732 results on '"Zhang, John H."'
Search Results
2. Corrigendum to "Activation of Galanin receptor 1 with M617 attenuates neuronal apoptosis via ERK/GSK-3β/TIP60 pathway after subarachnoid hemorrhage in Rats" [Neurotherapeutics 18 (3) (2021) 1905-1921].
- Author
-
Shi H, Fang Y, Huang L, Gao L, Lenahan C, Okada T, Travis ZD, Xie S, Tang H, Lu Q, Liu R, Tang J, Cheng Y, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
3. Inhibition of acid-sensing receptor GPR4 attenuates neuronal ferroptosis via RhoA/YAP signaling in a rat model of subarachnoid hemorrhage.
- Author
-
He Q, Zhou Y, Wu L, Huang L, Yuan Y, Flores JJ, Luo X, Tao Y, Chen X, Kanamaru H, Dong S, Zhu S, Yu Q, Han M, Sherchan P, Li J, Tang J, Xie Z, and Zhang JH
- Abstract
Background and Purpose: Subarachnoid hemorrhage (SAH) is a devastating stroke, in which acidosis is one of detrimental complications. The extracellular pH reduction can activate G protein-coupled receptor 4 (GPR4) in the brain. Yet, the extent to which proton-activated GPR4 contributes to the early brain injury (EBI) post-SAH remains largely unexplored. Ferroptosis, iron-dependent programmed cell death, has recently been shown to contribute to EBI. We aimed to investigate the effects of GPR4 inhibition on neurological deficits and neuronal ferroptosis after SAH in rats., Methods: A total 253 Sprague Dawley (SD) male rats (weighing 275-330g) were utilized in this study. SAH was induced by endovascular perforation. NE-52-QQ57 (NE), a selective antagonist of GPR4 was administered intraperitoneally 1-h post-SAH. To explore the mechanisms, RhoA activator U-46619 and YAP activator PY-60 were delivered intracerebroventricularly. Short- and long-term neurobehavior, SAH grading, Western blot assay, ELISA assay, immunofluorescence staining, and transmission electron microscopy was performed post-SAH., Results: Following SAH, there was an upregulation of GPR4 expression in neurons. GPR4 inhibition by NE improved both short-term and long-term neurological outcomes post-SAH. NE also reduced neuronal ferroptosis, as evidenced by decreased lipid peroxidation products 4HNE and MDA levels in brain tissues, and reduced mitochondrial shrinkage, increased mitochondria crista and decreased membrane density. The application of either U-46619 or PY-60 partially offset the neuroprotective effects of NE on neuronal ferroptosis in SAH rats., Conclusions: This study demonstrated that acid-sensing receptor GPR4 contributed to neuronal ferroptosis after SAH via RhoA/YAP pathway, and NE may be a potential therapeutic strategy to attenuate GPR4 mediated neuronal ferroptosis and EBI after SAH., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
4. Corrigendum to "Adiponectin attenuates neuronal apoptosis induced by hypoxia-ischemia via the activation of AdipoR1/APPL1/LKB1/AMPK pathway in neonatal rats" [Neuropharmacology 133 (2018) 415-428/NP_7092].
- Author
-
Xu N, Zhang Y, Doycheva DM, Ding Y, Zhang Y, Tang J, Guo H, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
5. Cerebrospinal fluid markers of neuroinflammation and coagulation in severe cerebral edema and chronic hydrocephalus after subarachnoid hemorrhage: a prospective study.
- Author
-
Fang Y, Liu Y, Chen L, Wang J, Zhang J, Zhang H, Tian S, Zhang A, Zhang J, Zhang JH, Wang X, Yu J, and Chen S
- Subjects
- Humans, Male, Female, Middle Aged, Prospective Studies, Animals, Aged, Mice, Neuroinflammatory Diseases cerebrospinal fluid, Neuroinflammatory Diseases etiology, Adult, Chronic Disease, Mice, Inbred C57BL, Blood Coagulation physiology, Hydrocephalus cerebrospinal fluid, Hydrocephalus etiology, Subarachnoid Hemorrhage cerebrospinal fluid, Subarachnoid Hemorrhage complications, Brain Edema cerebrospinal fluid, Brain Edema etiology, Cytokines cerebrospinal fluid, Cytokines blood, Biomarkers cerebrospinal fluid, Biomarkers blood
- Abstract
Background: Early severe cerebral edema and chronic hydrocephalus are the primary cause of poor prognosis in patients with subarachnoid hemorrhage (SAH). This study investigated the role of cerebrospinal fluid (CSF) inflammatory cytokines and coagulation factors in the development of severe cerebral edema and chronic hydrocephalus in patients with SAH., Methods: Patients with SAH enrolled in this study were categorized into mild and severe cerebral edema groups based on the Subarachnoid Hemorrhage Early Brain Edema Score at admission. During long-term follow-up, patients were further classified into hydrocephalus and non-hydrocephalus groups. CSF samples were collected within 48 h post-SAH, and levels of inflammatory cytokines and coagulation factors were measured. Univariate and multivariate logistic regression analyses were performed to identify independent factors associated with severe cerebral edema and chronic hydrocephalus. The correlation between inflammatory cytokines and coagulation factors was further investigated and validated in a mouse model of SAH., Results: Seventy-two patients were enrolled in the study. Factors from the extrinsic coagulation pathway and inflammatory cytokines were associated with both severe cerebral edema and chronic hydrocephalus. Coagulation products thrombin-antithrombin complexes (TAT) and fibrin, as well as inflammatory cytokines IL-1β, IL-2, IL-5, IL-7, and IL-4, were independently associated with severe cerebral edema. Additionally, Factor VII, fibrin, IL-2, IL-5, IL-12, TNF-α, and CCL-4 were independently associated with chronic hydrocephalus. A positive correlation between extrinsic coagulation factors and inflammatory cytokines was observed. In the SAH mouse model, tissue plasminogen activator was shown to alleviate neuroinflammation and cerebral edema, potentially by restoring glymphatic-meningeal lymphatic function., Conclusions: Elevated levels of inflammatory cytokines and extrinsic coagulation pathway factors in the CSF are associated with the development of early severe cerebral edema and chronic hydrocephalus following SAH. These factors are interrelated and may contribute to post-SAH glymphatic-meningeal lymphatic dysfunction., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Mitochondrial ferritin upregulation by deferiprone reduced neuronal ferroptosis and improved neurological deficits via NDRG1/Yap pathway in a neonatal rat model of germinal matrix hemorrhage.
- Author
-
Yuan Y, Yang X, Zhao Y, Flores JJ, Huang L, Gu L, Li R, Zhang X, Zhu S, Dong S, Kanamaru H, He Q, Tao Y, Yi K, Han M, Chen X, Wu L, Zhang JH, Xie Z, and Tang J
- Abstract
Ferroptosis contributes to brain injury after germinal matrix hemorrhage (GMH). Mitochondrial ferritin (FTMT), a novel mitochondrial outer membrane protein, reduces oxidative stress in neurodegenerative diseases. In vitro, Deferiprone has been shown to upregulate FTMT. However, the effects of FTMT upregulation by Deferiprone on neuronal ferroptosis after GMH and its underlying mechanism has not been investigated. In our study, 389 Sprague-Dawley rat pups of postnatal day 7 were used to establish a collagenase-induced GMH model and an iron-overload model of intracerebral FeCl
2 injection. The brain expressions of FTMT, N-myc downstream-regulated gene-1 (NDGR1), Yes-associated protein (YAP), ferroptosis-related molecules including transferrin receptor (TFR) and acyl-CoA synthase long-chain family member 4 (ACSL4) were increased after GMH. FTMT agonist Deferiprone improved neurological deficits and hydrocephalus after GMH. Deferiprone or Adenovirus-FTMT enhanced YAP phosphorylation at the Ser127 site and attenuated ferroptosis, which was reversed by NDRG1 CRISPR Knockout. Iron overload induced neuronal ferroptosis and neurological deficits, which were improved by YAP CRISPR Knockout. Collectively, FTMT upregulation by Deferiprone reduced neuronal ferroptosis and neurological deficits via the NDRG1/YAP signaling pathway after GMH. Deferiprone may serve as a potential non-invasive treatment for GMH patients., Competing Interests: Declaration of conflicting interestsThe author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.- Published
- 2024
- Full Text
- View/download PDF
7. Corrigendum to "Ezetimibe, a NPC1L1 inhibitor, attenuates neuronal apoptosis through AMPK dependent autophagy activation after MCAO in rats" [Experimental Neurology, 2018 Sep:307:12-23.].
- Author
-
Yu J, Li X, Matei N, McBride D, Tang J, Yan M, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
8. Corrigendum to "Neurogenesis changes and the fate of progenitor cells after subarachnoid hemorrhage in rats" [Experimental Neurology 311 (2019) 274-284].
- Author
-
Zuo Y, Wang J, Enkhjargal B, Doycheva D, Yan X, Zhang JH, and Liu F
- Published
- 2024
- Full Text
- View/download PDF
9. Correction to "Celastrol Ameliorates Neuronal Mitochondrial Dysfunction Induced by Intracerebral Hemorrhage via Targeting cAMP-Activated Exchange Protein-1".
- Author
-
Li X Sr, Liu W, Jiang G, Lian J, Zhong Y, Zhou J, Li H, Xu X, Liu Y, Cao C, Tao J, Cheng J, Zhang JH, and Chen G
- Published
- 2024
- Full Text
- View/download PDF
10. Correction to: Gas6 Promotes Microglia Efferocytosis and Suppresses Inflammation Through Activating Axl/Rac1 Signaling in Subarachnoid Hemorrhage Mice.
- Author
-
Tang J, Jin Y, Jia F, Lv T, Manaenko A, Zhang LF, Zhang Z, Qi X, Xue Y, Zhao B, Zhang X, Zhang JH, Lu J, and Hu Q
- Published
- 2024
- Full Text
- View/download PDF
11. Corrigendum to "Delayed recanalization at 3 days after permanent MCAO attenuates neuronal apoptosis through FGF21/FGFR1/PI3K/Caspase-3 pathway in rats" [Exp Neurol. 2019 Oct: 320:113007].
- Author
-
Zheng W, Matei N, Pang J, Luo X, Song Z, Tang J, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
12. PEDF-34 attenuates neurological deficit and suppresses astrocyte-dependent neuroinflammation by modulating astrocyte polarization via 67LR/JNK/STAT1 signaling pathway after subarachnoid hemorrhage in rats.
- Author
-
Wu L, Liu Y, He Q, Ao G, Xu N, He W, Liu X, Huang L, Yu Q, Kanamaru H, Dong S, Zhu S, Yuan Y, Han M, Ling Y, Liu L, Wu C, Zhou Y, Sherchan P, Flores JJ, Tang J, Chen X, He X, and Zhang JH
- Subjects
- Animals, Male, Rats, Cell Polarity, Cells, Cultured, MAP Kinase Signaling System, Rats, Sprague-Dawley, Signal Transduction, Astrocytes drug effects, Astrocytes metabolism, Nerve Growth Factors metabolism, Neuroinflammatory Diseases drug therapy, Neuroinflammatory Diseases metabolism, Serpins metabolism, STAT1 Transcription Factor metabolism, Subarachnoid Hemorrhage drug therapy, Subarachnoid Hemorrhage metabolism
- Abstract
Background: Reactive astrocytes participate in various pathophysiology after subarachnoid hemorrhage (SAH), including neuroinflammation, glymphatic-lymphatic system dysfunction, brain edema, BBB disruption, and cell death. Astrocytes transform into two new reactive phenotypes with changed morphology, altered gene expression, and secretion profiles, termed detrimental A1 and beneficial A2. This study investigates the effect of 67LR activation by PEDF-34, a PEDF peptide, on neuroinflammation and astrocyte polarization after the experimental SAH., Methods: A total of 318 male adult Sprague-Dawley rats were used in experiments in vivo, of which 272 rats were subjected to the endovascular perforation model of SAH and 46 rats underwent sham surgery. 67LR agonist (PEDF-34) was administrated intranasally 1 h after SAH. 67LR-specific inhibitor (NSC-47924) and STAT1 transcriptional activator (2-NP) were injected intracerebroventricularly 48 h before SAH. Short- and long-term neurological tests, brain water content, immunostaining, Nissl staining, western blot, and ELISA assay were performed. In experiments in vitro, primary astrocyte culture with hemoglobin (Hb) stimulation was used to mimic SAH. The expression of the PEDF-34/67LR signaling pathway and neuro-inflammatory cytokines were assessed using Western blot, ELISA, and immunohistochemistry assays both in vivo and in vitro., Results: Endogenous PEDF and 67LR expressions were significantly reduced at 6 h after SAH. 67LR was expressed in astrocytes and neurons. Intranasal administration of PEDF-34 significantly reduced brain water content, pro-inflammatory cytokines, and short-term and long-term neurological deficits after SAH. The ratio of p-JNK/JNK and p-STAT1/STAT1 and the expression of CFB and C3 (A1 astrocytes marker), significantly decreased after PEDF-34 treatment, along with fewer expression of TNF-α and IL-1β at 24 h after SAH. However, 2-NP (STAT1 transcriptional activator) and NSC-47924 (67LR inhibitor) reversed the protective effects of PEDF-34 in vivo and in vitro by promoting A1 astrocyte polarization with increased inflammatory cytokines., Conclusion: PEDF-34 activated 67LR, attenuating neuroinflammation and inhibiting astrocyte A1 polarization partly via the JNK/STAT1 pathway, suggesting that PEDF-34 might be a potential treatment for SAH patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Corrigendum to "AVE 0991 attenuates oxidative stress and neuronal apoptosis via Mas/PKA/CREB/UCP-2 pathway after subarachnoid hemorrhage in rats" [Redox Biol. 20 (2019) 75-86].
- Author
-
Mo J, Enkhjargal B, Travis ZD, Zhou K, Wu P, Zhang G, Zhu Q, Zhang T, Peng J, Xu W, Ocak U, Chen Y, Tang J, Zhang J, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
14. Corrigendum to "INT-777 attenuates NLRP3-ASC inflammasome-mediated neuroinflammation via TGR5/cAMP/PKA signaling pathway after subarachnoid hemorrhage in rats" [Brain Behav. Immun. 91 (2021) 587-600].
- Author
-
Hu X, Yan J, Huang L, Araujo C, Peng J, Gao L, Liu S, Tang J, Zuo G, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
15. Author Correction: Hemoglobin induced NO/cGMP suppression Deteriorate Microcirculation via Pericyte Phenotype Transformation after Subarachnoid Hemorrhage in Rats.
- Author
-
Li Q, Chen Y, Li B, Luo C, Zuo S, Liu X, Zhang JH, Ruan H, and Feng H
- Published
- 2024
- Full Text
- View/download PDF
16. Microglia Autophagy Mediated by TMEM166 Promotes Ischemic Stroke Secondary to Carotid Artery Stenosis.
- Author
-
Li L, Krafft PR, Zeng N, Duan R, Qi X, Shao A, Xue F, and Zhang JH
- Subjects
- Animals, Mice, Humans, Male, Aged, Middle Aged, Endarterectomy, Carotid adverse effects, Female, Disease Models, Animal, Mice, Knockout, TOR Serine-Threonine Kinases metabolism, TOR Serine-Threonine Kinases genetics, C-Reactive Protein metabolism, C-Reactive Protein genetics, Autophagy, Carotid Stenosis genetics, Carotid Stenosis metabolism, Ischemic Stroke metabolism, Ischemic Stroke genetics, Ischemic Stroke pathology, Microglia metabolism, Microglia pathology, Membrane Proteins genetics, Membrane Proteins metabolism
- Abstract
Ischemic stroke can be a serious complication of selective carotid endarterectomy (CEA) in patients with carotid artery stenosis (CAS). The underlying risk factors and mechanisms of these postoperative strokes are not completely understood. Our previous study showed that TMEM166-induced neuronal autophagy is involved in the development of secondary brain injury following cerebral ischemia-reperfusion injury in rats. This current study aimed to investigate the role of TMEM166 in ischemic stroke following CEA. In the clinical part of this study, the quantitative analysis demonstrated circulating TMEM166, interleukin 6 (IL-6), and C-reactive protein (CRP) levels were significantly elevated in patients who suffered an ischemic stroke after CEA compared to those who did not. Furthermore, non-survivors exhibited higher levels of these proteins than survivors. In the preclinical part of this study, a middle cerebral artery occlusion (MCAO) model was implemented following CAS simulation in TMEM166
-/- mice. We found TMEM166 expression was positively correlated with the degree of ischemic brain injury. Ad5-TMEM166 transfection aggravated ischemic brain injury by inducing microglial autophagy activation and release of inflammatory cytokines. Accordingly, TMEM166 deficiency reduced brain inflammation and inhibited excessive microglial autophagy through the mammalian target of rapamycin (mTOR) pathway. These findings suggest that TMEM166 may play a key role in the development of ischemic injury after CEA and may serve as a biomarker for risk assessment of postoperative ischemic stroke.- Published
- 2024
- Full Text
- View/download PDF
17. UDP-Glucose/P2Y14 Receptor Signaling Exacerbates Neuronal Apoptosis After Subarachnoid Hemorrhage in Rats.
- Author
-
Kanamaru H, Zhu S, Dong S, Takemoto Y, Huang L, Sherchan P, Suzuki H, Tang J, and Zhang JH
- Abstract
Background: Subarachnoid hemorrhage (SAH) is a severe subtype of stroke with poor outcomes. Abnormal glucose metabolism often occurs after SAH, but the strict control of blood glucose levels is not always beneficial. This study aimed to investigate the contribution of uridine diphosphate glucose (UDP-G), an intermediate of glucose/glycogen metabolism, and its receptor P2Y14 (P2Y purinoceptor 14) to SAH pathology and explored the potential targeted treatments in rats., Methods: A total of 218 Sprague-Dawley male rats were used. SAH was induced by endovascular perforation. Brain expressions of P2Y14, uridine diphosphate glucose (UDP-G), and its converting enzyme UGP2 (UDP-G pyrophosphorylase-2) were evaluated. Exogenous UDP-G or selective P2Y14 inhibitor was administered intranasally at 1 hour after SAH to explore their potential effects. Intranasal Ugp2 or P2ry14 siRNA was delivered 24 hours before SAH for mechanistic evaluation. Primary neuron culture and hemoglobin stimulation were used as in vitro model of SAH. Post-SAH evaluation included liquid chromatography-mass spectrometry measurement of brain endogenous UDP-G level, neurobehavioral assessments, Western blotting, immunohistochemistry, TUNEL staining, and Nissl staining., Results: There was an acute elevation of endogenous brain UDP-G and UGP2 after SAH, and P2Y14 was expressed in neurons. Although P2Y14 inhibitor decreased neurological dysfunction, neuronal apoptosis, and proapoptotic molecules, exogenous UDP-G exacerbated these outcomes at 24 hours after SAH. Early inhibition of P2Y14 preserved long-term neuronal survival in the hippocampus, amygdala, and cortex with improved neurocognition and depressive-like behavior. In addition, in vivo knockdown of Ugp2 - and P2ry14 -reduced neurological deficits and proapoptotic molecules at 24 hours after SAH, and furthermore in vitro knockdown of P2ry14 -reduced apoptosis in hemoglobin stimulated primary neuron., Conclusions: These findings suggest a detrimental role of brain UDP-G/P2Y14 signaling in SAH, as a part of glucose metabolic pathology at the tissue level. P2Y14 inhibitor 4-[4-(4-piperidinyl)phenyl]-7-[4-(trifluoromethyl)phenyl]-2-naphthalenecarboxylic acid hydrochloride may serve as a potential therapeutic target in treating patients with SAH., Competing Interests: Disclosures None.
- Published
- 2024
- Full Text
- View/download PDF
18. Cathepsin B as a key regulator of ferroptosis in microglia following intracerebral hemorrhage.
- Author
-
Lu J, Li H, Yu Z, Cao C, Xu Z, Peng L, Zhang JH, and Chen G
- Subjects
- Humans, Cerebral Hemorrhage pathology, Animals, Mice, Brain Injuries metabolism, Cathepsin B genetics, Cathepsin B metabolism, Ferroptosis, Microglia metabolism
- Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke marked by elevated mortality and disability rates. Recently, mounting evidence suggests a significant role of ferroptosis in the pathogenesis of ICH. Through a combination of bioinformatics analysis and basic experiments, our goal is to identify the primary cell types and key molecules implicated in ferroptosis post-ICH. This aims to propel the advancement of ferroptosis research, offering potential therapeutic targets for ICH treatment. Our study reveals pronounced ferroptosis in microglia and identifies the target gene, cathepsin B (Ctsb), by analyzing differentially expressed genes following ICH. Ctsb, a cysteine protease primarily located in lysosomes, becomes a focal point in our investigation. Utilizing in vitro and in vivo models, we explore the correlation between Ctsb and ferroptosis in microglia post-ICH. Results demonstrate that ICH and hemin-induced ferroptosis in microglia coincide with elevated levels and activity of Ctsb protein. Effective alleviation of ferroptosis in microglia after ICH is achieved through the inhibition of Ctsb protease activity and protein levels using inhibitors and shRNA. Additionally, a notable increase in m6A methylation levels of Ctsb mRNA post-ICH is observed, suggesting a pivotal role of m6A methylation in regulating Ctsb translation. These research insights deepen our comprehension of the molecular pathways involved in ferroptosis after ICH, underscoring the potential of Ctsb as a promising target for mitigating brain damage resulting from ICH., Competing Interests: Declaration of competing interest All authors claim that there are no conflicts of interest., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. Corrigendum to 'Osteopontin modulates microglial activation states and attenuates inflammatory responses after subarachnoid hemorrhage in rats' [Experimental Neurology 371 (2024) 114585].
- Author
-
Sun C, Rahman MSU, Enkhjargal B, Peng J, Zhou K, Xie Z, Wu L, Zhang T, Zhu Q, Tang J, Zeng Y, Zhang JH, and Xu S
- Published
- 2024
- Full Text
- View/download PDF
20. Corrigendum to "LRP1 activation attenuates white matter injury by modulating microglial polarization through Shc1/PI3K/Akt pathway after subarachnoid hemorrhage in rats" [Redox Biol. 21 (2019) 101121].
- Author
-
Peng J, Pang J, Huang L, Enkhjargal B, Zhang T, Mo J, Wu P, Xu W, Zuo Y, Peng J, Zuo G, Chen L, Tang J, Zhang JH, and Jiang Y
- Published
- 2024
- Full Text
- View/download PDF
21. Celastrol Ameliorates Neuronal Mitochondrial Dysfunction Induced by Intracerebral Hemorrhage via Targeting cAMP-Activated Exchange Protein-1.
- Author
-
Li X Sr, Liu W, Jiang G, Lian J, Zhong Y, Zhou J, Li H, Xu X, Liu Y, Cao C, Tao J, Cheng J, Zhang JH, and Chen G
- Subjects
- Animals, Mice, Male, Guanine Nucleotide Exchange Factors metabolism, Guanine Nucleotide Exchange Factors genetics, Neuroprotective Agents pharmacology, Triterpenes pharmacology, Mice, Inbred C57BL, Signal Transduction drug effects, Pentacyclic Triterpenes pharmacology, Cerebral Hemorrhage metabolism, Cerebral Hemorrhage drug therapy, Mitochondria metabolism, Mitochondria drug effects, Neurons metabolism, Neurons drug effects, Disease Models, Animal
- Abstract
Mitochondrial dysfunction contributes to the development of secondary brain injury (SBI) following intracerebral hemorrhage (ICH) and represents a promising therapeutic target. Celastrol, the primary active component of Tripterygium wilfordii, is a natural product that exhibits mitochondrial and neuronal protection in various cell types. This study aims to investigate the neuroprotective effects of celastrol against ICH-induced SBI and explore its underlying mechanisms. Celastrol improves neurobehavioral and cognitive abilities in mice with autologous blood-induced ICH, reduces neuronal death in vivo and in vitro, and promotes mitochondrial function recovery in neurons. Single-cell nuclear sequencing reveals that the cyclic adenosine monophosphate (cAMP)/cAMP-activated exchange protein-1 (EPAC-1) signaling pathways are impacted by celastrol. Celastrol binds to cNMP (a domain of EPAC-1) to inhibit its interaction with voltage-dependent anion-selective channel protein 1 (VDAC1) and blocks the opening of mitochondrial permeability transition pores. After neuron-specific knockout of EPAC1, the neuroprotective effects of celastrol are diminished. In summary, this study demonstrates that celastrol, through its interaction with EPAC-1, ameliorates mitochondrial dysfunction in neurons, thus potentially improving SBI induced by ICH. These findings suggest that targeting EPAC-1 with celastrol can be a promising therapeutic approach for treating ICH-induced SBI., (© 2024 The Authors. Advanced Science published by Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
22. Corrigendum to "Activation of TGR5 with INT-777 attenuates oxidative stress and neuronal apoptosis via cAMP/PKCε/ALDH2 pathway after subarachnoid hemorrhage in rats" [Free Radic. Biol. Med. (2019 Nov 1) 143 441-453].
- Author
-
Zuo G, Zhang T, Huang L, Araujo C, Peng J, Travis Z, Okadab T, Ocak U, Zhang G, Tang J, Lu X, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
23. Corrigendum to "Activation of galanin receptor 1 with M617 attenuates neuronal apoptosis via ERK/GSK-3β/TIP60 pathway after subarachnoid hemorrhage in rats" [Neurotherapeutics 18 (3) (2021) 1905-1921].
- Author
-
Shi H, Fang Y, Huang L, Gao L, Lenahan C, Okada T, Travis ZD, Xie S, Tang H, Lu Q, Liu R, Tang J, Cheng Y, and Zhang JH
- Published
- 2024
- Full Text
- View/download PDF
24. Mitochondrial ferritin upregulation reduced oxidative stress and blood-brain-barrier disruption by maintaining cellular iron homeostasis in a neonatal rat model of germinal matrix hemorrhage.
- Author
-
Yuan Y, He Q, Yang X, Flores JJ, Huang L, Luo X, Zhang X, Zhang Z, Li R, Gu L, Dong S, Zhu S, Yi K, Han M, Wu L, Zhou Y, Zhang JH, Xie Z, and Tang J
- Subjects
- Humans, Infant, Newborn, Rats, Animals, Animals, Newborn, Rats, Sprague-Dawley, Up-Regulation, Deferiprone metabolism, Deferiprone pharmacology, Cerebral Hemorrhage complications, Cerebral Hemorrhage metabolism, Oxidative Stress, Iron metabolism, Homeostasis, Ferritins metabolism, Tight Junction Proteins metabolism, Blood-Brain Barrier metabolism, Iron Overload metabolism
- Abstract
Germinal matrix hemorrhage (GMH) is a devasting neurological disease in premature newborns. After GMH, brain iron overload associated with hemoglobin degradation contributed to oxidative stress, causing disruption of the already vulnerable blood-brain barrier (BBB). Mitochondrial ferritin (FTMT), a novel mitochondrial outer membrane protein, is crucial in maintaining cellular iron homeostasis. We aimed to investigate the effect of FTMT upregulation on oxidative stress and BBB disruption associated with brain iron overload in rats. A total of 222 Sprague-Dawley neonatal rat pups (7 days old) were used to establish a collagenase-induced GMH model and an iron-overload model of intracerebral FeCl
2 injection. Deferiprone was administered via gastric lavage 1 h after GMH and given daily until euthanasia. FTMT CRISPR Knockout and adenovirus (Ad)-FTMT were administered intracerebroventricularly 48 h before GMH and FeCl2 injection, respectively. Neurobehavioral tests, immunofluorescence, Western blot, Malondialdehyde measurement, and brain water content were performed to evaluate neurobehavior deficits, oxidative stress, and BBB disruption, respectively. The results demonstrated that brain expressions of iron exporter Ferroportin (FPN) and antioxidant glutathione peroxidase 4 (GPX4) as well as BBB tight junction proteins including Claudin-5 and Zona Occulta (ZO)-1 were found to be decreased at 72 h after GMH. FTMT agonist Deferiprone attenuated oxidative stress and preserved BBB tight junction proteins after GMH. These effects were partially reversed by FTMT CRISPR Knockout. Iron overload by FeCl2 injection resulted in oxidative stress and BBB disruption, which were improved by Ad-FTMT mediated FTMT overexpression. Collectively, FTMT upregulation is neuroprotective against brain injury associated with iron overload. Deferiprone reduced oxidative stress and BBB disruption by maintaining cellular iron homeostasis partially by the upregulating of FTMT after GMH. Deferiprone may be an effective treatment for patients with GMH., Competing Interests: Declaration of competing interest All Authors declare that they have no competing financial interests or conflicts of interest in the current study., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. Exercise-Induced Reduction of IGF1R Sumoylation Attenuates Neuroinflammation in APP/PS1 Transgenic Mice.
- Author
-
Chen Y, Chen X, Luo Z, Kang X, Ge Y, Wan R, Wang Q, Han Z, Li F, Fan Z, Xie Y, Qi B, Zhang X, Yang Z, Zhang JH, Liu D, Xu Y, Wu D, and Chen S
- Abstract
Introduction: Alzheimer's Disease (AD), a progressive neurodegenerative disorder, is marked by cognitive deterioration and heightened neuroinflammation. The influence of Insulin-like Growth Factor 1 Receptor (IGF1R) and its post-translational modifications, especially sumoylation, is crucial in understanding the progression of AD and exploring novel therapeutic avenues., Objectives: This study investigates the impact of exercise on the sumoylation of IGF1R and its role in ameliorating AD symptoms in APP/PS1 mice, with a specific focus on neuroinflammation and innovative therapeutic strategies., Methods: APP/PS1 mice were subjected to a regimen of moderate-intensity exercise. The investigation encompassed assessments of cognitive functions, alterations in hippocampal protein expressions, neuroinflammatory markers, and the effects of exercise on IGF1R and SUMO1 nuclear translocation. Additionally, the study evaluated the efficacy of KPT-330, a nuclear export inhibitor, as an alternative to exercise., Results: Exercise notably enhanced cognitive functions in AD mice, possibly through modulations in hippocampal proteins, including Bcl-2 and BACE1. A decrease in neuroinflammatory markers such as IL-1β, IL-6, and TNF-α was observed, indicative of reduced neuroinflammation. Exercise modulated the nuclear translocation of SUMO1 and IGF1R in the hippocampus, thereby facilitating neuronal regeneration. Mutant IGF1R (MT IGF1R), lacking SUMO1 modification sites, showed reduced SUMOylation, leading to diminished expression of pro-inflammatory cytokines and apoptosis. KPT-330 impeded the formation of the IGF1R/RanBP2/SUMO1 complex, thereby limiting IGF1R nuclear translocation, inflammation, and neuronal apoptosis, while enhancing cognitive functions and neuron proliferation., Conclusion: Moderate-intensity exercise effectively mitigates AD symptoms in mice, primarily by diminishing neuroinflammation, through the reduction of IGF1R Sumoylation. KPT-330, as a potential alternative to physical exercise, enhances the neuroprotective role of IGF1R by inhibiting SUMOylation through targeting XPO1, presenting a promising therapeutic strategy for AD., (Copyright © 2023. Production and hosting by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
26. Intranasal administration of recombinant prosaposin attenuates neuronal apoptosis through GPR37/PI3K/Akt/ASK1 pathway in MCAO rats.
- Author
-
Yu J, Li J, Matei N, Wang W, Tang L, Pang J, Li X, Fang L, Tang J, Zhang JH, and Yan M
- Subjects
- Rats, Male, Female, Humans, Animals, Rats, Sprague-Dawley, Infarction, Middle Cerebral Artery complications, Infarction, Middle Cerebral Artery drug therapy, Infarction, Middle Cerebral Artery metabolism, Phosphatidylinositol 3-Kinases metabolism, Saposins metabolism, Saposins pharmacology, Saposins therapeutic use, Signal Transduction, Administration, Intranasal, Apoptosis, RNA, Small Interfering pharmacology, Proto-Oncogene Proteins c-akt metabolism, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use
- Abstract
Studies have reported that Prosaposin (PSAP) is neuroprotective in cerebrovascular diseases. We hypothesized that PSAP would reduce infarct volume by attenuating neuronal apoptosis and promoting cell survival through G protein-coupled receptor 37(GPR37)/PI3K/Akt/ASK1 pathway in middle cerebral artery occlusion (MCAO) rats. Two hundred and thirty-five male and eighteen female Sprague-Dawley rats were used. Recombinant human PSAP (rPSAP) was administered intranasally 1 h (h) after reperfusion. PSAP small interfering ribonucleic acid (siRNA), GPR37 siRNA, and PI3K specific inhibitor LY294002 were administered intracerebroventricularly 48 h before MCAO. Infarct volume, neurological score, immunofluorescence staining, Western blot, Fluoro-Jade C (FJC) and TUNEL staining were examined. The expression of endogenous PSAP and GPR37 were increased after MCAO. Intranasal administration of rPSAP reduced brain infarction, neuronal apoptosis, and improved both short- and long-term neurological function. Knockdown of endogenous PSAP aggravated neurological deficits. Treatment with exogenous rPSAP increased PI3K expression, Akt and ASK1 phosphorylation, and Bcl-2 expression; phosphorylated-JNK and Bax levels were reduced along with the number of FJC and TUNEL positive neurons. GPR37 siRNA and LY294002 abolished the anti-apoptotic effect of rPSAP at 24 h after MCAO. In conclusion, rPSAP attenuated neuronal apoptosis and improved neurological function through GPR37/PI3K/Akt/ASK1 pathway after MCAO in rats. Therefore, further exploration of PSAP as a potential treatment option in ischemic stroke is warranted., Competing Interests: Declaration of Competing Interest The authors declare that they have no conflict of interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
27. Mitochondrial stress: a key role of neuroinflammation in stroke.
- Author
-
Gao L, Peng L, Wang J, Zhang JH, and Xia Y
- Subjects
- Humans, Mitochondria pathology, Neuroinflammatory Diseases, Stroke pathology
- Abstract
Stroke is a clinical syndrome characterized by an acute, focal neurological deficit, primarily caused by the occlusion or rupture of cerebral blood vessels. In stroke, neuroinflammation emerges as a pivotal event contributing to neuronal cell death. The occurrence and progression of neuroinflammation entail intricate processes, prominently featuring mitochondrial dysfunction and adaptive responses. Mitochondria, a double membrane-bound organelle are recognized as the "energy workshop" of the body. Brain is particularly vulnerable to mitochondrial disturbances due to its high energy demands from mitochondria-related energy production. The interplay between mitochondria and neuroinflammation plays a significant role in the pathogenesis of stroke. The biological and pathological consequences resulting from mitochondrial stress have substantial implications for cerebral function. Mitochondrial stress serves as an adaptive mechanism aimed at mitigating the stress induced by the import of misfolded proteins, which occurs in response to stroke. This adaptive response involves a reduction in misfolded protein accumulation and overall protein synthesis. The influence of mitochondrial stress on the pathological state of stroke is underscored by its capacity to interact with neuroinflammation. The impact of mitochondrial stress on neuroinflammation varies according to its severity. Moderate mitochondrial stress can bolster cellular adaptive defenses, enabling cells to better withstand detrimental stressors. In contrast, sustained and excessive mitochondrial stress detrimentally affects cellular and tissue integrity. The relationship between neuroinflammation and mitochondrial stress depends on the degree of mitochondrial stress present. Understanding its role in stroke pathogenesis is instrumental in excavating the novel treatment of stroke. This review aims to provide the evaluation of the cross-talk between mitochondrial stress and neuroinflammation within the context of stroke. We aim to reveal how mitochondrial stress affects neuroinflammation environment in stroke., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
28. GW0742 reduces mast cells degranulation and attenuates neurological impairments via PPAR β/δ /CD300a/SHP1 pathway after GMH in neonatal rats.
- Author
-
Lu W, Huang J, Flores J, Li P, Wang W, Liu S, Zhang JH, and Tang J
- Subjects
- Humans, Rats, Animals, Animals, Newborn, Mast Cells metabolism, Chymases, Interleukin-17, Rats, Sprague-Dawley, Transforming Growth Factor beta1, Tryptases, Cerebral Hemorrhage, Thiazoles pharmacology, Inflammation, RNA, Small Interfering, PPAR delta genetics, PPAR delta metabolism, PPAR-beta genetics, PPAR-beta metabolism
- Abstract
Background: Activation of mast cells plays an important role in brain inflammation. CD300a, an inhibitory receptor located on mast cell surfaces, has been reported to reduce the production of pro-inflammatory cytokines and exert protective effects in inflammation-related diseases. Peroxisome proliferator-activated receptor β/δ (PPAR
β/δ ), a ligand-activated nuclear receptor, activation upregulates the transcription of CD300a. In this study, we aim to investigate the role of PPARβ/δ in the attenuation of germinal matrix hemorrhage (GMH)-induced mast cell activation via CD300a/SHP1 pathway., Methods: GMH model was induced by intraparenchymal injection of bacterial collagenase into the right hemispheric ganglionic eminence in P7 Sprague Dawley rats. GW0742, a PPARβ/δ agonist, was administered intranasally at 1 h post-ictus. CD300a small interfering RNA (siRNA) and PPARβ/δ siRNA were injected intracerebroventricularly 5 days and 2 days before GMH induction. Behavioral tests, Western blot, immunofluorescence, Toluidine Blue staining, and Nissl staining were applied to assess post-GMH evaluation., Results: Results demonstrated that endogenous protein levels of PPARβ/δ and CD300a were decreased, whereas chymase, tryptase, IL-17A and transforming growth factor β1 (TGF-β1) were elevated after GMH. GMH induced significant short- and long-term neurobehavioral deficits in rat pups. GW0742 decreased mast cell degranulation, improved neurological outcomes, and attenuated ventriculomegaly after GMH. Additionally, GW0742 increased expression of PPARβ/δ , CD300a and phosphorylation of SHP1, decreased phosphorylation of Syk, chymase, tryptase, IL-17A and TGF-β1 levels. PPARβ/δ siRNA and CD300a siRNA abolished the beneficial effects of GW0742., Conclusions: GW0742 inhibited mast cell-induced inflammation and improved neurobehavior after GMH, which is mediated by PPARβ/δ /CD300a/SHP1 pathway. GW0742 may serve as a potential treatment to reduce brain injury for GMH patients., Competing Interests: Declaration of Competing Interest None of the authors have any conflicts of interests related to this work., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
29. Delayed recanalization reduced neuronal apoptosis and neurological deficits by enhancing liver-derived trefoil factor 3-mediated neuroprotection via LINGO2/EGFR/Src signaling pathway after middle cerebral artery occlusion in rats.
- Author
-
Li D, Lian L, Huang L, Gamdzyk M, Huang Y, Doycheva D, Li G, Yu S, Guo Y, Kang R, Tang H, Tang J, Kong L, and Zhang JH
- Subjects
- Humans, Rats, Male, Animals, Rats, Sprague-Dawley, Neuroprotection, Infarction, Middle Cerebral Artery metabolism, Trefoil Factor-3 pharmacology, Trefoil Factor-3 therapeutic use, Signal Transduction, Apoptosis, ErbB Receptors metabolism, ErbB Receptors pharmacology, ErbB Receptors therapeutic use, Liver, RNA, Small Interfering pharmacology, Proto-Oncogene Proteins c-bcl-2 metabolism, Ischemic Stroke, Brain Ischemia, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use
- Abstract
Delayed recanalization at days or weeks beyond the therapeutic window was shown to improve functional outcomes in acute ischemic stroke (AIS) patients. However, the underlying mechanisms remain unclear. Previous preclinical study reported that trefoil factor 3 (TFF3) was secreted by liver after cerebral ischemia and acted a distant neuroprotective factor. Here, we investigated the liver-derived TFF3-mediated neuroprotective mechanism enhanced by delayed recanalization after AIS. A total of 327 male Sprague-Dawley rats and the model of middle cerebral artery occlusion (MCAO) with permanent occlusion (pMCAO) or with delayed recanalization at 3 d post-occlusion (rMCAO) were used. Partial hepatectomy was performed within 5 min after MCAO. Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein 2 (LINGO2) siRNA was administered intracerebroventricularly at 48 h after MCAO. Recombinant rat TFF3 (rr-TFF3, 30 μg/Kg) or recombinant rat epidermal growth factor (rr-EGF, 100 μg/Kg) was administered intranasally at 1 h after recanalization, and EGFR inhibitor Gefitinib (75 mg/Kg) was administered intranasally at 30 min before recanalization. The evaluation of outcomes included neurobehavior, ELISA, western blot and immunofluorescence staining. TFF3 in hepatocytes and serum were upregulated in a similar time-dependent manner after MCAO. Compared to pMCAO, delayed recanalization increased brain TFF3 levels and attenuated brain damage with the reduction in neuronal apoptosis, infarct volume and neurological deficits. Partial hepatectomy reduced TFF3 levels in serum and ipsilateral brain hemisphere, and abolished the benefits of delayed recanalization on neuronal apoptosis and neurobehavioral deficits in rMCAO rats. Intranasal rrTFF3 treatment reversed the changes associated with partial hepatectomy. Delayed recanalization after MCAO increased the co-immunoprecipitation of TFF3 and LINGO2, as well as expressions of p-EGFR, p-Src and Bcl-2 in the brain. LINGO2 siRNA knockdown or EGFR inhibitor reversed the effects of delayed recanalization on apoptosis and brain expressions of LINGO2, p-EGFR, p-Src and Bcl-2 in rMCAO rats. EGFR activator abolished the deleterious effects of LINGO2 siRNA. In conclusion, our investigation demonstrated for the first time that delayed recanalization may enhance the entry of liver-derived TFF3 into ischemic brain upon restoring blood flow after MCAO, which attenuated neuronal apoptosis and neurological deficits at least in part via activating LINGO2/EGFR/Src pathway., Competing Interests: Declaration of Competing Interest The authors declared no competing interests., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
30. Osteopontin modulates microglial activation states and attenuates inflammatory responses after subarachnoid hemorrhage in rats.
- Author
-
Sun C, Rahman MSU, Enkhjargal B, Peng J, Zhou K, Xie Z, Wu L, Zhang T, Zhu Q, Tang J, Zeng Y, Zhang JH, and Xu S
- Subjects
- Rats, Animals, Rats, Sprague-Dawley, NF-kappa B metabolism, Interleukin-10, Microglia metabolism, Tumor Necrosis Factor-alpha metabolism, Neuroinflammatory Diseases, Anti-Inflammatory Agents pharmacology, Integrins metabolism, Integrins therapeutic use, RNA, Small Interfering pharmacology, Disease Models, Animal, Osteopontin therapeutic use, Osteopontin metabolism, Osteopontin pharmacology, Subarachnoid Hemorrhage complications, Subarachnoid Hemorrhage drug therapy, Subarachnoid Hemorrhage metabolism
- Abstract
Aims: Osteopontin (OPN) has demonstrated neuroprotective effects in various stroke models. Its role in neuroinflammation after brain injury remains to be elucidated. This study aims to clarify the effect of OPN on neuroinflammation, particularly on the functional states of microglia after subarachnoid hemorrhage (SAH)., Methods: 77 rats were randomly divided into the following groups: Sham, SAH 24 h, SAH + rOPN, SAH + Vehicle (PBS), SAH + OPN siRNA, and SAH + Scr siRNA, SAH + rOPN+Fib-14 and SAH + rOPN+DMSO. Modified Garcia and beam balance tests were used to evaluate neurobehavioral outcomes. Semi-quantitative immunofluorescence staining was performed to measure expression of myeloperoxidase (MPO) and microglia activation state markers CD16, CD206 after SAH and recombinant OPN treatment. The quantification of microglia activation and functional markers CD16, CD206, TNF-α and IL-10 were further evaluated using Western-blotting., Results: Nasal administration of rOPN improved neurological dysfunction, attenuated neutrophil infiltration, and decreased expression of phenotypic and functional markers of pro-inflammatory microglia CD16 and TNF-α. It also promoted an anti-inflammatory microglial state, as evidenced by increased expression of CD206 and IL-10. Furthermore, after blocking the phosphorylation of FAK signaling, the effects of rOPN on microglial activation states were partially reversed. The downstream pathways of STAT3 and NF-κB also exhibited consistent changes, suggesting the involvement of the STAT3 and NF-κB pathways in OPN's modulation of microglial activation via integrin-FAK signaling., Conclusion: OPN attenuates inflammatory responses after SAH by promoting an anti-inflammatory microglial state, potentially mediated through the integrin-FAK-STAT3 and NF-κB signaling pathways., Competing Interests: Declaration of Competing Interest The authors declare no conflict of interest., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
31. Gas6 Promotes Microglia Efferocytosis and Suppresses Inflammation Through Activating Axl/Rac1 Signaling in Subarachnoid Hemorrhage Mice.
- Author
-
Tang J, Jin Y, Jia F, Lv T, Manaenko A, Zhang LF, Zhang Z, Qi X, Xue Y, Zhao B, Zhang X, Zhang JH, Lu J, and Hu Q
- Subjects
- Animals, Microglia pathology, Signal Transduction, Inflammation metabolism, Disease Models, Animal, Subarachnoid Hemorrhage pathology, Brain Injuries
- Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) is characterized by rapid development of neuron apoptosis and dysregulated inflammatory response. Microglia efferocytosis plays a critical role in the clearance of apoptotic cells, attenuation of inflammation, and minimizing brain injury in various pathological conditions. Here, using a mouse SAH model, we aim to investigate whether microglia efferocytosis is involved in post-SAH inflammation and to determine the underlying signaling pathway. We hypothesized that TAM receptors and their ligands regulate this process. To prove our hypothesis, the expression and cellular location of TAM (Tyro3, Axl, and Mertk) receptors and their ligands growth arrest-specific 6 (Gas6) and Protein S (ProS1) were examined by PCR, western blots, and fluorescence immunostaining. Thirty minutes after SAH, mice received an intraventricular injection of recombinant Gas6 (rGas6) or recombinant ProS1 (rPros1) and underwent evaluations of inflammatory mediator expression, neurological deficits, and blood-brain barrier integrity at 24 h. Microglia efferocytosis of apoptotic neurons was analyzed in vivo and in vitro. The potential mechanism was determined by inhibiting or knocking down TAM receptors and Rac1 by specific inhibitors or siRNA. SAH induced upregulation of Axl and its ligand Gas6. The administration of rGas6 but not rPros1 promoted microglia efferocytosis, alleviated inflammation, and ameliorated SAH-induced BBB breakdown and neurological deficits. The beneficial effects of rGas6 were arrogated by inhibiting or knocking down Axl and Rac1. We concluded that rGas6 attenuated the development of early brain injury in mice after SAH by facilitating microglia efferocytosis and preventing inflammatory response, which is partly dependent on activation of Axl and Rac1., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
32. Correction to: Endogenous Neural Stem Cell-induced Neurogenesis after Ischemic Stroke: Processes for Brain Repair and Perspectives.
- Author
-
Tang H, Li Y, Tang W, Zhu J, Parker GC, and Zhang JH
- Published
- 2023
- Full Text
- View/download PDF
33. Long Non-coding RNA H19 Promotes NLRP3-Mediated Pyroptosis After Subarachnoid Hemorrhage in Rats.
- Author
-
Liu Y, Luo Y, Zhang A, Wang Z, Wang X, Yu Q, Zhang Z, Zhu Z, Wang K, Chen L, Nie X, Zhang JH, Zhang J, Fang Y, Su Z, and Chen S
- Subjects
- Rats, Humans, Animals, Inflammasomes metabolism, NLR Family, Pyrin Domain-Containing 3 Protein genetics, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Pyroptosis, Rats, Sprague-Dawley, RNA, Long Noncoding genetics, Subarachnoid Hemorrhage complications, MicroRNAs genetics, MicroRNAs therapeutic use
- Abstract
NLRP3 inflammasomes have been reported to be an essential mediator in the inflammatory response during early brain injury (EBI) following subarachnoid hemorrhage (SAH). Recent studies have indicated that NLRP3 inflammasome-mediated pyroptosis and long non-coding RNA (lncRNA) H19 can participate in the inflammatory response. However, the roles and functions of lncRNA H19 in NLRP3 inflammasome-mediated pyroptosis during EBI after SAH are unknown and need to be further elucidated. NLRP3 inflammasome proteins were significantly elevated in CSF of human with SAH induced EBI and presented a positive correlation with severity. In ipsilateral hemisphere cortex of rats, these NLRP3 inflammasome proteins were also increased and accompanied with upregulation of H19, and both of NLRP3 and H19 were peaked at 24 h after SAH. However, knockdown of H19 markedly decreased the expression of NLRP3 inflammasome proteins at 24 h after SAH in rats and also ameliorated EBI, showing improved neurobehavioral deficits, cerebral edema, and neuronal injury. Moreover, knocking down of H19 downregulated the expression of Gasdermin D (GSDMD) in microglia in SAH rats. Similarly, knockdown of H19 also alleviated OxyHb-induced pyroptosis and NLRP3-mediated inflammasomes activation in primary microglia. Lastly, H19 competitively sponged with rno-miR-138-5p and then upregulated NLRP3 expression in the post-SAH inflammatory response. lncRNA H19 promotes NLRP3-mediated pyroptosis by functioning as rno-miR-138-5p sponge in rats during EBI after SAH, which might provide a potential therapeutic target for post-SAH inflammation regulation., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
34. Recombinant Slit2 suppresses neuroinflammation and Cdc42-mediated brain infiltration of peripheral immune cells via Robo1-srGAP1 pathway in a rat model of germinal matrix hemorrhage.
- Author
-
Li Q, Huang L, Ding Y, Sherchan P, Peng W, and Zhang JH
- Subjects
- Rats, Animals, Rats, Sprague-Dawley, Neuroinflammatory Diseases, Receptors, Immunologic genetics, Receptors, Immunologic metabolism, Brain metabolism, Cerebral Hemorrhage, RNA, Small Interfering genetics, RNA, Small Interfering therapeutic use, RNA, Small Interfering pharmacology, GTPase-Activating Proteins metabolism, Signal Transduction, Nerve Tissue Proteins metabolism
- Abstract
Background: Germinal matrix hemorrhage (GMH) is a devastating neonatal stroke, in which neuroinflammation is a critical pathological contributor. Slit2, a secreted extracellular matrix protein, plays a repulsive role in axon guidance and leukocyte chemotaxis via the roundabout1 (Robo1) receptor. This study aimed to explore effects of recombinant Slit2 on neuroinflammation and the underlying mechanism in a rat model of GMH., Methods: GMH was induced by stereotactically infusing 0.3 U of bacterial collagenase into the germinal matrix of 7-day-old Sprague Dawley rats. Recombinant Slit2 or its vehicle was administered intranasally at 1 h after GMH and daily for 3 consecutive days. A decoy receptor recombinant Robo1 was co-administered with recombinant Slit2 after GMH. Slit2 siRNA, srGAP1 siRNA or the scrambled sequences were administered intracerebroventricularly 24 h before GMH. Neurobehavior, brain water content, Western blotting, immunofluorescence staining and Cdc42 activity assays were performed., Results: The endogenous brain Slit2 and Robo1 expressions were increased after GMH. Robo1 was expressed on neuron, astrocytes and infiltrated peripheral immune cells in the brain. Endogenous Slit2 knockdown by Slit2 siRNA exacerbated brain edema and neurological deficits following GMH. Recombinant Slit2 (rSlit2) reduced neurological deficits, proinflammatory cytokines, intercellular adhesion molecules, peripheral immune cell markers, neuronal apoptosis and Cdc42 activity in the brain tissue after GMH. The anti-neuroinflammation effects were reversed by recombinant Robo1 co-administration or srGAP1 siRNA., Conclusions: Recombinant Slit2 reduced neuroinflammation and neuron apoptosis after GMH. Its anti-neuroinflammation effects by suppressing onCdc42-mediated brain peripheral immune cells infiltration was at least in part via Robo1-srGAP1 pathway. These results imply that recombinant Slit2 may have potentials as a therapeutic option for neonatal brain injuries., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
35. Fractalkine Enhances Hematoma Resolution and Improves Neurological Function via CX3CR1/AMPK/PPARγ Pathway After GMH.
- Author
-
Chen X, He X, Xu F, Xu N, Sharifi NH, Zhang P, Flores JJ, Wu L, He Q, Kanamaru H, Zhu S, Dong S, Han M, Yuan Y, Huang L, Miao L, Zhang JH, Zhou Y, and Tang J
- Subjects
- Rats, Animals, Humans, Infant, Newborn, PPAR gamma metabolism, AMP-Activated Protein Kinases metabolism, AMP-Activated Protein Kinases pharmacology, Rats, Sprague-Dawley, Neuroinflammatory Diseases, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage metabolism, Microglia metabolism, Hematoma metabolism, CX3C Chemokine Receptor 1 metabolism, Chemokine CX3CL1 metabolism, Chemokine CX3CL1 pharmacology, Infant, Newborn, Diseases
- Abstract
Background: Hematoma clearance has been a proposed therapeutic strategy for hemorrhagic stroke. This study investigated the impact of CX3CR1 (CX3C chemokine receptor 1) activation mediated by r-FKN (recombinant fractalkine) on hematoma resolution, neuroinflammation, and the underlying mechanisms involving AMPK (AMP-activated protein kinase)/PPARγ (peroxisome proliferator-activated receptor gamma) pathway after experimental germinal matrix hemorrhage (GMH)., Methods: A total of 313 postnatal day 7 Sprague Dawley rat pups were used. GMH was induced using bacterial collagenase by a stereotactically guided infusion. r-FKN was administered intranasally at 1, 25, and 49 hours after GMH for short-term neurological evaluation. Long-term neurobehavioral tests (water maze, rotarod, and foot-fault test) were performed 24 to 28 days after GMH with the treatment of r-FKN once daily for 7 days. To elucidate the underlying mechanism, CX3CR1 CRISPR, or selective CX3CR1 inhibitor AZD8797, was administered intracerebroventricularly 24 hours preinduction of GMH. Selective inhibition of AMPK/PPARγ signaling in microglia via intracerebroventricularly delivery of liposome-encapsulated specific AMPK (Lipo-Dorsomorphin), PPARγ (Lipo-GW9662) inhibitor. Western blot, Immunofluorescence staining, Nissl staining, Hemoglobin assay, and ELISA assay were performed., Results: The brain expression of FKN and CX3CR1 were elevated after GMH. FKN was expressed on both neurons and microglia, whereas CX3CR1 was mainly expressed on microglia after GMH. Intranasal administration of r-FKN improved the short- and long-term neurobehavioral deficits and promoted M2 microglia polarization, thereby attenuating neuroinflammation and enhancing hematoma clearance, which was accompanied by an increased ratio of p-AMPK (phosphorylation of AMPK)/AMPK, Nrf2 (nuclear factor erythroid 2-related factor 2), PPARγ, CD36 (cluster of differentiation 36), CD163 (hemoglobin scavenger receptor), CD206 (the mannose receptor), and IL (interleukin)-10 expression, and decreased CD68 (cluster of differentiation 68), IL-1β, and TNF (tumor necrosis factor) α expression. The administration of CX3CR1 CRISPR or CX3CR1 inhibitor (AZD8797) abolished the protective effect of FKN. Furthermore, selective inhibition of microglial AMPK/PPARγ signaling abrogated the anti-inflammation effects of r-FKN after GMH., Conclusions: CX3CR1 activation by r-FKN promoted hematoma resolution, attenuated neuroinflammation, and neurological deficits partially through the AMPK/PPARγ signaling pathway, which promoted M1/M2 microglial polarization. Activating CX3CR1 by r-FKN may provide a promising therapeutic approach for treating patients with GMH., Competing Interests: Disclosures None.
- Published
- 2023
- Full Text
- View/download PDF
36. Recruitment of regulatory T cells with rCCL17 promotes M2 microglia/macrophage polarization through TGFβ/TGFβR/Smad2/3 pathway in a mouse model of intracerebral hemorrhage.
- Author
-
Deng S, Jin P, Liu S, He Y, Sherchan P, Zhang JH, Gong Y, and Tang J
- Subjects
- Mice, Male, Animals, Chemokines, CC metabolism, Chemokines, CC therapeutic use, T-Lymphocytes, Regulatory, Ligands, Macrophages metabolism, Cerebral Hemorrhage metabolism, Immunologic Factors, Disease Models, Animal, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta therapeutic use, Hematoma metabolism, Microglia metabolism, Brain Edema metabolism
- Abstract
Aims: Intracerebral hemorrhage (ICH) is a severe neurological condition with high mortality and morbidity. Microglia activation and peripheral inflammatory cells infiltration play an important role in ICH prognosis. Previous studies demonstrated that regulatory T cells (Tregs) ameliorated neuroinflammation following experimental ICH. However, the molecular mechanism underlying such effects of Tregs remains unclear. The objective was to examine how Tregs recruitment induced by recombinant CC chemokine ligand 17 (rCCL17) influences microglia/macrophage polarization in an intrastriatal autologous blood injection ICH animal model, and to determine if TGFβ/TGFβ-R/Smad2/3 pathway was involved., Methods: 380 adult CD1 mice (male, eight weeks old) were subjected to sham surgery or autologous blood injection induced ICH. A CD25-specific mouse antibody or isotype control mAb was injected intraventricular (i.c.v) 48 h prior to ICH induction to deplete Tregs. rCCL17, a CC chemokine receptor 4 (CCR4) ligand, was delivered intranasally at 1 h post-ICH. SB431542, a specific inhibitor of TGF-β was administered intraperitoneally 1 h before ICH induction. Following the ICH, neurobehavioral testing, brain edema, hematoma volume, hemoglobin content, western blotting, double immunofluorescence labeling, and immunohistochemistry were performed., Results: Endogenous expressions of CCL17, Tregs marker Foxp3, and the number of Tregs in perihematomal region increased following ICH. Tregs depletion with a CD25 antibody aggravated neurological deficits and brain edema, increased inflammatory cytokines, neutrophil infiltration, oxidative stress, and reduced the rate of hematoma resolution in ICH mice. rCCL17 treatment increased the number of Tregs in the brain, ameliorated neurological deficits and brain edema after ICH, and promoted microglia/macrophage polarization toward M2 phenotype which was reversed with CD25 antibody. Moreover, rCCL17 increased the expressions of brain TGF-β/phosphorylated-Smad2/3 which was abrogated with the selective TGFβ inhibitor SB431542., Conclusions: rCCL17-mediated Tregs recruitment may be a potential therapeutic strategy to promote M2 microglia/macrophages polarization and alleviate early brain injury following ICH., Competing Interests: Declaration of Competing Interest None., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
37. Inhibition of LAR attenuates neuroinflammation through RhoA/IRS-1/Akt signaling pathway after intracerebral hemorrhage in mice.
- Author
-
Le C, Hu X, Tong L, Ye X, Zhang J, Yan J, Sherchan P, Zhang JH, Gao F, and Tang J
- Subjects
- Mice, Animals, Neuroinflammatory Diseases, Signal Transduction, Cerebral Hemorrhage complications, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage metabolism, Proto-Oncogene Proteins c-akt metabolism, Brain Edema drug therapy
- Abstract
Leukocyte common antigen-related phosphatase (LAR) is widely expressed in the central nervous system and is known to regulate a variety of processes including cell growth, differentiation, and inflammation. However, little is currently known about LAR signaling mediated neuroinflammation after intracerebral hemorrhage (ICH). The objective of this study was to investigate the role of LAR in ICH using autologous blood injection-induced ICH mouse model. Expression of endogenous proteins, brain edema and neurological function after ICH were evaluated. Extracellular LAR peptide (ELP), an inhibitor of LAR, was administered to ICH mice and outcomes were evaluated. LAR activating-CRISPR or IRS inhibitor NT-157 was administered to elucidate the mechanism. The results showed that expressions of LAR, its endogenous agonist chondroitin sulfate proteoglycans (CSPGs) including neurocan and brevican, and downstream factor RhoA increased after ICH. Administration of ELP reduced brain edema, improved neurological function, and decreased microglia activation after ICH. ELP decreased RhoA and phosphorylated serine-IRS1, increased phosphorylated tyrosine-IRS1 and p-Akt, and attenuated neuroinflammation after ICH, which was reversed by LAR activating-CRISPR or NT-157. In conclusion, this study demonstrated that LAR contributed to neuroinflammation after ICH via RhoA/IRS-1 pathway, and ELP may be a potential therapeutic strategy to attenuate LAR mediated neuroinflammation after ICH.
- Published
- 2023
- Full Text
- View/download PDF
38. Endogenous Neural Stem Cell-induced Neurogenesis after Ischemic Stroke: Processes for Brain Repair and Perspectives.
- Author
-
Tang H, Li Y, Tang W, Zhu J, Parker GC, and Zhang JH
- Subjects
- Humans, Neurogenesis, Brain, Ischemic Stroke metabolism, Stroke therapy, Stroke metabolism, Neural Stem Cells, Brain Ischemia therapy, Brain Ischemia metabolism
- Abstract
Ischemic stroke is a very common cerebrovascular accident that occurred in adults and causes higher risk of neural deficits. After ischemic stroke, patients are often left with severe neurological deficits. Therapeutic strategies for ischemic stroke might mitigate neuronal loss due to delayed neural cell death in the penumbra or seek to replace dead neural cells in the ischemic core. Currently, stem cell therapy is the most promising approach for inducing neurogenesis for neural repair after ischemic stroke. Stem cell treatments include transplantation of exogenous stem cells but also stimulating endogenous neural stem cells (NSCs) proliferation and differentiation into neural cells. In this review, we will discuss endogenous NSCs-induced neurogenesis after ischemic stroke and provide perspectives for the therapeutic effects of endogenous NSCs in ischemic stroke. Our review would inform future therapeutic development not only for patients with ischemic stroke but also with other neurological deficits., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
39. Inhibition of lysophosphatidic acid receptor 1 relieves PMN recruitment in CNS via LPA1/TSP1/CXCR2 pathway and alleviates disruption on blood-brain barrier following intracerebral haemorrhage in mice.
- Author
-
Gao L, Peng L, Sherchan P, Tang H, Liu Y, Xiao J, Shi H, Luo Y, Tang J, Zhang JH, and Xia Y
- Subjects
- Mice, Animals, Receptors, Lysophosphatidic Acid metabolism, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage metabolism, Brain metabolism, Blood-Brain Barrier metabolism, Brain Edema metabolism
- Abstract
Backgroud: The frequencies of morbidity and impairment associated with spontaneous intracerebral haemorrhage (ICH) are comparatively high. Blood-brain barrier (BBB) integrity was compromised due to subsequent brain injury induced by ICH, which is crucial for a poor prognosis. Polymorphonuclear leukocyte (PMN) strongly modulate the disruption of BBB in the central nervous system (CNS). The lysophosphatidic acid receptor 1 (LPA1) mediated thrombospondin-1 (TSP1) regulation in astrocytes, which induce macrophage inflammatory protein 2(MIP2) secretion. MIP2 enhance PMN recruitment through CXC chemokine type 2 (CXCR2) activation. The purpose of this study was to investigate whether the LPA1-mediated inhibition of PMN recruitment and BBB protection after ICH is regulated by TSP1 and CXCR2 networks., Methods: ICH induction was performed in CD1 mice using collagenase administration. AM966, a targeted LPA1 antagonist, was orally administered 1 and 12 h following ICH. further identify possible LPA1-mediated BBB protection mechanisms, we intracerebroventricularly (ICV) administered a CXCR2 ligand MIP2, as well as TSP1 CRISPR activation (ACT) with AM966. Consequently, we performed neurobehavioral, brain water content (BWC), Evans blue staining (EBS), immunofluorescence (IF), and western blot (WB) analyses., Results: After ICH, astrocytes showed signs of LPA1, which peaked after 24 h, while PMN\ displayed evidence of CXCR2. The AM966-mediated LPA1 suppression relieved PMN recruitment, diminished brain oedema, demonstrated extravasation (as evidenced by EBS), protected BBB integrity, and enhanced neurologic activity following ICH. AM966 treatment strongly reduced TSP1, CXCR2, Occludin, and Claudin-5 expressions and PMN recruitment following ICH, and their expressions were restored by MIP2 and TSP1 CRISPR (ACT)., Conclusions: This study shows that LAP1 suppression reduced PMN recruitment after ICH in mice via TSP1/CXCR2 signalling, which minimized BBB disruption and improved the CNS's neurobehavioral functioning. Hence, LPA1 is a strong candidate for therapy to reduce PMN recruitment and offer protection of BBB integrity after ICH., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
40. Glial cells: an important switch for the vascular function of the central nervous system.
- Author
-
Gao L, Pan X, Zhang JH, and Xia Y
- Abstract
In this review, we first describe the current understanding of glial-mediated vascular function affecting the role of the blood-brain barrier (BBB) in central nervous system (CNS) disorders. BBB, mainly composed of glial and endothelial cells (ECs), is the protective structure that orchestrates the transport of substances, including ions, molecules, and cells from brain vessels into or out of the CNS. Then, we display the multiple communication between glial and vascular function based on angiogenesis, vascular wrapping, and blood perfusion in the brain. Glial can support microvascular ECs to form a blood network connecting to neurons. Astrocytes, microglia, and oligodendrocytes are the common types of glial surrounding the brain vessel. Glial-vessel interaction is required for the permeability and integrity of BBB. Glial cells surrounding the cerebral blood vessels can transmit communication signals to ECs and regulate the activity of vascular endothelial growth factor (VEGF) or Wnt-dependent endothelial angiogenesis mechanism. In addition, these glial cells monitor the blood flow in the brain via Ca
2+ /K+ -dependent pathways. Finally, we provide a potential research direction for the glial-vessel axis in CNS disorders. Microglial activation can trigger astrocyte activation, which suggests that microglia-astrocyte interaction may play a key role in monitoring cerebral blood flow. Thus, microglia-astrocyte interaction can be the key point of follow-up studies focusing on the microglia-blood mechanism. More investigations focus on the mechanism of how oligodendrocyte progenitor cells communicate and interact with ECs. The direct role of oligodendrocytes in modulating vascular function needs to be explored in the future., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Gao, Pan, Zhang and Xia.)- Published
- 2023
- Full Text
- View/download PDF
41. Discovery of a hydrogen molecular target.
- Author
-
Sun X, Ohta S, and Zhang JH
- Subjects
- Hydrogen
- Abstract
Competing Interests: None
- Published
- 2023
- Full Text
- View/download PDF
42. Correction to: Role of Estrogen-related Receptor γ and PGC-1α/SIRT3 Pathway in Early Brain Injury After Subarachnoid Hemorrhage.
- Author
-
Guo Y, Hu Y, Huang Y, Huang L, Kanamaru H, Takemoto Y, Li H, Li D, Gu J, and Zhang JH
- Published
- 2023
- Full Text
- View/download PDF
43. Three Days Delayed Recanalization Improved Neurological Function in pMCAO Rats by Increasing M2 Microglia-Possible Involvement of the IL-4R/STAT6/PPARγ Pathway.
- Author
-
Kang R, Gamdzyk M, Luo Y, Tang H, Huang L, Lenahan C, Doycheva D, Li D, Tang J, Tan S, and Zhang JH
- Subjects
- Rats, Animals, Infarction, Middle Cerebral Artery metabolism, Microglia metabolism, PPAR gamma, Ischemic Stroke metabolism, Stroke drug therapy, Brain Ischemia drug therapy
- Abstract
Current approved therapies for acute ischemic stroke have a restricted therapeutic time window. Delayed recanalization, which has been utilized clinically in patients who have missed the time window for administration, may be a promising alternative for stroke patients. However, the underlying molecular mechanisms remain undiscovered. Herein, we hypothesized that delayed recanalization would increase M2 microglial polarization through the IL-4R (interleukin-4 receptor)/STAT6 (signal transducer and activators of transcription 6)/PPARγ (peroxisome proliferator-activated receptor γ) pathway, subsequently promoting stroke recovery in rats. The permanent middle cerebral artery occlusion (pMCAO) model was induced via intravascular filament insertion. Recanalization was induced by withdrawing the filament at 3 days after MCAO (rMCAO). Interleukin (IL)-4 was administered intranasally at 3 days after pMCAO. AS1517499, a specific STAT6 inhibitor, was administered intranasally at 3 days after MCAO induction. Immunofluorescence staining, enzyme-linked immunosorbent assay (ELISA), western blot analysis, volumetric measurements of brain infarct, and neurological behavior tests were conducted. Delayed recanalization at 3 days after MCAO increased the polarization of M2 microglia, decreased inflammation, and improved neurological behavior. IL-4 treatment administered on the 3rd day after pMCAO increased M2 microglial polarization, improved neurological behavior, and reduced infarction volume of pMCAO rats. The inhibition of STAT6 decreased the level of p-STAT6 and PPARγ in rats treated with delayed recanalization. Delayed recanalization improved neurological function by increasing microglial M2 polarization, possibly involved with the IL-4R/STAT6/PPARγ pathway after MCAO in rats., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
44. Role of Estrogen-Related Receptor γ and PGC-1α/SIRT3 Pathway in Early Brain Injury After Subarachnoid Hemorrhage.
- Author
-
Guo Y, Hu Y, Huang Y, Huang L, Kanamaru H, Takemoto Y, Li H, Li D, Gu J, and Zhang JH
- Subjects
- Animals, Rats, Apoptosis, Estrogens, Rats, Sprague-Dawley, RNA, Small Interfering pharmacology, Humans, Brain Injuries drug therapy, Brain Injuries etiology, Brain Injuries metabolism, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use, Sirtuin 3 pharmacology, Sirtuin 3 therapeutic use, Subarachnoid Hemorrhage metabolism
- Abstract
Estrogen-related receptors (ERRs) were shown to play an important role in the regulation of free radical-mediated pathology. This study aimed to investigate the neuroprotective effect of ERRγ activation against early brain injury (EBI) after subarachnoid hemorrhage (SAH) and the potential underlying mechanisms. In a rat model of SAH, the time course of ERRs and SIRT3 and the effects of ERRγ activation were investigated. ERRγ agonist DY131, selective inhibitor GSK5182, or SIRT3 selective inhibitor 3-TYP were administered intracerebroventricularly (icv) in the rat model of SAH. The use of 3-TYP was for validating SIRT3 as the downstream signaling of ERRγ activation. Post-SAH assessments included SAH grade, neurological score, Western blot, Nissl staining, and immunofluorescence staining in rats. In an vitro study, the ERRγ agonist DY131 and ERRγ siRNA were administered to primary cortical neurons stimulated by Hb, after which cell viability and neuronal deaths were accessed. Lastly, the brain ERRγ levels and neuronal death were accessed in SAH patients. We found that brain ERRγ expressions were significantly increased, but the expression of SIRT3 dramatically decreased after SAH in rats. In the brains of SAH rats, ERRγ was expressed primarily in neurons, astrocytes, and microglia. The activation of ERRγ with DY131 significantly improved the short-term and long-term neurological deficits, accompanied by reductions in oxidative stress and neuronal apoptosis at 24 h after SAH in rats. DY131 treatment significantly increased the expressions of PGC-1α, SIRT3, and Bcl-2 while downregulating the expressions of 4-HNE and Bax. ERRγ antagonist GSK5182 and SIRT3 inhibitor 3-TYP abolished the neuroprotective effects of ERRγ activation in the SAH rats. An in vitro study showed that Hb stimulation significantly increased intracellular oxidative stress in primary cortical neurons, and DY131 reduced such elevations. Primary cortical neurons transfected with the ERRγ siRNA exhibited notable apoptosis and abolished the protective effect of DY131. The examination of SAH patients' brain samples revealed increases in ERRγ expressions and neuronal apoptosis marker CC3. We concluded that ERRγ activation with DY131 ameliorated oxidative stress and neuronal apoptosis after the experimental SAH. The effects were, at least in part, through the ERRγ/PGC-1α/SIRT3 signaling pathway. ERRγ may serve as a novel therapeutic target to ameliorate EBI after SAH., (© 2022. The American Society for Experimental Neurotherapeutics, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
45. Neuroprotection by Nrf2 via modulating microglial phenotype and phagocytosis after intracerebral hemorrhage.
- Author
-
Liang C, Liu L, Bao S, Yao Z, Bai Q, Fu P, Liu X, Zhang JH, and Wang G
- Abstract
Activated microglia are divided into pro-inflammatory and anti-inflammatory functional states. In anti-inflammatory state, activated microglia contribute to phagocytosis, neural repair and anti-inflammation. Nrf2 as a major endogenous regulator in hematoma clearance after intracerebral hemorrhage (ICH) has received much attention. This study aims to investigate the mechanism underlying Nrf2-mediated regulation of microglial phenotype and phagocytosis in hematoma clearance after ICH. In vitro experiments, BV-2 cells were assigned to normal group and administration group (Nrf2-siRNA, Nrf2 agonists Monascin and Xuezhikang). In vivo experiments, mice were divided into 5 groups: sham, ICH + vehicle, ICH + Nrf2-/-, ICH + Monascin and ICH + Xuezhikang. In vitro and in vivo, 72 h after administration of Monascin and Xuezhikang, the expression of Nrf2, inflammatory-associated factors such as Trem1, TNF-α and CD80, anti-inflammatory, neural repair and phagocytic associated factors such as Trem2, CD206 and BDNF were analyzed by the Western blot method. In vitro, fluorescent latex beads or erythrocytes were uptaken by BV-2 cells in order to study microglial phagocytic ability. In vivo, hemoglobin levels reflect the hematoma volume. In this study, Nrf2 agonists (Monascin and Xuezhikang) upregulated the expression of Trem2, CD206 and BDNF while decreased the expression of Trem1, TNF-α and CD80 both in vivo and in vitro. At the same time, after Monascin and Xuezhikang treatment, the phagocytic capacity of microglia increased in vitro, neurological deficits improved and hematoma volume lessened in vivo. These results were reversed in the Nrf2-siRNA or the Nrf2-/- mice. All these results indicated that Nrf2 enhanced hematoma clearance and neural repair, improved neurological outcomes through enhancing microglial phagocytosis and alleviating neuroinflammation., Competing Interests: The authors declare no conflict of interest., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
46. Delayed revascularization in acute ischemic stroke patients.
- Author
-
Eastin TM, Dye JA, Pillai P, Lopez-Gonzalez MA, Huang L, Zhang JH, and Boling WW
- Abstract
Stroke shares a significant burden of global mortality and disability. A significant decline in the quality of life is attributed to the so-called post-stroke cognitive impairment including mild to severe cognitive alterations, dementia, and functional disability. Currently, only two clinical interventions including pharmacological and mechanical thrombolysis are advised for successful revascularization of the occluded vessel. However, their therapeutic effect is limited to the acute phase of stroke onset only. This often results in the exclusion of a significant number of patients who are unable to reach within the therapeutic window. Advances in neuroimaging technologies have allowed better assessment of salvageable penumbra and occluded vessel status. Improvement in diagnostic tools and the advent of intravascular interventional devices such as stent retrievers have expanded the potential revascularization window. Clinical studies have demonstrated positive outcomes of delayed revascularization beyond the recommended therapeutic window. This review will discuss the current understanding of ischemic stroke, the latest revascularization doctrine, and evidence from clinical studies regarding effective delayed revascularization in ischemic stroke., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Eastin, Dye, Pillai, Lopez-Gonzalez, Huang, Zhang and Boling.)
- Published
- 2023
- Full Text
- View/download PDF
47. EphA4/EphrinB2 signaling mediates pericyte-induced transient glia limitans formation as a secondary protective barrier after subarachnoid hemorrhage in mice.
- Author
-
Zhou J, Guo P, Duan M, Li J, Ru X, Li L, Guo Z, Zhang JH, Feng H, Chen Y, and Sun X
- Subjects
- Animals, Male, Mice, Astrocytes, Blood-Brain Barrier, Ephrin-B2, Mice, Inbred C57BL, Pericytes, Receptor, EphA4 metabolism, Brain Edema, Brain Injuries, Subarachnoid Hemorrhage complications
- Abstract
Background: Most patients with subarachnoid hemorrhage (SAH) do not exhibit brain parenchymal injury upon imaging but present significant blood-brain barrier (BBB) disruption and secondary neurological deficits. The aim of this study was to investigate whether stressed astrocytes act as a secondary barrier to exert a protective effect after SAH and to investigate the mechanism of glial limitan formation., Methods: A total of 204 adult male C57BL/6 mice and an endovascular perforation SAH model were employed. The spatiotemporal characteristics of glial limitan formation after SAH were determined by immunofluorescence staining and transmission electron microscopy. The molecular mechanisms by which pericytes regulate glia limitans formation were analyzed using polymerase chain reaction, Western blotting, immunofluorescence staining and ELISA in a pericyte-astrocyte contact coculture system. The findings were validated ex vivo and in vivo using lentiviruses and inhibitors. Finally, pericytes were targeted to regulate glial limitan formation, and the effect of the glia limitans on secondary brain injury after SAH was evaluated by flow cytometry and analysis of neurological function., Results: Stress-induced glial limitan formation occurred 1 day after SAH and markedly subsided 3 days after ictus. Pericytes regulated astrocyte glia limitan formation via EphA4/EphrinB2 signaling, inhibited inflammatory cell infiltration and altered neurological function., Conclusions: Astrocyte-derived glia limitans serve as a secondary protective barrier following BBB disruption after SAH in mice, and pericytes can regulate glial limitan formation and alter neurological function via EphA4/EphrinB2 signaling. Strategies for maintaining this secondary protective barrier may be novel treatment approaches for alleviating early brain injury after SAH., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
48. Secukinumab attenuates neuroinflammation and neurobehavior defect via PKCβ/ERK/NF-κB pathway in a rat model of GMH.
- Author
-
Liu S, Deng S, Ding Y, Flores JJ, Zhang X, Jia X, Hu X, Peng J, Zuo G, Zhang JH, Gong Y, and Tang J
- Subjects
- Animals, Rats, Rats, Sprague-Dawley, Animals, Newborn, Cerebral Hemorrhage metabolism, NF-kappa B, Neuroinflammatory Diseases
- Abstract
Aims: Germinal matrix hemorrhage (GMH) is a disastrous clinical event for newborns. Neuroinflammation plays an important role in the development of neurological deficits after GMH. The purpose of this study is to investigate the anti-inflammatory role of secukinumab after GMH and its underlying mechanisms involving PKCβ/ERK/NF-κB signaling pathway., Methods: A total of 154 Sprague-Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of bacterial collagenase. Secukinumab was administered intranasally post-GMH. PKCβ activator PMA and p-ERK activator Ceramide C6 were administered intracerebroventricularly at 24 h prior to GMH induction, respectively. Neurobehavioral tests, western blot and immunohistochemistry were used to evaluate the efficacy of Secukinumab in both short-term and long-term studies., Results: Endogenous IL-17A, IL-17RA, PKCβ and p-ERK were increased after GMH. Secukinumab treatment improved short- and long-term neurological outcomes, reduced the synthesis of MPO and Iba-1 in the perihematoma area, and inhibited the synthesis of proinflammatory factors, such as NF-κB, IL-1β, TNF-α and IL-6. Additionally, PMA and ceramide C6 abolished the beneficial effects of Secukinumab., Conclusion: Secukinumab treatment suppressed neuroinflammation and attenuated neurological deficits after GMH, which was mediated through the downregulation of the PKCβ/ERK/NF-κB pathway. Secukinumab treatment may provide a promising therapeutic strategy for GMH patients., Competing Interests: Declaration of Competing Interest The authors declare that they have no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
49. Autophagy and Apoptosis in Acute Brain Injuries: From Mechanism to Treatment.
- Author
-
He C, Xu Y, Sun J, Li L, Zhang JH, and Wang Y
- Subjects
- Humans, Brain metabolism, Autophagy physiology, Apoptosis physiology, Brain Injuries therapy, Brain Injuries metabolism
- Abstract
Significance: Autophagy and apoptosis are two important cellular mechanisms behind brain injuries, which are severe clinical situations with increasing incidences worldwide. To search for more and better treatments for brain injuries, it is essential to deepen the understanding of autophagy, apoptosis, and their interactions in brain injuries. This article first analyzes how autophagy and apoptosis participate in the pathogenetic processes of brain injuries respectively and mutually, then summarizes some promising treatments targeting autophagy and apoptosis to show the potential clinical applications in personalized medicine and precision medicine in the future. Recent Advances: Most current studies suggest that apoptosis is detrimental to brain recovery. Several studies indicate that autophagy can cause unnecessary death of neurons after brain injuries, while others show that autophagy is beneficial for acute brain injuries (ABIs) by facilitating the removal of damaged proteins and organelles. Whether autophagy is beneficial or detrimental in ABIs depends on many factors, and the results from different research groups are diverse or even controversial, making this topic more appealing to be explored further. Critical Issues: Neuronal autophagy and apoptosis are two primary pathological processes in ABIs. How they interact with each other and how their regulations affect the outcome and prognosis of brain injuries remain uncertain, making these answers more critical. Future Directions: Insights into the interplay between autophagy and apoptosis and the accurate regulations of their balance in ABIs may promote personalized and precise treatments in the field of brain injuries. Antioxid. Redox Signal. 38, 234-257.
- Published
- 2023
- Full Text
- View/download PDF
50. Annexin A1 upregulates hematoma resolution via the FPR2/p-ERK(1/2)/DUSP1/CD36 signaling pathway after germinal matrix hemorrhage.
- Author
-
Flores JJ, Ding Y, Sherchan P, Zhang JH, and Tang J
- Subjects
- Animals, Humans, Infant, Newborn, Rats, CD36 Antigens genetics, Cerebral Hemorrhage complications, Dual Specificity Phosphatase 1 metabolism, Hematoma, Infant, Premature, Receptors, Lipoxin metabolism, Signal Transduction, Extracellular Signal-Regulated MAP Kinases, Annexin A1 genetics, Annexin A1 metabolism, Receptors, Formyl Peptide genetics, Receptors, Formyl Peptide metabolism
- Abstract
Germinal matrix hemorrhage (GMH) is one of the leading causes of morbidity and mortality in preterm infants in the United States, with little progress made in its clinical management. Blood clots disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage are key contributors towards post-hemorrhagic hydrocephalus development. n-formyl peptide receptor 2 (FPR2), a G-protein-coupled receptor, has been associated with the activation of p-ERK1/2, which in turn promotes the transcription of the DUSP1 gene, which may play a role in CD36 signaling. CD36 scavenger, a transmembrane glycoprotein, plays an essential role in microglia phagocytic blood clot clearance after GMH. FPR2's role in blood clot clearance after hemorrhagic stroke is unknown. We hypothesize that FPR2 activation by FPR2 agonist Annexin A1 (AnxA1) will enhance hematoma resolution via the upregulation of the CD36 signaling pathway, thereby improving short- and long-term neurological outcomes. Bacterial collagenase (0.3 U) was infused intraparenchymally into the right hemispheric ganglionic eminence in P7 rat pups to induce GMH. AnxA1 and FPR2 Inhibitor (Boc2) were given at 1-h post-GMH via intranasal administration. FPR2 CRISPR was given 48-h prior to GMH induction. Short-term neurological deficits were assessed using negative geotaxis test. Hematoma volume was assessed using hemoglobin assay. Protein expression was assessed using western blots. Long-term neurocognitive deficits and motor coordination were assessed using Morris water maze, rotarod, and foot fault tests. We have demonstrated that AnxA1 treatment enhances hematoma resolution and improved short and long-term outcomes. Lastly, FPR2 agonist AnxA1 treatment resulted in the upregulation of the FPR2/p-ERK(1/2)/DUSP1/CD36 signaling pathway., Competing Interests: Declaration of Competing Interest The authors of this manuscript declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.