Chengliang Luo's research while affiliated with Soochow University (PRC) and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (47)


The Interplay between Ferroptosis and Neuroinflammation in Central Neurological Disorders
  • Literature Review
  • Full-text available

March 2024

·

24 Reads

·

2 Citations

Antioxidants

Yejia Xu

·

Bowen Jia

·

·

[...]

·

Chengliang Luo

Central neurological disorders are significant contributors to morbidity, mortality, and long-term disability globally in modern society. These encompass neurodegenerative diseases, ischemic brain diseases, traumatic brain injury, epilepsy, depression, and more. The involved pathogenesis is notably intricate and diverse. Ferroptosis and neuroinflammation play pivotal roles in elucidating the causes of cognitive impairment stemming from these diseases. Given the concurrent occurrence of ferroptosis and neuroinflammation due to metabolic shifts such as iron and ROS, as well as their critical roles in central nervous disorders, the investigation into the co-regulatory mechanism of ferroptosis and neuroinflammation has emerged as a prominent area of research. This paper delves into the mechanisms of ferroptosis and neuroinflammation in central nervous disorders, along with their interrelationship. It specifically emphasizes the core molecules within the shared pathways governing ferroptosis and neuroinflammation, including SIRT1, Nrf2, NF-κB, Cox-2, iNOS/NO·, and how different immune cells and structures contribute to cognitive dysfunction through these mechanisms. Researchers’ findings suggest that ferroptosis and neuroinflammation mutually promote each other and may represent key factors in the progression of central neurological disorders. A deeper comprehension of the common pathway between cellular ferroptosis and neuroinflammation holds promise for improving symptoms and prognosis related to central neurological disorders.

Download
Share

Origin, gene, and the protein structure of IL‐38. (A) The milestone events associated with IL‐38. (B) The IL‐38 gene is located on human chromosome 2q13‐14.1, between the two antagonist genes IL‐1Ra and IL‐36Ra. The IL‐38 gene is mainly composed of 5 exons of 7.8 kb DNA genome, encoding a precursor protein of 152 amino acids (AA) with a molecular weight of 17 kDa. The existence forms of IL‐38 protein can be divided into two categories: IL‐38 full‐length form and IL‐38 mature form. There are many types of mature forms of IL‐38, such as IL‐38 (aa2‐152, aa3‐152, aa5‐152, aa7‐152, and aa20‐152). Due to the lack of a signal peptide and caspase‐1 cleavage site, IL‐38, like most members of IL‐1F, requires N‐terminal cleavage to be biologically active. These predicted cleavage enzymes (e.g., Calpain, MMP2/9, Cathepsin G, Granzyme B) are indicated by arrows with the corresponding cleavage sites. (C) The 3D crystal structure of the IL‐38‐IL‐36R complex is shown. The two critical binding sites have been marked by dotted boxes (black).
The putative effects and mechanisms of IL‐38 in NMOD and AD. (A) In NMOD, abnormally elevated AQP4‐specific antibody IgG1 in the serum crosses the BBB disrupted by pro‐inflammatory cytokines and enters the brain parenchyma. Subsequently, AQP4‐specific antibodies induce AQP4 internalization by binding to AQP4 on astrocyte terminal foot processes, thereby reducing astrocyte surface AQP4 expression and activating astrocytes. Ultimately, activation of the complement cascade forms the MAC, leading to astrocyte lysis and neuronal necrosis. Notably, IL‐38 can block IL‐36/IL‐36R signaling by competitively antagonizing IL‐36 receptor (IL‐36R) and ILRAPL1 on Th17 cell membranes, thereby inhibiting pro‐inflammatory cytokines (i.e., IL‐17 and IL‐6) expression. Downregulation of IL‐17 and IL‐6 expression levels reduced plasma cell differentiation and antibody production, ultimately alleviating BBB disruption in NMOD. (B) In AD, IL‐38 inhibits Tau phosphorylation and neuronal plaque formation by competing with IL‐1β and TNF‐α for binding to IL‐1R1 and TNFR1, thereby reducing neuronal degeneration and necrosis.
The mechanisms of IL‐38 underpinning in ASD, IS, TBI, and SCI. IL‐38 regulates immune and inflammatory responses after ASD, IS, TBI, and SCI by competitively binding to IL36R with IL‐36. Briefly, IL‐1R1 and IL‐36R are activated by the agonists IL‐1 and IL‐36 (α, γ and β), respectively. The heterodimeric receptor complexes composed of IL‐1R1 or IL‐36R and IL‐1RAcP contribute to myeloid differentiation primary response 88 (MyD88) recruitment through the intracellular toll‐interleukin 1 receptor (TIR) domain, and the downstream signaling pathways that are activated include NF‐κB and MAPK pathways by extracellular regulated protein kinases (ERK), p38 or c‐Jun N‐terminal kinase (JNK), which stimulate the activator protein‐1 (AP‐1). NF‐κB and AP‐1 then bind DNA and stimulate the production of proinflammatory cytokines and chemokines. Conversely, IL‐38 acts as an antagonist to IL‐1R1 and IL‐36R, inhibiting IL‐1RAcP recruitment and agonist binding, similar to what occurs for IL‐1Ra and IL‐36Ra. In addition, IL‐38 selectively recruits MEK or upregulates channel protein‐dependent SIRT1 by binding to IL1RAPL1, thereby reducing NF‐κB, AP‐1, and MAPK, and ultimately regulating the expression of multiple cytokines involved in the pathological process of ASD, IS, TBI, and SCI.
Emerging functions and therapeutic targets of IL‐38 in central nervous system diseases
CNS Neuroscience & Therapeutics

CNS Neuroscience & Therapeutics

Interleukin (IL)‐38 is a newly discovered cytokine of the IL‐1 family, which binds various receptors (i.e., IL‐36R, IL‐1 receptor accessory protein‐like 1, and IL‐1R1) in the central nervous system (CNS). The hallmark physiological function of IL‐38 is competitive binding to IL‐36R, as does the IL‐36R antagonist. Emerging research has shown that IL‐38 is abnormally expressed in the serum and brain tissue of patients with ischemic stroke (IS) and autism spectrum disorder (ASD), suggesting that IL‐38 may play an important role in neurological diseases. Important advances include that IL‐38 alleviates neuromyelitis optica disorder (NMOD) by inhibiting Th17 expression, improves IS by protecting against atherosclerosis via regulating immune cells and inflammation, and reduces IL‐1β and CXCL8 release through inhibiting human microglial activity post‐ASD. In contrast, IL‐38 mRNA is markedly increased and is mainly expressed in phagocytes in spinal cord injury (SCI). IL‐38 ablation attenuated SCI by reducing immune cell infiltration. However, the effect and underlying mechanism of IL‐38 in CNS diseases remain inadequately characterized. In this review, we summarize the biological characteristics, pathophysiological role, and potential mechanisms of IL‐38 in CNS diseases (e.g., NMOD, Alzheimer's disease, ASD, IS, TBI, and SCI), aiming to explore the therapeutic potential of IL‐38 in the prevention and treatment of CNS diseases.


The MT1 receptor as the target of ramelteon neuroprotection in ischemic stroke

November 2023

·

29 Reads

·

2 Citations

Journal of Pineal Research

Stroke is the leading cause of death and disability worldwide. Novel and effective therapies for ischemic stroke are urgently needed. Here, we report that melatonin receptor 1A (MT1) agonist ramelteon is a neuroprotective drug candidate as demonstrated by comprehensive experimental models of ischemic stroke, including a middle cerebral artery occlusion (MCAO) mouse model of cerebral ischemia in vivo, organotypic hippocampal slice cultures ex vivo, and cultured neurons in vitro; the neuroprotective effects of ramelteon are diminished in MT1‐knockout (KO) mice and MT1‐KO cultured neurons. For the first time, we report that the MT1 receptor is significantly depleted in the brain of MCAO mice, and ramelteon treatment significantly recovers the brain MT1 losses in MCAO mice, which is further explained by the Connectivity Map L1000 bioinformatic analysis that shows gene‐expression signatures of MCAO mice are negatively connected to melatonin receptor agonist like Ramelteon. We demonstrate that ramelteon improves the cerebral blood flow signals in ischemic stroke that is potentially mediated, at least, partly by mechanisms of activating endothelial nitric oxide synthase. Our results also show that the neuroprotection of ramelteon counteracts reactive oxygen species‐induced oxidative stress and activates the nuclear factor erythroid 2‐related factor 2/heme oxygenase‐1 pathway. Ramelteon inhibits the mitochondrial and autophagic death pathways in MCAO mice and cultured neurons, consistent with gene set enrichment analysis from a bioinformatics perspective angle. Our data suggest that Ramelteon is a potential neuroprotective drug candidate, and MT1 is the neuroprotective target for ischemic stroke, which provides new insights into stroke therapy. MT1‐KO mice and cultured neurons may provide animal and cellular models of accelerated ischemic damage and neuronal cell death.


Models of neurodegenerative diseases and outcomes after intervention.
ACSL4-Mediated Ferroptosis and Its Potential Role in Central Nervous System Diseases and Injuries

June 2023

·

23 Reads

·

10 Citations

International Journal of Molecular Sciences

As an iron-dependent regulated form of cell death, ferroptosis is characterized by iron-dependent lipid peroxidation and has been implicated in the occurrence and development of various diseases, including nervous system diseases and injuries. Ferroptosis has become a potential target for intervention in these diseases or injuries in relevant preclinical models. As a member of the Acyl-CoA synthetase long-chain family (ACSLs) that can convert saturated and unsaturated fatty acids, Acyl—CoA synthetase long-chain familymember4 (ACSL4) is involved in the regulation of arachidonic acid and eicosapentaenoic acid, thus leading to ferroptosis. The underlying molecular mechanisms of ACSL4-mediated ferroptosis will promote additional treatment strategies for these diseases or injury conditions. Our review article provides a current view of ACSL4-mediated ferroptosis, mainly including the structure and function of ACSL4, as well as the role of ACSL4 in ferroptosis. We also summarize the latest research progress of ACSL4-mediated ferroptosis in central nervous system injuries and diseases, further proving that ACSL4-medicated ferroptosis is an important target for intervention in these diseases or injuries.


The change in IL-33 and ST2 expressions in human or mice skin wounds over time. A, B, and C Representative images of H&E and Immunohistochemical staining for IL-33 and ST2 (scale bar = 50 μm) in injured and control groups of human skin. D Semi-quantitative analysis of IL-33- and ST2-positive cells relative to the total number of cells. The data were expressed as means ± SEM (n=11). ##P<0.01 vs Control group. E The cartoon displays the experimental setup. F The expression of IL-33 protein showed a hump-like trend after skin wound healing, with a trough at 3 hs and peaks at 24 hs and 10 ds. G The expression trends of ST2 presented a parabolic form, along with the peaks at 12 hs and 7 ds. H and I The optical densities of IL-33 or ST2 protein bands mentioned above were quantitatively analyzed. J Immunohistochemical results showed expression and distribution of IL-33 in each group at indicated times post-SWH (scale bar = 50 μm). K Semi-quantitative analysis of IL-33-positive cells relative to the total number of cells. The data were expressed as means ± SEM (n=5). ##P<0.01 vs Sham group, #P<0.05 vs Sham group. **P<0.01 vs preceding posttraumatic group, *P<0.05 vs preceding posttraumatic group. ns represents no significance. Experiments are representative of three independent experiments
The expressions and distributions of IL-33 and ST2 in macrophages after SWH. Representative microphotographs of co-localization of IL-33-like immunoreactivity with F4/80 in the different groups on day 3 post-SWH (scale bar = 50 μm). B and C Analysis of the contribution of IL-33 to the F4/80-positive cell population and Pearson’s correlation coefficient between them. D The co-localization of ST2-like immunoreactivity and F4/80 (scale bar = 50 μm). E and F Analysis of the contribution of ST2 to F4/80-positive cell population and Pearson’s correlation coefficient between them. Fiji image J software was used to calculate colocalization coefficients. The data were expressed as means ± SEM (n=5). ##P < 0.01 vs Sham group. Experiments are representative of three independent experiments
The expression and distribution of IL-33 in myofibroblasts after SWH. Representative microphotographs of co-localization of IL-33-like immunoreactivity with CD31 in the different groups on day 3 post-SWH (scale bar = 50 μm). B and C Analysis of the contribution of IL-33 to CD31-positive cell population and Pearson’s correlation coefficient between them. Fiji image J software was used to calculate colocalization coefficients. D Representative microphotographs of co-localization of IL-33-like immunoreactivity with α-SMA in the different groups on day 3 post-SWH (scale bar = 50 μm). E and F Analysis of the contribution of IL-33 to α-SMA-positive cell population and Pearson’s correlation coefficient between them. Fiji image J software was used to calculate colocalization coefficients. The data were expressed as means ± SEM (n=5). ##P<0.01 vs Sham group. Experiments are representative of three independent experiments
IL-33-induced facilitation in wound area closure after SWH is dependent on the ST2 signaling (A) The cartoon displays the experimental setup. (B) Representative images of wound area closure on day 3 post-SWH. Scale Bar = 5 mm. (C) Quantification of wound healing is expressed as a percent of original wound size. (D, E, and G) Representative images of H&E, immunohistochemical staining for vimentin (scale bar = 50 μm), and immunofluorescence staining for K14 (scale bar = 100 μm) of different groups on day 3 post-SWH. (F and H) Quantification of the rates of K14-positive keratinocytes and vimentin-positive fibroblasts. The data were expressed as means ± SEM (n=5). ##P<0.01 vs Sham+PBS group. **P<0.05 vs SWH+PBS group; *P<0.05 vs SWH+PBS group. &&P<0.01 vs SWH+IL-33 group. $$P<0.01 vs SWH+Anti-IL-33 group; $P<0.05 vs SWH+Anti-IL-33 group. ns represents no significance. Experiments are representative of three independent experiments
Extended characterization of IL-33/ST2 as a predictor for wound age determination in skin wound tissue samples of humans and mice

May 2023

·

38 Reads

·

2 Citations

International Journal of Legal Medicine

Interleukin (IL)-33, an important inflammatory cytokine, is highly expressed in skin wound tissue and serum of humans and mice, and plays an essential role in the process of skin wound healing (SWH) dependent on the IL-33/suppression of tumorigenicity 2 (ST2) pathway. However, whether IL-33 and ST2 themselves, as well as their interaction, can be applied for skin wound age determination in forensic practice remains incompletely characterized. Human skin samples with injured intervals of a few minutes to 24 hours (hs) and mouse skin samples with injured intervals of 1 h to 14 days (ds) were collected. Herein, the results demonstrated that IL-33 and ST2 are increased in the human skin wounds, and that in mice skin wounds, there is an increase over time, with IL-33 expression peaking at 24 hs and 10 ds, and ST2 expression peaking at 12 hs and 7 ds. Notably, the relative quantity of IL-33 and ST2 proteins < 0.35 suggested a wound age of 3 hs; their relative quantity > 1.0 suggested a wound age of 24 hs post-mouse skin wounds. In addition, immunofluorescent staining results showed that IL-33 and ST2 were consistently expressed in the cytoplasm of F4/80-positive macrophages and CD31-positive vascular endothelial cells with or without skin wounds, whereas nuclear localization of IL-33 was absent in α-SMA-positive myofibroblasts with skin wounds. Interestingly, IL-33 administration facilitated the wound area closure by increasing the proliferation of cytokeratin (K) 14 -positive keratinocytes and vimentin-positive fibroblasts. In contrast, treating with its antagonist (i.e., anti-IL-33) or receptor antagonist (e.g., anti-ST2) exacerbated the aforementioned pathological changes. Moreover, treatment with IL-33 combined with anti-IL-33 or anti-ST2 reversed the effect of IL-33 on facilitating skin wound closure, suggesting that IL-33 administration facilitated skin wound closure through the IL-33/ST2 signaling pathway. Collectively, these findings indicate that the detection of IL-33/ST2 might be a reliable biomarker for the determination of skin wound age in forensic practice.


Molecular component changes due to AUD, PTSD, and AUD/PTSD.
Summary of important data that emerged from the review.
Molecular Toxicology and Pathophysiology of Comorbid Alcohol Use Disorder and Post-Traumatic Stress Disorder Associated with Traumatic Brain Injury

May 2023

·

70 Reads

·

3 Citations

International Journal of Molecular Sciences

The increasing comorbidity of alcohol use disorder (AUD) and post-traumatic stress disorder (PTSD) associated with traumatic brain injury (TBI) is a serious medical, economic, and social issue. However, the molecular toxicology and pathophysiological mechanisms of comorbid AUD and PTSD are not well understood and the identification of the comorbidity state markers is significantly challenging. This review summarizes the main characteristics of comorbidity between AUD and PTSD (AUD/PTSD) and highlights the significance of a comprehensive understanding of the molecular toxicology and pathophysiological mechanisms of AUD/PTSD, particularly following TBI, with a focus on the role of metabolomics, inflammation, neuroendocrine, signal transduction pathways, and genetic regulation. Instead of a separate disease state, a comprehensive examination of comorbid AUD and PTSD is emphasized by considering additive and synergistic interactions between the two diseases. Finally, we propose several hypotheses of molecular mechanisms for AUD/PTSD and discuss potential future research directions that may provide new insights and translational application opportunities.


Ruxolitinib, a promising therapeutic candidate for traumatic brain injury through maintaining the homeostasis of cathepsin B

February 2023

·

8 Reads

·

2 Citations

Experimental Neurology

Traumatic brain injury (TBI) is one of the main causes of death and disability in the world. Owing to the heterogeneity and complexity of TBI pathogenesis, there is still no specific drug. Our previous studies have proved the neuroprotective effect of Ruxolitinib (Ruxo) on TBI, but further are needed to explore the potent mechanisms and potential translational application. Compelling evidence indicates that Cathepsin B (CTSB) plays an important role in TBI. However, the relationships between Ruxo and CTSB upon TBI remain non-elucidated. In this study, we established a mouse model of moderate TBI to clarify it. The neurological deficit in the behavioral test was alleviated when Ruxo administrated at 6 h post-TBI. Additionally, Ruxo significantly reduced the lesion volume. As for the pathological process of acute phase, Ruxo remarkably reduced the expression of proteins associated with cell demise, neuroinflammation, and neurodegeneration. Then the expression and location of CTSB were detected respectively. We found that the expression of CTSB exhibits a transient decrease and then persistent increase following TBI. The distribution of CTSB, mainly located at NeuN-positive neurons was unchanged. Importantly, the dysregulation of CTSB expression was reversed with the treatment of Ruxo. The timepoint was chosen when CTSB decreased, to further analyze its change in the extracted organelles; and Ruxo maintained the homeostasis of it in sub-cellular. In summary, our results demonstrate that Ruxo plays neuroprotection through maintaining the homeostasis of CTSB, and will be a promising therapeutic candidate for TBI in clinic.


Melatonin ameliorates neurological deficits through MT2/IL-33/ferritin H signaling-mediated inhibition of neuroinflammation and ferroptosis after traumatic brain injury

February 2023

·

54 Reads

·

17 Citations

Free Radical Biology and Medicine

Although traumatic brain injury (TBI) is a common cause of death and disability worldwide, there is currently a lack of effective therapeutic drugs and targets. To reveal the complex pathophysiologic mechanisms of TBI, we performed transcriptome analysis of the mouse cerebral cortex and immunohistochemical analysis of human cerebral tissues. The genes Mt1, Mt2, Il33, and Fth1 were upregulated post-TBI and enriched in pathways associated with the inflammatory response, oxidative phosphorylation, and ferroptosis. As an agonist of MT1/2, melatonin (MLT) confers anti-oxidant, anti-inflammatory, and anti-ferroptosis effects after TBI. However, whether these upregulated genes and their corresponding pathways are involved in the neuroprotective effect of MLT remains unclear. In this study, interventions to inhibit MT1/2, IL-33, and ferroptosis (i.e., ferritin H (Fth)-KO) were applied post-TBI. The results showed that MLT attenuated TBI-induced cerebral edema and neurological outcomes by inhibiting inflammation and ferroptosis. Mechanistically, MLT mainly suppressed inflammatory responses and ferroptosis via the activation of MT2 and IL-33 pathways. Building on the previous finding that Fth deletion increases susceptibility to ferroptosis post-TBI, we demonstrated that Fth depletion remarkably exacerbated the post-TBI inflammatory response, and abolished the anti-inflammatory effects of MLT both in vivo and in vitro. Furthermore, the post-TBI anti-inflammatory effect of MLT, which occurs by promoting the polarization of CD206+ macrophages, was dependent on Fth. Taken together, these results clarified that MLT alleviates inflammation- and ferroptosis-mediated brain edema and neurological deficits by activating the MT2/IL-33/Fth pathway, which provides a novel target and theoretical basis for MLT to treat TBI patients.


Figure 3
Figure 7
Melatonin ameliorates neurological deficits through MT2/IL-33/ferritin H signaling-mediated inhibition of neuroinflammation and ferroptosis after traumatic brain injury

December 2022

·

76 Reads

Although traumatic brain injury (TBI) is a common cause of death and disability worldwide, there is currently a lack of effective therapeutic drugs or targets. To reveal the complex pathophysiologic mechanisms of TBI, we performed transcriptome analysis of the mouse cerebral cortex and immunohistochemical analysis of human cerebral tissues. The genes Mt1, Mt2, Il33, and Fth1 were upregulated post-TBI and enriched in pathways associated with inflammatory response, oxidative phosphorylation, and ferroptosis. As an agonist of MT1/2, melatonin (MLT) has anti-oxidative stress, anti-inflammatory, and anti-ferroptosis properties after TBI. However, whether these upregulated genes and their corresponding pathways are involved in the neuroprotective effect of MLT remains unclear. In this study, interventions to inhibit MT1/2, IL-33, and ferroptosis (i.e., ferritin H (Fth)- KO) were applied post-TBI. The results showed that MLT attenuated TBI-induced cerebral edema and neurological outcomes by inhibiting inflammation and ferroptosis. Mechanistically, MLT mainly suppressed inflammatory responses and ferroptosis via the activation of MT2 and IL-33 pathways. Building on the previous finding that Fth deletion increases susceptibility to ferroptosis post-TBI, we demonstrated that Fth depletion remarkably exacerbated the post-TBI inflammatory response, and abolished the anti-inflammatory effect of MLT in vivo and in vitro. What’s more, the post-TBI anti-inflammatory effect of MLT, which occurs by promoting the transformation of M2 phenotype macrophages, was dependent on Fth. Taken together, these results clarified that MLT alleviates inflammation- and ferroptosis-mediated brain edema and neurological deficits by activating the MT2/IL-33/Fth pathway, which provides a novel target and theoretical basis for MLT to treat TBI patients.


TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway

December 2022

·

42 Reads

·

28 Citations

Free Radical Biology and Medicine

Ferroptosis is a form of regulated cell death that is mainly triggered by iron-dependent lipid peroxidation. A growing body of evidence suggests that ferroptosis is involved in the pathophysiology of traumatic brain injury (TBI), and tropomyosin-related kinase B (TrkB) deficiency would mediate TBI pathologies. As an agonist of TrkB and an immediate precursor of melatonin, N-acetyl serotonin (NAS) exerts several beneficial effects on TBI, but there is no information regarding the role of NAS in ferroptosis after TBI. Here, we examined the effect of NAS treatment on TBI-induced functional outcomes and ferroptosis. Remarkably, the administration of NAS alleviated TBI-induced neurobehavioral deficits, lesion volume, and neurodegeneration. NAS also rescued TBI-induced mitochondrial shrinkage, the changes in ferroptosis-related molecule expression, and iron accumulation in the ipsilateral cortex. Similar results were obtained with a well-established ferroptosis inhibitor, liproxstatin-1. Furthermore, NAS activated the TrkB/PI3K/Akt/Nrf2 pathway in the mouse model of TBI, while inhibition of PI3K and Nrf2 weakened the protection of NAS against ferroptosis both in vitro and in vivo, suggesting that a possible pathway linking NAS to the action of anti-ferroptosis was TrkB/PI3K/Akt/Nrf2. Given that ferritin H (Fth) is a known transcription target of Nrf2, we then investigated the effects of NAS on neuron-specific Fth knockout (Fth-KO) mice. Strikingly, Fth deletion almost abolished the protective effects of NAS against TBI-induced ferroptosis and synaptic damage, although Fth deletion-induced susceptibility toward ferroptosis after TBI was reversed by an iron chelator, deferoxamine. Taken together, these data indicate that the TrkB agonist NAS treatment appears to improve brain function after TBI by suppressing ferroptosis, at least in part, through activation of the PI3K/Akt/Nrf2/Fth pathway, providing evidence that NAS is likely to be a promising anti-ferroptosis agent for further treatment for TBI.


Citations (38)


... Ferroptosis is a recently discovered form of cell death controlled by iron [13]. Prior research has indicated a strong connection between ferroptosis and neuroinflammation [14,15]. Studies have shown that neuroinflammation can trigger intracellular iron buildup, speed up iron overload, facilitate the synthesis of reactive oxygen species (ROS), and further result in ferroptosis [16,17], whereas ferroptosis enhances neuroinflammatory reactions [18,19]. ...

Reference:

New Insight into Neuropathic Pain: The Relationship between α7nAChR, Ferroptosis, and Neuroinflammation
The Interplay between Ferroptosis and Neuroinflammation in Central Neurological Disorders

Antioxidants

... xCT inhibition also attenuates glutamine metabolism and lowers nutrient flexibility (12). Acyl-CoA synthetase long-chain family member 4 (ACSL4) increases levels of polyunsaturated fatty acids (PUFAs), raising sensitivity to ferroptosis by increasing lipid peroxidation (13). Moreover, high levels of lipid droplets could proportionally elevate susceptibility to ferroptosis because of elevated PUFAs and lipophagy activation (14). ...

ACSL4-Mediated Ferroptosis and Its Potential Role in Central Nervous System Diseases and Injuries

International Journal of Molecular Sciences

... However, this trauma may be physical or emotional, suggesting that the disorder does not require neurologic injury. Though it may not be inherent in all cases, many studies indicate a correlation between traumatic brain injury (TBI) and PTSD [2][3][4][5][6]. One meta-analysis found a 13.5% prevalence rate of PTSD in civilians who had been diagnosed with mild traumatic brain injury (mTBI) [2]. ...

Molecular Toxicology and Pathophysiology of Comorbid Alcohol Use Disorder and Post-Traumatic Stress Disorder Associated with Traumatic Brain Injury

International Journal of Molecular Sciences

... The intricate interplay between melatonin and microglial activation (Hardeland R, 2021;Guo er al., 2023). Through an integrative analysis, we explore the neuroprotective effects of melatonin against dBL-induced hippocampal inflammation and microglial activation, drawing upon the foundational work of Gao et al. (2023), which underscore the hormone's capacity to modulate neuroinflammatory responses. Moreover, the study by Wongprayoon and Govitrapong (2015) highlights melatonin's ability to counteract neuroinflammation through receptormediated pathways, providing a molecular basis for its therapeutic potential. ...

Melatonin ameliorates neurological deficits through MT2/IL-33/ferritin H signaling-mediated inhibition of neuroinflammation and ferroptosis after traumatic brain injury
  • Citing Article
  • February 2023

Free Radical Biology and Medicine

... The N-acetyl-5-hydroxytryptamine (NAS), acting as a TrkB agonist and direct melatonin precursor, exhibits multiple advantageous effects on traumatic brain injury (TBI) [130]. Administration of NAS reduces TBI-induced neurobehavioral deficits, lesion volume, and neurodegeneration. ...

TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway
  • Citing Article
  • December 2022

Free Radical Biology and Medicine

... Additionally, IL-33 disrupts the Th17/Treg balance in the lungs, leading to cytokine storms and immune damage. IL-33 also causes hyperactivation of ILC2s, differentiation of M2 macrophages, and secretion of TGF-β and IL-13 [31]. Common coronavirus infections in children trigger host responses similar to those induced by other viruses, such as increased expression of IL6 and ACE2 [32]. ...

Emerging Effects of IL-33 on COVID-19

International Journal of Molecular Sciences

... 27 Multimechanistic pharmacologic interventions for TBI, including antioxidants, antiexcitatory, and anti-inflammatory treatments, as well as combined therapies, have been investigated. [28][29][30][31][32] Combining medications from different classes aims to enhance synergistic effects, yet not all have translated successfully to clinical practice. 33 TBI pathophysiology is complex and evolves over time, requiring adaptable interventions tailored to different stages. ...

Targeting Molecular Mediators of Ferroptosis and Oxidative Stress for Neurological Disorders

... Liproxstatin-1, an inhibitor of ferroptosis, significantly improves the neurological function of SAH mice and inhibits inflammatory response and proinflammatory polarization of microglia in the brain, suggesting that suppression of ferroptosis may inhibit M1 polarization of microglia [19]. In addition, activation of ferroptosis in microglia may suppress the activity of M2 microglia, and promoting ferroptosis in M1 microglia may inhibit M1 microglia-mediated activation of inflammatory pathways and improve inflammatory response in SAH [20,21]. Thus, activation of ferroptosis in microglia following SAH may promote M1 polarization and subsequent inflammatory response and suppress M2 polarization, thus aggravating SAH-induced brain injury. ...

Targeting iNOS Alleviates Early Brain Injury After Experimental Subarachnoid Hemorrhage via Promoting Ferroptosis of M1 Microglia and Reducing Neuroinflammation

Molecular Neurobiology

... In addition, longterm neurodevelopment improves directly correlates to the serum decrease maintained over time. The role of IL-13 in brain damage is still unclear and the interpretation of its plasma levels is controversial, since recent animal experimentation attributes to this cytokine: anti-inflammatory and neuroprotective effects [53,54], reactivation of neuronal pyroptosis and increased neurological deficits in models of cerebral hemorrhage [55,56]. In a recent clinical trial in asphyxiated human neonates, Massaro et al. observed that an early plasma increase (24 hours) of cytokines such as IL-1 and IL-13 was related to greater brain injury as seen on magnetic resonance imaging. ...

Interleukin-13 Affects the Recovery Processes in a Mouse Model of Hemorrhagic Stroke with Bilateral Tibial Fracture

Molecular Neurobiology

... When administered, Fer-II can reduce the expression of TfR1 in neurons, which helps protect them from damage and neurodegeneration caused by TBI [98]. The molecular mechanism behind Fer-II involves reducing Fe3+ content and iron deposits; reversing the expression of proteins related to iron homeostasis such as R1 and lipid peroxidation genes; and lowering high levels of MDA after TBI [128]. When recombinant Annexin A5 (A5) was administered via the tail vein in TBI mice, it exhibited ameliorative effects on neurological deficits, weight loss, cerebral hypoperfusion, cerebral edema, blood-brain barrier disruption, neuronal apoptosis, and iron-induced cell death. ...

Ferristatin II, an Iron Uptake Inhibitor, Exerts Neuroprotection against Traumatic Brain Injury via Suppressing Ferroptosis
  • Citing Article
  • February 2022

ACS Chemical Neuroscience