Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.127
1.
Brain Behav ; 14(5): e3504, 2024 May.
Article En | MEDLINE | ID: mdl-38698583

BACKGROUND: Electroacupuncture (EA) has been shown to facilitate brain plasticity-related functional recovery following ischemic stroke. The functional magnetic resonance imaging technique can be used to determine the range and mode of brain activation. After stroke, EA has been shown to alter brain connectivity, whereas EA's effect on brain network topology properties remains unclear. An evaluation of EA's effects on global and nodal topological properties in rats with ischemia reperfusion was conducted in this study. METHODS AND RESULTS: There were three groups of adult male Sprague-Dawley rats: sham-operated group (sham group), middle cerebral artery occlusion/reperfusion (MCAO/R) group, and MCAO/R plus EA (MCAO/R + EA) group. The differences in global and nodal topological properties, including shortest path length, global efficiency, local efficiency, small-worldness index, betweenness centrality (BC), and degree centrality (DC) were estimated. Graphical network analyses revealed that, as compared with the sham group, the MCAO/R group demonstrated a decrease in BC value in the right ventral hippocampus and increased BC in the right substantia nigra, accompanied by increased DC in the left nucleus accumbens shell (AcbSh). The BC was increased in the right hippocampus ventral and decreased in the right substantia nigra after EA intervention, and MCAO/R + EA resulted in a decreased DC in left AcbSh compared to MCAO/R. CONCLUSION: The results of this study provide a potential basis for EA to promote cognitive and motor function recovery after ischemic stroke.


Electroacupuncture , Infarction, Middle Cerebral Artery , Magnetic Resonance Imaging , Rats, Sprague-Dawley , Reperfusion Injury , Animals , Electroacupuncture/methods , Male , Rats , Reperfusion Injury/physiopathology , Reperfusion Injury/therapy , Reperfusion Injury/diagnostic imaging , Infarction, Middle Cerebral Artery/therapy , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/diagnostic imaging , Brain/physiopathology , Brain/diagnostic imaging , Brain Ischemia/therapy , Brain Ischemia/physiopathology , Brain Ischemia/diagnostic imaging , Disease Models, Animal , Nerve Net/physiopathology , Nerve Net/diagnostic imaging , Ischemic Stroke/therapy , Ischemic Stroke/physiopathology , Ischemic Stroke/diagnostic imaging , Hippocampus/diagnostic imaging , Hippocampus/physiopathology
3.
Oxid Med Cell Longev ; 2022: 8123157, 2022.
Article En | MEDLINE | ID: mdl-35281468

Objectives: Although a recent study reported that stimulator of interferon genes (STING) in macrophages has an important regulatory effect on liver ischemia-reperfusion injury (IRI), the underlying mechanism of STING-dependent innate immune activation in liver macrophages (Kupffer cells, KCs) remains unclear. Here, we investigated the effect of STING on liver macrophage pyroptosis and the associated regulatory mechanism of liver IRI. Methods: Clodronate liposomes were used to block liver macrophages. AAV-STING-RNAi-F4/80-EGFP, an adenoassociated virus (AAV), was transfected into the portal vein of mice in vivo, and the liver IRI model was established 14 days later. In vitro, liver macrophages were treated with STING-specific siRNA, and a hypoxia-reoxygenation (H/R) model was established. The level of STING was detected via Western blotting (WB), RT-PCR, and immunostaining. Liver tissue and blood samples were collected. Pathological changes in liver tissue were detected by hematoxylin and eosin (H&E) staining. Macrophage pyroptosis was detected by WB, confocal laser scanning microscopy (CLSM), transmission electron microscopy (TEM), and enzyme-linked immunosorbent assay (ELISA). The calcium concentration was measured by immunofluorescence and analyzed with a fluorescence microplate reader. Results: The expression of STING increased with liver IRI but decreased significantly after the clodronate liposome blockade of liver macrophages. After knockdown of STING, the activation of caspase 1-GSDMD in macrophages and liver IRI was alleviated. More interestingly, hypoxia/reoxygenation (H/R) increased the calcium concentration in liver macrophages, but the calcium concentration was decreased after STING knockdown. Furthermore, after the inhibition of calcium in H/R-induced liver macrophages by BAPTA-AM, pyroptosis was significantly reduced, but the expression of STING was not significantlydecreased. Conclusions: Knockdown of STING reduces calcium-dependent macrophage caspase 1-GSDMD-mediated liver IRI, representing a potential therapeutic approach in the clinic.


Caspase 1/metabolism , Liver/pathology , Macrophages/metabolism , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Reperfusion Injury/physiopathology , Animals , Humans , Male , Mice , Signal Transduction , Transfection
4.
Hum Exp Toxicol ; 41: 9603271221078870, 2022.
Article En | MEDLINE | ID: mdl-35230166

AIM: To explore whether LPA5 was involved in the inflammatory responses in CI/R injury by regulation of NLRC4. METHOD: The cerebral I/R model in rats was constructed with ischemia of 2h and different time points of reperfusion. After that, western blot was used to determine protein expression (LPA5, NLRC4, AIM2, caspase-1, cleaved-caspase-1, mature IL-1ß, and precursor IL-1ß). And LPA5 and NLRC4 expression were also detected by using immunofluorescence experiment. Afterward, two sequence of LPA5-siRNA were injected into rats via intracerebroventricular administration. TTC staining and HE staining were performed. RESULT: As the reperfusion time was prolonged, LPA5 content was continuously increased, and the highest expression of NLRC4 was found at 4h of reperfusion. And protein expression of AIM2, cleaved-caspase-1, and mature IL-1ß was also at highest level at 4h. And after reperfusion of 4h, LPA5 siRNA1# or 2# was injected into lateral ventricles. LPA5 silence markedly reduced the infract volume and improved the histological change of ischemic zone. And LPA5 silence significantly downregulated NLRC4, AIM2, and the ratio of cleaved-caspase-1/caspase-1 and mature IL-1ß/precursor IL-1ß. And compared with LPA5-siRNA2#, LPA5-siRNA1# exerted a more significant effect. CONCLUSION: Low expression of LPA5 can protect against the inflammatory responses in CI/R model of rats through inhibiting NLRC4 inflammasomes.


Brain/drug effects , Inflammasomes/drug effects , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/therapeutic use , Animals , Brain/metabolism , Disease Models, Animal , Humans , Male , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
5.
Comput Math Methods Med ; 2022: 9275406, 2022.
Article En | MEDLINE | ID: mdl-35211189

BACKGROUND: Renal dysfunction after kidney transplantation may be influenced by many reasons. This study was designed to evaluate whether the administration of dexmedetomidine (Dex) could ameliorate renal function and prognosis after kidney transplantation. METHODS: A total of 65 patients were divided into Dex group (n = 33) and Con group (Con, n = 32). Dex group intravenously received an initial loading dose of 0.6 µg/kg Dex for 15 min before anaesthesia induction, followed by a rate of 0.4 µg/kg/h until 30 min after kidney reperfusion. By contrast, Con group received saline. The concentration of urinary kidney injury molecule-1 (KIM-1), serum creatinine (Cr), blood urea, urine output, ß2 microglobulin (ß2-MG), Cystatin C (CysC), and estimated glomerular filtration rate (eGFR) was recorded and compared between two groups during the course of the hospitalization or follow-up. Mean arterial pressure (MAP) and heart rate (HR), vasoactive drugs, and anaesthetics were recorded during the operation. Pain degree was evaluated using a visual analogue scale (VAS) after operation. Delayed graft function (DGF), graft loss, length of hospital stay, and mortality were compared between groups. RESULTS: The concentration of KIM-1 in Dex group was lower than Con group at 2 h (P = 0.018), 24 h (P = 0.013), 48 h (P < 0.01), and 72 h (P < 0.01) after reperfusion. MAP of Dex group after tracheal intubation (P = 0.012) and incision (P = 0.018) and HR after intubation (P = 0.021) were lower than that of Con group. The dosage of sufentanil during operation in Dex group was less than Con group (P = 0.039). Patients that used atropine in Dex group were more than Con group (P = 0.027). Patients who received Dex presented with lower VAS scores at 6 h (P = 0.01) and 12 h (P = 0.002) after operation. Concentration of serum Cr and blood urea had no significant differences between groups before operation and on postoperative day 1 to 6. Urine output was recorded for 6 days after operation and had no differences between groups. Also, no differences were identified between two groups in urea, Cr, ß2-MG, CysC, and eGFR in the first 3 months after operation. Incidence of DGF after operation was detected no difference between groups, while length of hospital stay in Dex group was less than Con group (P = 0.012). CONCLUSION: Dex can decrease kidney injury marker level, attenuate perioperative stress, relieve the dosage of sufentanil and postoperative pain, and reduce length of hospital stay. However, Dex is not associated with changes in prognosis in the first 3 months after transplantation.


Dexmedetomidine/administration & dosage , Kidney Transplantation/methods , Acute Kidney Injury/etiology , Acute Kidney Injury/physiopathology , Acute Kidney Injury/prevention & control , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Adult , Biomarkers/blood , Biomarkers/urine , Computational Biology , Female , Humans , Intraoperative Period , Kidney Failure, Chronic/physiopathology , Kidney Failure, Chronic/surgery , Kidney Function Tests , Kidney Transplantation/adverse effects , Male , Middle Aged , Postoperative Complications/etiology , Postoperative Complications/physiopathology , Postoperative Complications/prevention & control , Prognosis , Protective Agents/administration & dosage , Reperfusion Injury/etiology , Reperfusion Injury/physiopathology , Reperfusion Injury/prevention & control
6.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article En | MEDLINE | ID: mdl-35216473

Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.


Apoptosis , Liver Diseases/physiopathology , Mitochondria/physiology , Reperfusion Injury/physiopathology , Animals , Endoplasmic Reticulum Stress , Humans
7.
Nat Commun ; 13(1): 438, 2022 01 21.
Article En | MEDLINE | ID: mdl-35064106

Loss of Klotho, an anti-aging protein, plays a critical role in the pathogenesis of chronic kidney diseases. As Klotho is a large transmembrane protein, it is challenging to harness it as a therapeutic remedy. Here we report the discovery of a Klotho-derived peptide 1 (KP1) protecting kidneys by targeting TGF-ß signaling. By screening a series of peptides derived from human Klotho protein, we identified KP1 that repressed fibroblast activation by binding to TGF-ß receptor 2 (TßR2) and disrupting the TGF-ß/TßR2 engagement. As such, KP1 blocked TGF-ß-induced activation of Smad2/3 and mitogen-activated protein kinases. In mouse models of renal fibrosis, intravenous injection of KP1 resulted in its preferential accumulation in injured kidneys. KP1 preserved kidney function, repressed TGF-ß signaling, ameliorated renal fibrosis and restored endogenous Klotho expression. Together, our findings suggest that KP1 recapitulates the anti-fibrotic action of Klotho and offers a potential remedy in the fight against fibrotic kidney diseases.


Kidney/metabolism , Kidney/pathology , Klotho Proteins/chemistry , Peptides/pharmacology , Protective Agents/pharmacology , Signal Transduction , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Cell Line , Disease Models, Animal , Fibrosis , Humans , Inflammation/pathology , Kidney/injuries , Kidney/physiopathology , Kidney Diseases/complications , Kidney Diseases/pathology , Male , Mice, Inbred BALB C , Peptides/chemistry , Phosphorylation/drug effects , Protein Binding , Rats , Receptors, Transforming Growth Factor beta/metabolism , Reperfusion Injury/complications , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Smad Proteins/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
8.
Int J Med Sci ; 19(1): 13-24, 2022.
Article En | MEDLINE | ID: mdl-34975295

Retinal ischemia-reperfusion injury (RIRI) is of common occurrence in retinal and optic nerve diseases. The BDNF/TrkB signaling pathway has been examined to be neuroprotective in RIRI. In this study, we investigated the role of a potent selective TrkB agonist 7,8-dihydroxyfavone (DHF) in rat retinas with RIRI. Our results showed that RIRI inhibited the conversion of BDNF precursor (proBDNF) to mature BDNF (mBDNF) and increased the level of neuronal cell apoptosis. Compared with RIRI, DHF+RIRI reduced proBDNF level and at the same time increased mBDNF level. Moreover, DHF administration effectively activated TrkB signaling and and downstream Akt and Erk signaling pathways which increased nerve cell survival. The combined effects of mBDNF/proBDNF increase and TrkB signaling activation lead to reduction of apoptosis level and protection of retinas with RIRI. Moreover, it was also found that astrocytes labeled by GFAP were activated in RIRI and NF-kB mediated the increased expressions of inflammatory factors and these effects were partially reversed by DHF administration. Besides, we also used RNA sequencing to analyze the differently expressed genes (DEGs) and their enriched (Kyoto Encyclopedia of Genes and Genomes) KEGG pathways between Sham, RIRI, and DHF+RIRI. It was found that 1543 DEGs were differently expressed in RIRI and 619 DEGs were reversed in DHF+RIRI. The reversed DEGs were typically enriched in PI3K-Akt signaling pathway, Jak-STAT signaling pathway, NF-kB signaling pathway, and Apoptosis. To sum up, the DHF administration alleviated apoptosis and inflammation induced by RIRI via activating TrkB signaling pathway and may serve as a promising drug candidate for RIRI related ophthalmopathy.


Apoptosis/drug effects , Flavanones/pharmacology , Inflammation/prevention & control , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkB/metabolism , Reperfusion Injury/physiopathology , Retina/physiopathology , Animals , Disease Models, Animal , Male , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Signal Transduction
9.
Comput Math Methods Med ; 2022: 8202975, 2022.
Article En | MEDLINE | ID: mdl-35082916

OBJECTIVE: To investigate the influence of melatonin on behavioral and neurological function of rats with focal cerebral ischemia-reperfusion injury via the JNK/FoxO3a/Bim pathway. METHODS: One hundred and twenty healthy male SD rats were randomized into the model group (Model: the middle cerebral artery occlusion (MCAO) model was constructed and received an equal volume of normal saline containing 5% DMSO), sham operation group (Sham: received no treatment except normal feeding), and low, medium, and high dose of melatonin group (L-MT, M-MT, and H-MT intraperitoneally injected 10, 20, and 40 mg/kg melatonin 30 min after IR, respectively), with 24 rats in each group. Following 24 h of reperfusion, the rats in each of the above groups were tested for neurological deficit symptoms and behavioral changes to screen the rats included in the study. HE and TUNEL stainings were performed to observe pathological changes. Levels of oxidative stress-related indexes, inflammatory factor-related indexes, nuclear factor-κB p65 (NF-κB p65), and interferon-γ (IFN-γ) in the rat brain were measured by ELISA. The JNK/FoxO3a/Bim pathway-related proteins as well as Bcl-2, Caspase-3, and Bax were examined using Western blot. RESULTS: Detection of behavioral indicators showed that the MACO model was successfully constructed in rats. L-MT, M-MT, and L-MT groups presented reduced malondialdehyde (MDA), reactive oxygen species (ROS), tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, IL-1ß, IFN-γ, NF-κB p65, and apoptosis compared with the Model group (P < 0.05), and the improvement degree was better in the M-MT group versus the L-HT group. Bcl-2 protein expression in the brain tissue of L-MT, M-MT, and H-MT groups increased significantly, while Bax, Caspase-3, p-JNK, p-FoxO3a, and Bim protein expression declined markedly, versus the Model group (P < 0.05). The changes of indexes were greater in the M-MT group compared with that in the L-MT group. No significant difference was observed in all the above indexes between the M-MT group and the H-MT group (P > 0.05). CONCLUSIONS: In the MACO rat model, melatonin can effectively reduce Bax and Caspase-3 levels by modulating the JNK/FoxO3a/Bim pathway, inhibit neuronal apoptosis, and alleviate neurological deficits by reducing the release of proinflammatory mediators, with anti-inflammatory and antioxidant effects. In addition, 20 mg/kg is the optimal melatonin concentration.


Brain Ischemia/drug therapy , Melatonin/pharmacology , Reperfusion Injury/drug therapy , Animals , Bcl-2-Like Protein 11/metabolism , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Ischemia/physiopathology , Brain Ischemia/psychology , Computational Biology , Disease Models, Animal , Forkhead Box Protein O3 , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Male , Melatonin/administration & dosage , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Reperfusion Injury/psychology
10.
Int J Neurosci ; 132(12): 1254-1260, 2022 Dec.
Article En | MEDLINE | ID: mdl-33527868

PURPOSE: Spontaneous axonal plasticity and functional restoration after stroke may be limited by Nogo-A, a myelin-associated inhibitor, via activation of the Rho/Rho-associated protein kinase (ROCK) pathway. Constraint-induced movement therapy (CIMT) is a rehabilitation technique based on neuroplasticity and neural recombination. We recently reported that CIMT promoted neurogenesis after cerebral ischemia/reperfusion in part by inhibiting the Nogo-A-RhoA-ROCK pathway. Here, we examine the hypothesis that CIMT combined with the ROCK inhibitor fasudil further amplifies neurogenesis during stroke recovery. METHODS: Four groups of rats were randomized as follows: Cerebral ischemia-reperfusion (IR), Fasudil, CIMT and CIMT + Fasudil. Seven days after stroke, CIMT and/or intraperitoneal infusion of fasudil were initiated and continued for 3 weeks. The behavioral outcomes and immunohistochemical markers of neurogenesis were quantified. RESULTS: Compared with other groups, the combination of CIMT with fasudil after IR significantly improved motor and memory function recovery. In addition, BrdU, BrdU/doublecortin and BrdU/GFAP all increased significantly in the brain tissue of the combined treatment group compared to the CIMT or Fasudil group. CONCLUSION: These results suggest that the effects of CIMT on neurogenesis are amplified by fasudil during the recovery phase after stroke.


Brain Ischemia , Cerebral Infarction , Reperfusion Injury , Animals , Rats , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Bromodeoxyuridine , Cerebral Infarction/physiopathology , Cerebral Infarction/therapy , Neurogenesis/physiology , Nogo Proteins , Reperfusion Injury/physiopathology , Reperfusion Injury/therapy , Stroke/physiopathology , Stroke/therapy
12.
Life Sci ; 290: 120158, 2022 Feb 01.
Article En | MEDLINE | ID: mdl-34822798

Neutrophil extracellular trap (NETosis), the web-like structures induced by neutrophil death, is an important inflammatory mechanism of the immune system leading to reactive oxygen species production/coagulopathy, endothelial dysfunction, atherosclerosis, and ischemia. NETosis exerts its role through different mechanisms such as triggering Toll-like receptors, inflammatory cytokines, platelet aggregation, neutrophil activation/infiltration, and vascular impairment. NETosis plays a key role in the prognosis of coronary artery disease, ischemic injury of kidney, lung, gastrointestinal tract and skeletal muscles. In this review, we explored the molecular mechanisms involved in NETosis, and ischemic/reperfusion injuries in body organs.


Extracellular Traps/immunology , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Extracellular Traps/metabolism , Extracellular Traps/physiology , Humans , Neutrophils/immunology , Reperfusion Injury/physiopathology
13.
Br J Anaesth ; 128(3): 501-512, 2022 Mar.
Article En | MEDLINE | ID: mdl-34930601

BACKGROUND: Intestinal ischaemia/reperfusion (I/R) injury is a grave surgical event with high morbidity and mortality. Preoperative fasting might confer protection against intestinal I/R injury by altering the composition of gut microbiota and their respective metabolites. METHODS: An intestinal I/R mouse model was established and subjected to preoperative fasting for 24 h or fed ad libitum. Intestinal I/R injury was assessed using histological examination and survival analysis. Faecal samples were collected for 16S rDNA sequencing and metabolomic analysis. Faecal transplantation of fasted and non-fasted mice and humans was conducted to evaluate the effects of gut microbiota on intestinal I/R. Murine small intestinal cells wecre subjected to oxygen and glucose deprivation/reoxygenation as an in vitro I/R model. RESULTS: Preoperative fasting protected against intestinal I/R injury and improved survival in mice (P<0.001). In addition, 16S rDNA sequencing revealed that preoperative fasting increased the diversity and restructured the composition of the gut microbiota after intestinal I/R. Mice that received microbiota from fasted mice and humans showed less intestinal damage than those that received microbiota from fed subjects. Metabolomic analysis showed that the profiles of gut microbial metabolites differed between fasted and fed groups. Specifically, the concentration of petroselinic acid was significantly higher in the fasted group (P=0.009). Treatment of intestinal I/R mice with petroselinic acid alleviated intestinal injury in vivo and decreased cell apoptosis by mediating AMP-activated protein kinase-mammalian target of rapamycin-P70S6K signaling in vitro. CONCLUSIONS: Preoperative fasting protected against intestinal I/R injury by modulating gut microbiota and petroselinic acid, suggesting a novel therapeutic strategy.


Fasting/metabolism , Gastrointestinal Microbiome/physiology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Animals , Apoptosis/physiology , Disease Models, Animal , Glucose/metabolism , Mice , Oleic Acids/pharmacology , Oxygen/metabolism , Preoperative Period , Reperfusion Injury/drug therapy
14.
Neuropharmacology ; 207: 108935, 2022 04 01.
Article En | MEDLINE | ID: mdl-34968475

AIMS: Acetaminophen or paracetamol (PAR), the recommended antipyretic in COVID-19 and clinically used to alleviate stroke-associated hyperthermia interestingly activates cannabinoid receptor (CB1) through its AM404 metabolite, however, to date, no study reports the in vivo activation of PAR/AM404/CB1 axis in stroke. The current study deciphers the neuroprotective effect off PAR in cerebral ischemia/reperfusion (IR) rat model and unmasks its link with AM404/CB1/PI3K/Akt axis. MATERIALS AND METHODS: Animals were allocated into 5 groups: (I) sham-operated (SO), (II) IR, (III) IR + PAR (100 mg/kg), (IV) IR + PAR (100 mg/kg) + URB597; anandamide degradation inhibitor (0.3 mg/kg) and (V) IR + PAR (100 mg/kg) + AM4113; CB1 Blocker (5 mg/kg). All drugs were intraperitoneally administered at the inception of the reperfusion period. KEY FINDINGS: PAR administration alleviated the cognitive impairment in the Morris Water Maze as well as hippocampal histopathological and immunohistochemical examination of GFAP. The PAR signaling was associated with elevation of anandamide level, CB1 receptor expression and survival proteins as pS473-Akt. P(tyr202/thr204)-ERK1/2 and pS9-GSK3ß. Simultaneously, PAR increased hippocampal BDNF and ß-arrestin1 levels and decreased glutamate level. PAR restores the deranged redox milieu induced by IR Injury, by reducing lipid peroxides, myeloperoxidase activity and NF-κB and increasing NPSH, total antioxidant capacity, nitric oxide and Nrf2 levels. The pre-administration of AM4113 reversed PAR effects, while URB597 potentiated them. SIGNIFICANCE: PAR poses a significant neuroprotective effect which may be mediated, at least in part, via activation of anandamide/CB1/PI3K/Akt pathway in the IR rat model.


Acetaminophen/pharmacology , Antipyretics/pharmacology , Benzamides/pharmacology , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Receptor, Cannabinoid, CB1/metabolism , Reperfusion Injury/metabolism , Amidohydrolases/antagonists & inhibitors , Animals , Arachidonic Acids/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Endocannabinoids/metabolism , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Polyunsaturated Alkamides/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reperfusion Injury/physiopathology
15.
J Am Soc Nephrol ; 33(2): 279-289, 2022 02.
Article En | MEDLINE | ID: mdl-34853151

BACKGROUND: Single-cell sequencing technologies have advanced our understanding of kidney biology and disease, but the loss of spatial information in these datasets hinders our interpretation of intercellular communication networks and regional gene expression patterns. New spatial transcriptomic sequencing platforms make it possible to measure the topography of gene expression at genome depth. METHODS: We optimized and validated a female bilateral ischemia-reperfusion injury model. Using the 10× Genomics Visium Spatial Gene Expression solution, we generated spatial maps of gene expression across the injury and repair time course, and applied two open-source computational tools, Giotto and SPOTlight, to increase resolution and measure cell-cell interaction dynamics. RESULTS: An ischemia time of 34 minutes in a female murine model resulted in comparable injury to 22 minutes for males. We report a total of 16,856 unique genes mapped across our injury and repair time course. Giotto, a computational toolbox for spatial data analysis, enabled increased resolution mapping of genes and cell types. Using a seeded nonnegative matrix regression (SPOTlight) to deconvolute the dynamic landscape of cell-cell interactions, we found that injured proximal tubule cells were characterized by increasing macrophage and lymphocyte interactions even 6 weeks after injury, potentially reflecting the AKI to CKD transition. CONCLUSIONS: In this transcriptomic atlas, we defined region-specific and injury-induced loss of differentiation markers and their re-expression during repair, as well as region-specific injury and repair transcriptional responses. Lastly, we created an interactive data visualization application for the scientific community to explore these results (http://humphreyslab.com/SingleCell/).


Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Cell Communication/genetics , Disease Models, Animal , Female , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Mice , Mice, Inbred C57BL , RNA-Seq , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Single-Cell Analysis/methods , Single-Cell Analysis/statistics & numerical data , Software
16.
J Neurochem ; 160(3): 392-411, 2022 02.
Article En | MEDLINE | ID: mdl-34837397

TBC1Domain Family Member 25 (TBC1D25) is a protein that contains a TBC/RAB-GTPase activating protein (GAP) domain, which was shown to participate in autophagy in previous studies. However, the role of TBC1D25 in cerebral ischemia-reperfusion (I/R) injury remains unknown. In this study, we found that the mRNA and protein expression levels of TBC1D25 decreased in mouse brain after I/R injury and primary cortical neurons treated with oxygen and glucose deprivation/reoxygenation (OGD/R). Then TBC1D25 knockout (KO) mice were applied to demonstrate that TBC1D25 ablation aggravated cerebral I/R-induced neuronal loss and infarct size. In addition, neuronal apoptosis and inflammation were significantly potentiated in the TBC1D25-KO group. In in vitro OGD/R model, TBC1D25 knockdown can attenuate neuronal cell viability and aggravate the process of inflammation and apoptosis. Conversely, over-expression of TBC1D25 in primary neurons ameliorated the aforementioned processes. Mechanistically, RNA-sequencing (RNA-seq) analysis revealed mitogen-activated protein kinase (MAPK) signaling pathway was the most significant pathway that contributed to TBC1D25-mediated brain I/R injury process. Through experimental verification, TBC1D25 deficiency increased the phosphorylation of the transforming growth factor-ß-activated kinase 1 (TAK1)-c-Jun N-terminal kinase (JNK)/p38 axis in neurons during the brain I/R injury. Furthermore, we found that TAK1 blockade abrogated the apoptosis and inflammatory response produced by TBC1D25 knockdown in vitro. In conclusion, this study is the first to demonstrate the functional significance of TBC1D25 in the pathophysiology of brain I/R injury, and the protective mechanism of TBC1D25 is dependent on the TAK1-JNK/p38 pathway.


Brain Ischemia/genetics , GTPase-Activating Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase Kinases/genetics , Reperfusion Injury/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis , Brain Ischemia/physiopathology , GTPase-Activating Proteins/deficiency , Glucose/deficiency , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/genetics , Inflammation/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA-Seq , Reperfusion Injury/physiopathology
17.
Front Immunol ; 12: 785229, 2021.
Article En | MEDLINE | ID: mdl-34899752

The complement system has long been recognized as a potential druggable target for a variety of inflammatory conditions. Very few complement inhibitors have been approved for clinical use, but a great number are in clinical development, nearly all of which systemically inhibit complement. There are benefits of targeting complement inhibition to sites of activation/disease in terms of efficacy and safety, and here we describe P-selectin targeted complement inhibitors, with and without a dual function of directly blocking P-selectin-mediated cell-adhesion. The constructs are characterized in vitro and in murine models of hindlimb ischemia/reperfusion injury and hindlimb transplantation. Both constructs specifically targeted to reperfused hindlimb and provided protection in the hindlimb ischemia/reperfusion injury model. The P-selectin blocking construct was the more efficacious, which correlated with less myeloid cell infiltration, but with similarly reduced levels of complement deposition. The blocking construct also improved tissue perfusion and, unlike the nonblocking construct, inhibited coagulation, raising the possibility of differential application of each construct, such as in thrombotic vs. hemorrhagic conditions. Similar outcomes were obtained with the blocking construct following vascularized composite graft transplantation, and treatment also significantly increased graft survival. This is outcome may be particularly pertinent in the context of vascularized composite allograft transplantation, since reduced ischemia reperfusion injury is linked to a less rigorous alloimmune response that may translate to the requirement of a less aggressive immunosuppressive regime for this normally nonlife-threatening procedure. In summary, we describe a new generation of targeted complement inhibitor with multi-functionality that includes targeting to vascular injury, P-selectin blockade, complement inhibition and anti-thrombotic activity. The constructs described also bound to both mouse and human P-selectin which may facilitate potential translation.


Cell Adhesion/drug effects , Complement Activation/drug effects , Complement Inactivating Agents/administration & dosage , Hindlimb/blood supply , Hindlimb/transplantation , P-Selectin/antagonists & inhibitors , Receptors, Complement 3b/administration & dosage , Reperfusion Injury/prevention & control , Single-Chain Antibodies/administration & dosage , Vascularized Composite Allotransplantation , Animals , Complement Inactivating Agents/pharmacokinetics , Disease Models, Animal , Fibrinolytic Agents/administration & dosage , Graft Survival/drug effects , Male , Mice, Inbred C57BL , P-Selectin/metabolism , Regional Blood Flow , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Signal Transduction , Single-Chain Antibodies/pharmacokinetics
18.
Int J Mol Sci ; 22(24)2021 Dec 13.
Article En | MEDLINE | ID: mdl-34948185

Testicular ischemia reperfusion injury (tIRI) causes oxidative stress-induced DNA damage leading to germ cell apoptosis (GCA). The aim of the study is to establish a direct link between JAK2 activation and the DNA damage response (DDR) signaling pathways and their role in tIRI-induced GCA using AG490, a JAK2 specific inhibitor. Male Sprague Dawley rats (n = 36) were divided into three groups: sham, unilateral tIRI and tIRI + AG490 (40 mg/kg). During tIRI, augmentation in the phosphorylation levels of the JAK2/STAT1/STAT3 was measured by immunohistochemistry. Observed spermatogenic arrest was explained by the presence of considerable levels of DSB, AP sites and 8OHdG and activation of caspase 9, caspase 3 and PARP, which were measured by colorimetric assays and TUNEL. The ATM/Chk2/H2AX and ATR/Chk1 pathways were also activated as judged by their increased phosphorylation using Western blot. These observations were all prevented by AG490 inhibition of JAK2 activity. Our findings demonstrate that JAK2 regulates tIRI-induced GCA, oxidative DNA damage and activation of the ATM/Chk2/H2AX and ATR/Chk1 DDR pathways, but the cell made the apoptosis decision despite DDR efforts.


DNA Repair/physiology , Janus Kinase 2/metabolism , Reperfusion Injury/metabolism , Animals , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Caspase 3 , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/metabolism , DNA Damage/physiology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/physiology , Janus Kinase Inhibitors/pharmacology , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , STAT1 Transcription Factor , STAT3 Transcription Factor , Spermatogenesis , Testis/metabolism , Testis/physiology , Tyrphostins/pharmacology
19.
Bioengineered ; 12(2): 12917-12930, 2021 12.
Article En | MEDLINE | ID: mdl-34852714

Ischemia injury can cause cell death or impairment of neuron and astrocytes, and thus induce loss of nerve function. central nervous systems injury induces an aberrant activation of astrocytes called astrogliosis. Pyroptosis, which is a kind of programmed cell death, was proved play an important role in ischemia injury. Zinc Finger E-Box Binding Homeobox 2 (ZEB2) promoted neuron astrogliosis, which play a protected role in neuron regeneration. However, its precise mechanism remains unclear. This study investigated the mechanism of ZEB2 on astrogliosis and neuron regeneration after cerebral ischemia reperfusion condition. To confirm our hypothesis, Neurons and astrocytes were isolated from fetal Sprague Dawley rats, in vivo Middle Cerebral Artery Occlusion/reperfusion (MCAO/R) rat model and in vitro oxygen-glucose deprivation/reperfusion (OGD/R)-treated astrocytes and neurocytes model were constructed. Our results showed that ZEB2 was expressed in nucleus of astrocyte and upregulated after OGD/R induction, ZEB2 promoted astrogliosis. Further upregulation of ZEB2 promoted the astrogliosis, which promoted neuron proliferation and regeneration by decreased pyroptosis. Moreover, this finding was further confirmed in vivo MCAO/R rat model. Overexpression of ZEB2 promoted astrogliosis, which decreased infarct volume and accumulated recovery of neurological function by alleviated pyroptosis. This finding revealed that ZEB2 was a regulator of the astrogliosis after ischemia/reperfusion injury, and then astrogliosis promoted neuron regeneration via decreased neuron pyroptosis. Therefore, ZEB2 may be a potential therapeutic target for ischemia/reperfusion injury.


Brain Ischemia/pathology , Gliosis/metabolism , Gliosis/pathology , Neuroprotection , Pyroptosis , Reperfusion Injury/pathology , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/pathology , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Gliosis/complications , Gliosis/physiopathology , Glucose/deficiency , Nerve Regeneration , Oxygen , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology
20.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article En | MEDLINE | ID: mdl-34768879

Oxidative stress plays an important role in the pathophysiology of acute kidney injury (AKI). Previously, we reported that vanin-1, which is involved in oxidative stress, is associated with renal tubular injury. This study was aimed to determine whether urinary vanin-1 is a biomarker for the early diagnosis of AKI in two experimental models: in vivo and in vitro. In a rat model of AKI, ischemic AKI was induced in uninephrectomized rats by clamping the left renal artery for 45 min and then reperfusing the kidney. On Day 1 after renal ischemia/reperfusion (I/R), serum creatinine (SCr) in I/R rats was higher than in sham-operated rats, but this did not reach significance. Urinary N-acetyl-ß-D-glucosaminidase (NAG) exhibited a significant increase but decreased on Day 2 in I/R rats. In contrast, urinary vanin-1 significantly increased on Day 1 and remained at a significant high level on Day 2 in I/R rats. Renal vanin-1 protein decreased on Days 1 and 3. In line with these findings, immunofluorescence staining demonstrated that vanin-1 was attenuated in the renal proximal tubules of I/R rats. Our in vitro results confirmed that the supernatant from HK-2 cells under hypoxia/reoxygenation included significantly higher levels of vanin-1 as well as KIM-1 and NGAL. In conclusion, our results suggest that urinary vanin-1 might be a potential novel biomarker of AKI induced by I/R.


Acute Kidney Injury/metabolism , Amidohydrolases/metabolism , Reperfusion Injury/metabolism , Acute Kidney Injury/physiopathology , Acute Kidney Injury/urine , Amidohydrolases/urine , Animals , Biomarkers/urine , Creatinine/analysis , Creatinine/blood , Early Diagnosis , Hexosaminidases/metabolism , Hexosaminidases/urine , Ischemia/metabolism , Kidney/metabolism , Male , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reperfusion , Reperfusion Injury/physiopathology , Reperfusion Injury/urine , Urinary Tract/metabolism
...