubMed/NCBI | |
Fumoto T, Naraoka M, Katagai T, Li Y, Shimamura N and Ohkuma H: The role of oxidative stress in microvascular disturbances after experimental subarachnoid hemorrhage. Transl Stroke Res. 10:684–694. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ghonim HT, Shah SS, Thompson JW, Ambekar S, Peterson EC and Elhammady MS: Stem cells as a potential adjunctive therapy in aneurysmal subarachnoid hemorrhage. J Vasc Interv Neurol. 8:30–37. 2016.PubMed/NCBI | |
Lee WD, Wang KC, Tsai YF, Chou PC, Tsai LK and Chien CL: Subarachnoid hemorrhage promotes proliferation, differentiation, and migration of neural stem cells via BDNF upregulation. PLoS One. 11:e01654602016. View Article : Google Scholar : PubMed/NCBI | |
Chang H, Lin C, Li Z, Shen Y, Zhang G, Mao L, Ma C, Liu N and Lu H: T3 alleviates neuroinflammation and reduces early brain injury after subarachnoid haemorrhage by promoting mitophagy via PINK 1-parkin pathway. Exp Neurol. 357:1141752022. View Article : Google Scholar : PubMed/NCBI | |
Xia DY, Yuan JL, Jiang XC, Qi M, Lai NS, Wu LY and Zhang XS: SIRT1 promotes M2 microglia polarization via reducing ROS-mediated NLRP3 inflammasome signaling after subarachnoid hemorrhage. Front Immunol. 12:7707442021. View Article : Google Scholar : PubMed/NCBI | |
Mitsui K, Ikedo T, Kamio Y, Furukawa H, Lawton MT and Hashimoto T: TLR4 (toll-like receptor 4) mediates the development of intracranial aneurysm rupture. Hypertension. 75:468–476. 2020. View Article : Google Scholar : PubMed/NCBI | |
Suzuki H, Fujimoto M, Kawakita F, Liu L, Nakano F, Nishikawa H, Okada T, Imanaka-Yoshida K, Yoshida T and Shiba M: Toll-like receptor 4 and tenascin-C signaling in cerebral vasospasm and brain injuries after subarachnoid hemorrhage. Acta Neurochir Suppl. 127:91–96. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ma C, Zhou W, Yan Z, Qu M and Bu X: Toll-like receptor 4 (TLR4) is associated with cerebral vasospasm and delayed cerebral ischemia in aneurysmal subarachnoid hemorrhage. Neurol Med Chir (Tokyo). 55:878–884. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lee SJ and Lee S: Toll-like receptors and inflammation in the CNS. Curr Drug Targets Inflamm Allergy. 1:181–191. 2002. View Article : Google Scholar : PubMed/NCBI | |
Karimy JK, Reeves BC and Kahle KT: Targeting TLR4-dependent inflammation in post-hemorrhagic brain injury. Expert Opin Ther Targets. 24:525–533. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Luo Q, Zhao YH and Chen X: Toll-like receptor-4 pathway as a possible molecular mechanism for brain injuries after subarachnoid hemorrhage. Int J Neurosci. 130:953–964. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yamamoto M, Okamoto T, Takeda K, Sato S, Sanjo H, Uematsu S, Saitoh T, Yamamoto N, Sakurai H, Ishii KJ, et al: Key function for the Ubc13 E2 ubiquitin-conjugating enzyme in immune receptor signaling. Nat Immunol. 7:962–970. 2006. View Article : Google Scholar : PubMed/NCBI | |
Wang YC, Zhou Y, Fang H, Lin S, Wang PF, Xiong RP, Chen J, Xiong XY, Lv FL, Liang QL and Yang QW: Toll-like receptor 2/4 heterodimer mediates inflammatory injury in intracerebral hemorrhage. Ann Neurol. 75:876–889. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Geng G, Zhu T, Chen W, Li X, Gu J and Jiang E: Progress in research on TLR4-mediated inflammatory response mechanisms in brain injury after subarachnoid hemorrhage. Cells. 11:37812022. View Article : Google Scholar : PubMed/NCBI | |
Okada T, Kawakita F, Nishikawa H, Nakano F, Liu L and Suzuki H: Selective toll-like receptor 4 antagonists prevent acute blood-brain barrier disruption after subarachnoid hemorrhage in mice. Mol Neurobiol. 56:976–985. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S and Zhang JH: Immunological response in early brain injury after SAH. Acta Neurochir Suppl. 110:57–61. 2011.PubMed/NCBI | |
Khan D, Cornelius JF and Muhammad S: The role of NF-κB in intracranial aneurysm pathogenesis: A systematic review. Int J Mol Sci. 24:142182023. View Article : Google Scholar : PubMed/NCBI | |
Hu J, Chen R, An J, Wang Y, Liang M and Huang K: Dauricine attenuates vascular endothelial inflammation through inhibiting NF-κB pathway. Front Pharmacol. 12:7589622021. View Article : Google Scholar : PubMed/NCBI | |
Tang P, Wang Y, Yang X, Wu Z, Chen W, Ye Y, Jiang Y, Lin L and Lin B and Lin B: Protective role of endothelial SIRT1 in deep vein thrombosis and hypoxia-induced endothelial dysfunction mediated by NF-κB deacetylation. Inflammation. 46:1887–1900. 2023. View Article : Google Scholar : PubMed/NCBI | |
Kirsebom FCM, Kausar F, Nuriev R, Makris S and Johansson C: Neutrophil recruitment and activation are differentially dependent on MyD88/TRIF and MAVS signaling during RSV infection. Mucosal Immunol. 12:1244–1255. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ahmed H, Khan MA, Kahlert UD, Niemelä M, Hänggi D, Chaudhry SR and Muhammad S: Role of adaptor protein myeloid differentiation 88 (MyD88) in post-subarachnoid hemorrhage inflammation: A systematic review. Int J Mol Sci. 22:41852021. View Article : Google Scholar : PubMed/NCBI | |
Klepinowski T, Skonieczna-Żydecka K, Pala B, Stachowska E and Sagan L: Gut microbiome in intracranial aneurysm growth, subarachnoid hemorrhage, and cerebral vasospasm: A systematic review with a narrative synthesis. Front Neurosci. 17:12471512023. View Article : Google Scholar : PubMed/NCBI | |
Liu GJ, Zhang QR, Gao X, Wang H, Tao T, Gao YY, Zhou Y, Chen XX, Li W and Hang CH: MiR-146a ameliorates hemoglobin-induced microglial inflammatory response via TLR4/IRAK1/TRAF6 associated pathways. Front Neurosci. 14:3112020. View Article : Google Scholar : PubMed/NCBI | |
Wang YH, Gao X, Tang YR, Yu Y, Sun MJ, Chen FQ and Li Y: The role of NF-κB/NLRP3 inflammasome signaling pathway in attenuating pyroptosis by melatonin upon spinal nerve ligation models. Neurochem Res. 47:335–346. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Zhang Y, Li R, Zhu L, Fu B and Yan T: Salidroside ameliorates Parkinson's disease by inhibiting NLRP3-dependent pyroptosis. Aging (Albany NY). 12:9405–9426. 2020. View Article : Google Scholar : PubMed/NCBI | |
Man SM, Karki R and Kanneganti TD: Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 277:61–75. 2017. View Article : Google Scholar : PubMed/NCBI | |
Shao BZ, Cao Q and Liu C: Targeting NLRP3 inflammasome in the treatment of CNS diseases. Front Mol Neurosci. 11:3202018. View Article : Google Scholar : PubMed/NCBI | |
Zhang K, Qin Z, Chen J, Guo G, Jiang X, Wang F, Zhuang J and Zhang Z: TRPV1 modulated NLRP3 inflammasome activation via calcium in experimental subarachnoid hemorrhage. Aging (Albany NY). 16:1096–1110. 2024. View Article : Google Scholar : PubMed/NCBI | |
Almeida-da-Silva CLC, Savio LEB, Coutinho-Silva R and Ojcius DM: The role of NOD-like receptors in innate immunity. Front Immunol. 14:11225862023. View Article : Google Scholar : PubMed/NCBI | |
Nagar A, Bharadwaj R, Shaikh MOF and Roy A: What are NLRP3-ASC specks? An experimental progress of 22 years of inflammasome research. Front Immunol. 14:11888642023. View Article : Google Scholar : PubMed/NCBI | |
Martín-Sánchez F, Compan V, Peñín-Franch A, Tapia-Abellán A, Gómez AI, Baños-Gregori MC, Schmidt FI and Pelegrin P: ASC oligomer favors caspase-1CARD domain recruitment after intracellular potassium efflux. J Cell Biol. 222:e2020030532023. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Zhai H, Alemayehu H, Boulanger J, Hopkins LJ, Borgeaud AC, Heroven C, Howe JD, Leigh KE, Bryant CE and Modis Y: Cryo-electron tomography of NLRP3-activated ASC complexes reveals organelle co-localization. Nat Commun. 14:72462023. View Article : Google Scholar : PubMed/NCBI | |
Pétrilli V, Dostert C, Muruve DA and Tschopp J: The inflammasome: A danger sensing complex triggering innate immunity. Curr Opin Immunol. 19:615–622. 2007. View Article : Google Scholar : PubMed/NCBI | |
Devant P and Kagan JC: Molecular mechanisms of gasdermin D pore-forming activity. Nat Immunol. 24:1064–1075. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Chen S and Zhang JM: The updated role of oxidative stress in subarachnoid hemorrhage. Curr Drug Deliv. 14:832–842. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Zhang C, Yan T, Yang L, Wang Y, Shi Z, Li M and Chen Q: Atorvastatin ameliorates early brain injury after subarachnoid hemorrhage via inhibition of pyroptosis and neuroinflammation. J Cell Physiol. 236:6920–6931. 2021. View Article : Google Scholar : PubMed/NCBI | |
Akar A, Öztopuz RÖ, Büyük B, Ovali MA, Aykora D and Malçok ÜA: Neuroprotective effects of piceatannol on olfactory bulb injury after subarachnoid hemorrhage. Mol Neurobiol. 60:3695–3706. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhou R, Yazdi AS, Menu P and Tschopp J: A role for mitochondria in NLRP3 inflammasome activation. Nature. 469:221–225. 2011. View Article : Google Scholar : PubMed/NCBI | |
Elliott EI and Sutterwala FS: Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol Rev. 265:35–52. 2015. View Article : Google Scholar : PubMed/NCBI | |
Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F and Shao F: Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 526:660–665. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zheng D, Liwinski T and Elinav E: Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 6:362020. View Article : Google Scholar : PubMed/NCBI | |
He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ and Han J: Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 25:1285–1298. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ji XY, Tan BK and Zhu YZ: Salvia miltiorrhiza and ischemic diseases. Acta Pharmacol Sin. 21:1089–1094. 2000.PubMed/NCBI | |
Zhang H and Chen X: Effects of salvianolic acid B on osteogenic differentiation and oxidative stress of periodontal ligament stem cells. Genomics Appl Biol. 39:3232–3240. 2020. | |
Guarente L: Sirtuins as potential targets for metabolic syndrome. Nature. 444:868–874. 2006. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Wu Q, Lu Y, Wan J, Dai H, Zhou X, Lv S, Chen X, Zhang X, Hang C and Wang J: Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways. Free Radic Biol Med. 124:504–516. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Xie X, Tang M, Zhang J, Zhang B, Zhao Q, Han Y, Yan W, Peng C and You Z: Salvianolic acid B promotes microglial M2-polarization and rescues neurogenesis in stress-exposed mice. Brain Behav Immun. 66:111–124. 2017. View Article : Google Scholar : PubMed/NCBI | |
Shu T, Pang M, Rong L, Liu C, Wang J, Zhou W, Wang X and Liu B: Protective effects and mechanisms of salvianolic acid B against H2O2-induced injury in induced pluripotent stem cell-derived neural stem cells. Neurochem Res. 40:1133–1143. 2015. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Zhao L, Yue L, Wang B, Li X, Guo H, Ma Y, Yao C, Gao L, Deng J, et al: Pterostilbene attenuates early brain injury following subarachnoid hemorrhage via inhibition of the NLRP3 inflammasome and Nox2-related oxidative stress. Mol Neurobiol. 54:5928–5940. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Xu P, Liu Y, Wang Z, Lenahan C, Fang Y, Lu J, Zheng J, Wang K, Wang W, et al: New insights of early brain injury after subarachnoid hemorrhage: A focus on the caspase family. Curr Neuropharmacol. 21:392–408. 2023. View Article : Google Scholar : PubMed/NCBI | |
Duckett CS, Nava VE, Gedrich RW, Clem RJ, Van Dongen JL, Gilfillan MC, Shiels H, Hardwick JM and Thompson CB: A conserved family of cellular genes related to the baculovirus iap gene and encoding apoptosis inhibitors. EMBO J. 15:2685–2694. 1996. View Article : Google Scholar : PubMed/NCBI | |
Iseda K, Ono S, Onoda K, Satoh M, Manabe H, Nishiguchi M, Takahashi K, Tokunaga K, Sugiu K and Date I: Antivasospastic and antiinflammatory effects of caspase inhibitor in experimental subarachnoid hemorrhage. J Neurosurg. 107:128–135. 2007. View Article : Google Scholar : PubMed/NCBI | |
Fang Y, Wang X, Lu J, Shi H, Huang L, Shao A, Zhang A, Liu Y, Ren R, Lenahan C, et al: Inhibition of caspase-1-mediated inflammasome activation reduced blood coagulation in cerebrospinal fluid after subarachnoid haemorrhage. EbioMedicine. 76:1038432022. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Liu Y, Zhou C, Wu Y, Sun J, Gao X and Huang Y: Biological effects and mechanisms of caspases in early brain injury after subarachnoid hemorrhage. Oxid Med Cell Longev. 2022:33456372022. View Article : Google Scholar : PubMed/NCBI | |
Delgado A, Cholevas C and Theoharides TC: Neuroinflammation in Alzheimer's disease and beneficial action of luteolin. Biofactors. 47:207–217. 2021. View Article : Google Scholar : PubMed/NCBI | |
Theoharides TC, Conti P and Economu M: Brain inflammation, neuropsychiatric disorders, and immunoendocrine effects of luteolin. J Clin Psychopharmacol. 34:187–189. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhang ZH, Liu JQ, Hu CD, Zhao XT, Qin FY, Zhuang Z and Zhang XS: Luteolin confers cerebroprotection after subarachnoid hemorrhage by suppression of NLPR3 inflammasome activation through Nrf2-dependent pathway. Oxid Med Cell Longev. 2021:58381012021. View Article : Google Scholar : PubMed/NCBI | |
Zhou W, Hu M, Hu J, Du Z, Su Q and Xiang Z: Luteolin suppresses microglia neuroinflammatory responses and relieves inflammation-induced cognitive impairments. Neurotox Res. 39:1800–1811. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu FY, Cai J, Wang C, Ruan W, Guan GP, Pan HZ, Li JR, Qian C, Chen JS, Wang L and Chen G: Fluoxetine attenuates neuroinflammation in early brain injury after subarachnoid hemorrhage: A possible role for the regulation of TLR4/MyD88/NF-κB signaling pathway. J Neuroinflammation. 15:3472018. View Article : Google Scholar : PubMed/NCBI | |
Hu HM, Li B, Wang XD, Guo YS, Hui H, Zhang HP, Wang B, Huang DG and Hao DJ: Fluoxetine is neuroprotective in early brain injury via its anti-inflammatory and anti-apoptotic effects in a rat experimental subarachnoid hemorrhage model. Neurosci Bull. 34:951–962. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhou K, Shi L, Wang Z, Zhou J, Manaenko A, Reis C, Chen S and Zhang J: RIP1-RIP3-DRP1 pathway regulates NLRP3 inflammasome activation following subarachnoid hemorrhage. Exp Neurol. 295:116–124. 2017. View Article : Google Scholar : PubMed/NCBI | |
Endo Y, Winarski KL, Sajib MS, Ju A and Wu WJ: Atezolizumab induces necroptosis and contributes to hepatotoxicity of human hepatocytes. Int J Mol Sci. 24:116942023. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Jin H, Xu H, Peng Y, Jie L, Xu D, Chen L, Li T, Fan L, He P, et al: The neuroprotective effects of necrostatin-1 on subarachnoid hemorrhage in rats are possibly mediated by preventing blood-brain barrier disruption and RIP3-mediated necroptosis. Cell Transplant. 28:1358–1372. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang C, Liu J, Wang J, Yin A, Jiang Z, Ye S, Liu X, Zhang X, Wang F and Xiong L: Activation of astroglial CB1R mediates cerebral ischemic tolerance induced by electroacupuncture. J Cereb Blood Flow Metab. 41:2295–2310. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ulloa L: Electroacupuncture activates neurons to switch off inflammation. Nature. 598:573–574. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Yang X, Cao Y, Li X, Xu R, Yan J, Guo Z, Sun S, Sun X and Wu Y: Electroacupuncture alleviates early brain injury via modulating microglia polarization and suppressing neuroinflammation in a rat model of subarachnoid hemorrhage. Heliyon. 9:e144752023. View Article : Google Scholar : PubMed/NCBI |