Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 470
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell Biochem ; 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38294731

ABSTRACT

Subdural hematoma (SDH) drains into the extracranial lymphatic system through the meningeal lymphatic vessels (mLVs) but the formation of SDH impairs mLVs. Because vitamin D (Vit D) can protect the endothelial cells, we hypothesized that Vit D may enhance the SDH clearance. SDH was induced in Sprague-Dawley rats and treated with Vit D or vehicle. Hematoma volume in each group was measured by H&E staining and hemoglobin quantification. Evans blue (EB) quantification and red blood cells injection were used to evaluated the drainage of mLVs. Western blot analysis and immunofluorescence were conducted to assess the expression of lymphatic protein markers. We also examined the inflammatory factors levels in subdural space by ELISA. Vit D treatment significantly reduced SDH volume and improved the drainage of SDH to cervical lymph nodes. The structure of mLVs in SDH rats were protected by Vit D, and the expressions of LYVE1, PROX1, FOXC2, and VE-cadherin were increased after Vit D treatment. The TNF-α, IL-6, and IL-8 levels were reduced in Vit D group. In vitro, Vit D also increased the VE-cadherin expression levels under inflammation. Vit D protects the structure of mLVs and enhances the absorption of SDH, partly by the anti-inflammatory effect of Vit D.

2.
Environ Toxicol ; 39(2): 562-571, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37449671

ABSTRACT

BACKGROUND: Glioma, a type of malignant brain tumor, has become a challenging health issue globally in recent years. METHODS: In this study, we investigated the potential therapeutic role of scoparone in glioma and the underlying mechanism. Initially, transcriptome sequencing was conducted to identify genes that exhibited differential expression in glioma cells treated with scoparone compared to untreated cells. Subsequently, the impact of scoparone on the proliferation, migration, and invasion of glioma cells was assessed in vitro using a range of assays including cell viability, colony formation, wound healing, and transwell assays. Moreover, the apoptotic effects of scoparone on glioma cells were evaluated through flow cytometry and western blot analysis. Furthermore, we established a glioma xenograft mouse model to assess the in vivo antitumor activity of scoparone. Lastly, by integrating transcriptome analysis, we endeavored to unravel the molecular mechanisms underlying the observed antitumor effects of scoparone by examining the expression levels of RhoA/ROCK1 signaling pathway components using western blot analysis and qRT-PCR. RESULTS: Our transcriptome sequencing results revealed that scoparone significantly downregulated RhoA/ROCK1 signaling in glioma cells. Furthermore, scoparone treatment inhibited glioma cell proliferation, migration, and invasion, and promoted cell apoptosis in vitro. Moreover, scoparone reduced tumor growth and prolonged survival in a glioma xenograft mouse model, and improved the toxicity of temozolomide. Finally, our results showed that the antitumor effects of scoparone were mediated by the suppression of RhoA/ROCK1 signaling. CONCLUSION: Scoparone could be a promising therapeutic agent for glioma by suppressing RhoA/ROCK1 signaling. These findings pave the way for future research endeavors aimed at the development and optimization of scoparone-based therapeutic strategies.


Subject(s)
Glioma , Signal Transduction , Animals , Humans , Mice , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Glioma/genetics , rho-Associated Kinases/metabolism , Temozolomide/pharmacology , Temozolomide/therapeutic use , Coumarins/pharmacology , Coumarins/therapeutic use
3.
J Craniofac Surg ; 35(4): 1209-1213, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38709059

ABSTRACT

INTRODUCTION: Primary central nervous system lymphoma (PCNSL) presents a diagnostic enigma due to the inherent absence of lymphoid tissue in the central nervous system (CNS). The hypothesis posits that lymphocytes infiltrating the CNS during inflammatory responses could represent a cellular source for PCNSL, challenging traditional understandings of its etiology. PATIENT CONCERNS: In 2 illustrative cases, patients presented with neurological symptoms initially misdiagnosed as encephalitis and demyelinating disease, respectively. These diagnoses were established based on clinical assessments and initial biopsy findings. DIAGNOSIS: Subsequent biopsies, conducted months after the first signs of disease, confirmed the diagnosis of PCNSL in both patients. Identifying CD20-positive tumor cells was pivotal, indicating a B-cell lymphoma origin. INTERVENTIONS: Treatment strategies included high-dose methotrexate chemotherapy for both patients. In addition, the second patient underwent adjuvant whole-brain radiotherapy after the chemotherapy regimen. OUTCOMES: The therapeutic approach significantly reduced tumor size in both cases, with no evidence of recurrence observed during the follow-up period. This outcome underscores the potential efficacy of the chosen interventions. CONCLUSION: In response to inflammatory lesions, lymphocyte infiltration into the CNS may serve as a pivotal origin for tumor cells in PCNSL. These cases highlight the complexity of diagnosing CNS disorders and suggest that various forms of encephalitis in the early stages could influence the prognosis of lymphoma. This insight into the cellular origins and treatment responses of PCNSL contributes to a broader understanding of its pathophysiology and management.


Subject(s)
Central Nervous System Neoplasms , Methotrexate , Humans , Male , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/diagnosis , Female , Middle Aged , Methotrexate/therapeutic use , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/diagnosis , Aged , Diagnosis, Differential , Biopsy , Encephalitis/pathology , Encephalitis/diagnosis , Magnetic Resonance Imaging
4.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928496

ABSTRACT

The tumor microenvironment (TME) is crucial in tumor development, metastasis, and response to immunotherapy. DNA methylation can regulate the TME without altering the DNA sequence. However, research on the methylation-driven TME in clear-cell renal cell carcinoma (ccRCC) is still lacking. In this study, integrated DNA methylation and RNA-seq data were used to explore methylation-driven genes (MDGs). Immune scores were calculated using the ESTIMATE, which was employed to identify TME-related genes. A new signature connected with methylation-regulated TME using univariate, multivariate Cox regression and LASSO regression analyses was developed. This signature consists of four TME-MDGs, including AJAP1, HOXB9, MYH14, and SLC6A19, which exhibit high methylation and low expression in tumors. Validation was performed using qRT-PCR which confirmed their downregulation in ccRCC clinical samples. Additionally, the signature demonstrated stable predictive performance in different subtypes of ccRCC. Risk scores are positively correlated with TMN stages, immune cell infiltration, tumor mutation burden, and adverse outcomes of immunotherapy. Interestingly, the expression of four TME-MDGs are highly correlated with the sensitivity of first-line drugs in ccRCC treatment, especially pazopanib. Molecular docking indicates a high affinity binding between the proteins and pazopanib. In summary, our study elucidates the comprehensive role of methylation-driven TME in ccRCC, aiding in identifying patients sensitive to immunotherapy and targeted therapy, and providing new therapeutic targets for ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell , DNA Methylation , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Tumor Microenvironment , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Humans , Tumor Microenvironment/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Pyrimidines/therapeutic use , Indazoles/therapeutic use , Indazoles/pharmacology , Sulfonamides/therapeutic use , Sulfonamides/pharmacology , Biomarkers, Tumor/genetics , Female , Molecular Docking Simulation , Gene Expression Profiling/methods , Male
5.
J Proteome Res ; 22(11): 3640-3651, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37851947

ABSTRACT

Inflammation plays an important role in the development of sepsis-acute respiratory distress syndrome (ARDS). Olink inflammation-related biomarker panels were used to analyze the levels of 92 inflammation-related proteins in plasma with sepsis-ARDS (n = 25) and healthy subjects (n = 25). There were significant differences in 64 inflammatory factors, including TNFRSF11B in sepsis-ARDS, which was significantly higher than that in controls. Functional analysis showed that TNFRSF11B was closely focused on signal transduction, immune response, and inflammatory response. The TNFRSF11B level in sepsis-ARDS plasma, LPS-induced mice, and LPS-stimulated HUVECs significantly increased. The highest plasma concentration of TNFRSF11B in patients with sepsis-ARDS was 10-20 ng/mL, and 10 ng/mL was selected to stimulate HUVECs. Western blot results demonstrated that the levels of syndecan-1, claudin-5, VE-cadherin, occludin, aquaporin-1, and caveolin-1 in TNFRSF11B-stimulated HUVECs decreased, whereas that of connexin-43 increased in TNFRSF11B-stimulated HUVECs. To the best of the authors' knowledge, this study was the first to reveal elevated TNFRSF11B in sepsis-ARDS associated with vascular endothelial dysfunction. In summary, TNFRSF11B may be a new potential predictive and diagnostic biomarker for vascular endothelium damage in sepsis-ARDS.


Subject(s)
Osteoprotegerin , Respiratory Distress Syndrome , Sepsis , Vascular Diseases , Animals , Humans , Mice , Biomarkers/blood , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Osteoprotegerin/blood , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/diagnosis , Sepsis/blood , Sepsis/complications , Sepsis/diagnosis , Vascular Diseases/blood , Vascular Diseases/complications , Vascular Diseases/diagnosis
6.
Stroke ; 54(8): 2087-2095, 2023 08.
Article in English | MEDLINE | ID: mdl-37306018

ABSTRACT

BACKGROUND: The current understanding of untreated ruptured intracranial aneurysms has been limited by study design and inaccurate patient data. Multicenter clinical registry studies on untreated ruptured intracranial aneurysms in Chinese patients are scarce. We aimed to calculate the mortality of patients with untreated ruptured intracranial aneurysms in a current, clearly defined hospital cohort in China, with emphasis on mortality predictors over a 2-year period. METHODS: Patients with saccular untreated ruptured intracranial aneurysms were identified from the Chinese Multicenter Cerebral Aneurysm Database, a multicenter, prospective, observational database registered in China, which involved 32 tertiary medical centers covering 4 northern Chinese provincial regions. Patients with intracranial aneurysms, regardless of ruptured status, shape, age, or comorbidities, were consecutively included in 12 of 32 medical centers between 2017 and 2020. Survival probabilities were computed using the Kaplan-Meier method. Univariate and multivariate Cox regression analyses were conducted to determine the risk factors for the cumulative 2-year mortality. We analyzed the reasons for treatment decisions stratified by demographic characteristics and clinical features. RESULTS: For 941 enrolled patients, 58.6% of patients died within 1 month of symptom onset; and 68.1% within 2 years. 98 patients underwent surgical repair during follow-up. Multivariate Cox regression analysis identified Hunt and Hess grades 3 to 5 (hazard ratio, 1.54 [95% CI, 1.01-2.35]; P=0.047), loss of consciousness at symptom onset (hazard ratio, 1.56 [95% CI, 1.18-2.07]; P=0.002), and largest aneurysm size of ≥5 mm (hazard ratio, 1.29 [95% CI, 1.05-1.59]; P=0.014) as mortality predictors during the 2-year follow-up. Among patients who were successfully followed up, 42.6% (280) of them refused surgical treatment. CONCLUSIONS: Patients with poor Hunt and Hess grades, loss of consciousness at symptom onset, or largest aneurysms ≥5 mm in size showed a high mortality rate. A high number of treatment refusals was present in this study. These findings have implications for medical insurance policy, doctor-patient communication, and popular science education.


Subject(s)
Aneurysm, Ruptured , Embolization, Therapeutic , Intracranial Aneurysm , Humans , Intracranial Aneurysm/therapy , Follow-Up Studies , Prospective Studies , Treatment Outcome , Aneurysm, Ruptured/surgery , Risk Factors , Unconsciousness , Retrospective Studies
7.
Biochem Biophys Res Commun ; 671: 205-214, 2023 09 03.
Article in English | MEDLINE | ID: mdl-37302296

ABSTRACT

Sodium-glucose cotransporter 2 inhibitors (SGLT2i) provide added vascular protection beyond glucose lowering to patients with type 2 diabetes mellitus (T2DM). Endothelial progenitor cells (EPCs) are an important endogenous repair mechanism for diabetic vascular complications. Yet, whether SGLT2i protect vessels in diabetic patients by improving the function of EPCs remains to be elucidated. Here we enrolled Sixty-three T2DM patients and 60 healthy participants and 15 of T2DM group took dapagliflozin for 3 months. Retinal capillary density (RCD) was examined before and after meditation. Moreover, vasculogenic capacity of EPCs cocultured with or without dapagliflozin in vitro and in vivo (hind limb ischemia model) were assessed. Mechanically, genes related to inflammation/oxidative stress, and the AMPK signaling of EPCs were determined. Our results found T2DM demonstrated a declined RCD and a decreased number of circulating EPCs compared with healthy controls. Compared with the EPCs from healthy individuals, vasculogenic capacity of T2DM EPCs was significantly impaired, which could be restored by dapagliflozin meditation or dapagliflozin coculture. Increased expression of inflammation correlative genes and decreased anti-oxidative stress related genes expression were found in EPCs form T2DM, which were accompanied with reduced phosphorylation level of AMPK. Dapagliflozin treatment activated AMPK signaling, decreased the level of inflammation and oxidative stress, and rescued vasculogenic capacity of EPCs from T2DM. Furthermore, AMPK inhibitor pretreatment diminished the enhancement vasculogenic capacity of diabetic EPCs from dapagliflozin treatment. This study demonstrates for the first time that dapagliflozin restores vasculogenic capacity of EPCs via activating AMPK-mediated inhibition of inflammation and oxidative stress in T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Endothelial Progenitor Cells , Humans , Endothelial Progenitor Cells/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , AMP-Activated Protein Kinases/metabolism , Oxidative Stress , Glucose/metabolism
8.
Biochem Biophys Res Commun ; 682: 46-55, 2023 11 19.
Article in English | MEDLINE | ID: mdl-37801989

ABSTRACT

Previous studies have proved that cardiac dysfunction and myocardial damage can be found in TBI patients, but the underlying mechanisms of myocardial damage induced by TBI can't be illustrated. We want to investigate the function of ferroptosis in myocardial damage after TBI and determine if inhibiting iron overload might lessen myocardial injury after TBI due to the involvement of iron overload in the process of ferroptosis and inflammation. We detect the expression of ferroptosis-related proteins in cardiac tissue at different time points after TBI, indicating that TBI can cause ferroptosis in the heart in vivo. The echocardiography and myocardial enzymes results showed that ferroptosis can aggravate TBI-induced cardiac dysfunction. The result of DHE staining and 4-HNE expression showed that inhibition of ferroptosis can reduce ROS production and lipid peroxidation in myocardial tissue. In further experiments, DFO intervention was used to explore the effect of iron overload inhibition on myocardial ferroptosis after TBI, the production of ROS, expression of p38 MAPK and NF-κB was detected to explore the effect of iron overload on myocardial inflammation after TBI. The results above show that TBI can cause heart ferroptosis in vivo. Inhibition of iron overload can alleviate myocardial injury after TBI by reducing ferroptosis and inflammatory response induced by TBI.


Subject(s)
Ferroptosis , Heart Injuries , Iron Overload , Humans , Reactive Oxygen Species , Arrhythmias, Cardiac , Inflammation , Iron Overload/complications
9.
Biochem Biophys Res Commun ; 665: 141-151, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37163934

ABSTRACT

Traumatic brain injury (TBI) can negatively impact systemic organs, which can lead to more death and disability. However, the mechanism underlying the effect of TBI on systemic organs remains unclear. In previous work, we found that brain-derived extracellular vesicles (BDEVs) released from the injured brain can induce systemic coagulation with a widespread fibrin deposition in the microvasculature of the lungs, kidney, and heart in a mouse model of TBI. In this study, we investigated whether BDEVs can induce heart, lung, liver, and kidney injury in TBI mice. The results of pathological staining and related biomarkers indicated that BDEVs can induce histological damage and systematic dysfunction. In vivo imaging system demonstrated that BDEVs can gather in systemic organs. We also found that BDEVs could induce cell apoptosis in the lung, liver, heart, and kidney. Furthermore, we discovered that BDEVs could cause multi-organ endothelial cell damage. Finally, this secondary multi-organ damage could be relieved by removing circulating BDEVs. Our research provides a novel perspective and potential mechanism of TBI-associated multi-organ damage.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Extracellular Vesicles , Mice , Animals , Brain/pathology , Brain Injuries/pathology , Apoptosis , Extracellular Vesicles/pathology
10.
Biochem Biophys Res Commun ; 688: 149065, 2023 12 25.
Article in English | MEDLINE | ID: mdl-37979398

ABSTRACT

Intestinal injury caused by traumatic brain injury (TBI) seriously affects patient prognosis; however, the underlying mechanisms are unknown. Recent studies have demonstrated that ferritinophagy-mediated ferroptosis is involved in several intestinal disorders. However, uncertainty persists regarding the role of ferritinophagy-mediated ferroptosis in the intestinal damage caused by TBI. High-throughput transcriptional sequencing was used to identify the genes that were differentially expressed in the intestine after TBI. The intestinal tissues were harvested for hematoxylin and eosin staining (HE), immunofluorescence, and western blot (WB). Lipid peroxide markers and iron content in the intestines were determined using the corresponding kits. High throughput sequencing revealed that the ferroptosis signaling pathway was enriched, demonstrating that intestinal damage caused by TBI may include ferroptosis. Chiu's score, tight junction proteins, and lipid peroxide indicators demonstrated that TBI caused an intestinal mucosal injury that persisted for several days. The ferroptosis pathway-related proteins, ferritin heavy polypeptide 1 (Fth1) and glutathione peroxidase 4 (GPX4), exhibited dynamic changes. The results indicated that lipid peroxide products were markedly increased, whereas antioxidant enzymes were markedly decreased. WB analysis demonstrated that the expression levels of nuclear receptor coactivator 4 (NCOA4), LC3II/LC3I, and p62 were markedly upregulated, whereas those of GPX4 and Fth1 were markedly downregulated. In addition, ferrostatin-1 attenuates intestinal ferroptosis and injury post-TBI in vivo. Intriguingly, 3-methyladenine (3-MA) reduces intestinal ferritin decomposition, iron accumulation, and ferroptosis after TBI. Moreover, 3-MA markedly reduced intestinal apoptosis. In conclusion, NCOA4 mediated ferritinophagy and ferroptosis play roles in intestinal oxidative stress injury post-TBI. This study provides a deeper understanding of the mechanisms underlying intestinal damage following TBI.


Subject(s)
Brain Injuries, Traumatic , Ferroptosis , Humans , Lipid Peroxides , Intestines , Oxidative Stress , Transcription Factors , Ferritins , Iron , Autophagy , Nuclear Receptor Coactivators/genetics
11.
BMC Med ; 21(1): 199, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37254196

ABSTRACT

BACKGROUND: Contact sports athletes and military personnel who suffered a repetitive mild traumatic brain injury (rmTBI) are at high risk of neurodegenerative diseases such as advanced dementia and chronic traumatic encephalopathy (CTE). However, due to the lack of specific biological indicators in clinical practice, the diagnosis and treatment of rmTBI are quite limited. METHODS: We used 2-methacryloyloxyethyl phosphorylcholine (MPC)-nanocapsules to deliver immunoglobulins (IgG), which can increase the delivery efficiency and specific target of IgG while reducing the effective therapeutic dose of the drug. RESULTS: Our results demonstrated that MPC-capsuled immunoglobulins (MPC-n (IgG)) significantly alleviated cognitive impairment, hippocampal atrophy, p-Tau deposition, and myelin injury in rmTBI mice compared with free IgG. Furthermore, MPC-n (IgG) can also effectively inhibit the activation of microglia and the release of inflammatory factors. CONCLUSIONS: In the present study, we put forward an efficient strategy for the treatment of rmTBI-related cognitive impairment and provide evidence for the administration of low-dose IgG.


Subject(s)
Brain Concussion , Cognitive Dysfunction , Neurodegenerative Diseases , Mice , Animals , Brain Concussion/complications , Brain Concussion/drug therapy , Brain Concussion/psychology , Disease Models, Animal , Cognitive Dysfunction/drug therapy , Immunoglobulin G , Brain
12.
J Neuroinflammation ; 20(1): 222, 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37777772

ABSTRACT

BACKGROUND: Neuroinflammation is one of the most important pathogeneses in secondary brain injury after traumatic brain injury (TBI). Neutrophil extracellular traps (NETs) forming neutrophils were found throughout the brain tissue of TBI patients and elevated plasma NET biomarkers correlated with worse outcomes. However, the biological function and underlying mechanisms of NETs in TBI-induced neural damage are not yet fully understood. Here, we used Cl-amidine, a selective inhibitor of NETs to investigate the role of NETs in neural damage after TBI. METHODS: Controlled cortical impact model was performed to establish TBI. Cl-amidine, 2'3'-cGAMP (an activator of stimulating Interferon genes (STING)), C-176 (a selective STING inhibitor), and Kira6 [a selectively phosphorylated inositol-requiring enzyme-1 alpha [IRE1α] inhibitor] were administrated to explore the mechanism by which NETs promote neuroinflammation and neuronal apoptosis after TBI. Peptidyl arginine deiminase 4 (PAD4), an essential enzyme for neutrophil extracellular trap formation, is overexpressed with adenoviruses in the cortex of mice 1 day before TBI. The short-term neurobehavior tests, magnetic resonance imaging (MRI), laser speckle contrast imaging (LSCI), Evans blue extravasation assay, Fluoro-Jade C (FJC), TUNEL, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative-PCR were performed in this study. RESULTS: Neutrophils form NETs presenting in the circulation and brain at 3 days after TBI. NETs inhibitor Cl-amidine treatment improved short-term neurological functions, reduced cerebral lesion volume, reduced brain edema, and restored cerebral blood flow (CBF) after TBI. In addition, Cl-amidine exerted neuroprotective effects by attenuating BBB disruption, inhibiting immune cell infiltration, and alleviating neuronal death after TBI. Moreover, Cl-amidine treatment inhibited microglia/macrophage pro-inflammatory polarization and promoted anti-inflammatory polarization at 3 days after TBI. Mechanistically, STING ligand 2'3'-cGAMP abolished the neuroprotection of Cl-amidine via IRE1α/ASK1/JNK signaling pathway after TBI. Importantly, overexpression of PAD4 promotes neuroinflammation and neuronal death via the IRE1α/ASK1/JNK signaling pathway after TBI. However, STING inhibitor C-176 or IRE1α inhibitor Kira6 effectively abolished the neurodestructive effects of PAD4 overexpression after TBI. CONCLUSION: Altogether, we are the first to demonstrate that NETs inhibition with Cl-amidine ameliorated neuroinflammation, neuronal apoptosis, and neurological deficits via STING-dependent IRE1α/ASK1/JNK signaling pathway after TBI. Thus, Cl-amidine treatment may provide a promising therapeutic approach for the early management of TBI.


Subject(s)
Brain Injuries, Traumatic , Extracellular Traps , Humans , Mice , Animals , MAP Kinase Signaling System , Interferon-alpha/metabolism , Neuroinflammatory Diseases , Endoribonucleases , Disease Models, Animal , Protein Serine-Threonine Kinases/metabolism , Brain Injuries, Traumatic/pathology , Apoptosis , Mice, Inbred C57BL
13.
Blood ; 137(4): 544-555, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33507292

ABSTRACT

Traumatic brain injury-induced coagulopathy (TBI-IC) causes life-threatening secondary intracranial bleeding. Its pathogenesis differs mechanistically from that of coagulopathy arising from extracranial injuries and hemorrhagic shock, but it remains poorly understood. We report results of a study designed to test the hypothesis that von Willebrand factor (VWF) released during acute TBI is intrinsically hyperadhesive because its platelet-binding A1-domain is exposed and contributes to TBI-induced vascular leakage and consumptive coagulopathy. This hyperadhesive VWF can be selectively blocked by a VWF A2-domain protein to prevent TBI-IC and to improve neurological function with a minimal risk of bleeding. We demonstrated that A2 given through intraperitoneal injection or IV infusion reduced TBI-induced death by >50% and significantly improved the neurological function of C57BL/6J male mice subjected to severe lateral fluid percussion injury. A2 protected the endothelium from extracellular vesicle-induced injury, reducing TBI-induced platelet activation and microvesiculation, and preventing a TBI-induced hypercoagulable state. A2 achieved this therapeutic efficacy by specifically blocking the A1 domain exposed on the hyperadhesive VWF released during acute TBI. These results suggest that VWF plays a causal role in the development of TBI-IC and is a therapeutic target for this life-threatening complication of TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Peptide Fragments/pharmacology , von Willebrand Factor/antagonists & inhibitors , Acute-Phase Reaction , Animals , Blood Platelets/metabolism , Brain Injuries, Traumatic/complications , Capillary Leak Syndrome/etiology , Capillary Leak Syndrome/prevention & control , Case-Control Studies , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/prevention & control , Cerebrovascular Circulation , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/prevention & control , Endothelium, Vascular/drug effects , Extracellular Vesicles , Humans , Infusions, Intravenous , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Platelet Activation/drug effects , Protein Conformation , Protein Domains/drug effects , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , von Willebrand Factor/chemistry , von Willebrand Factor/physiology , von Willebrand Factor/therapeutic use
14.
Blood ; 138(25): 2714-2726, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34610086

ABSTRACT

Severe traumatic brain injury (TBI) often causes an acute systemic hypercoagulable state that rapidly develops into consumptive coagulopathy. We have recently demonstrated that TBI-induced coagulopathy (TBI-IC) is initiated and disseminated by brain-derived extracellular vesicles (BDEVs) and propagated by extracellular vesicles (EVs) from endothelial cells and platelets. Here, we present results from a study designed to test the hypothesis that anticoagulation targeting anionic phospholipid-expressing EVs prevents TBI-IC and improves the outcomes of mice subjected to severe TBI. We evaluated the effects of a fusion protein (ANV-6L15) for improving the outcomes of TBI in mouse models combined with in vitro experiments. ANV-6L15 combines the phosphatidylserine (PS)-binding annexin V (ANV) with a peptide anticoagulant modified to preferentially target extrinsic coagulation. We found that ANV-6L15 reduced intracranial hematoma by 70.2%, improved neurological function, and reduced death by 56.8% in mice subjected to fluid percussion injury at 1.9 atm. It protected the TBI mice by preventing vascular leakage, tissue edema, and the TBI-induced hypercoagulable state. We further showed that the extrinsic tenase complex was formed on the surfaces of circulating EVs, with the highest level found on BDEVs. The phospholipidomic analysis detected the highest levels of PS on BDEVs, as compared with EVs from endothelial cells and platelets (79.1, 15.2, and 3.5 nM/mg of protein, respectively). These findings demonstrate that TBI-IC results from a trauma-induced hypercoagulable state and may be treated by anticoagulation targeting on the anionic phospholipid-expressing membrane of EVs from the brain and other cells.


Subject(s)
Annexin A5/therapeutic use , Anticoagulants/therapeutic use , Brain Injuries, Traumatic/drug therapy , Extracellular Vesicles/drug effects , Phospholipids/metabolism , Recombinant Fusion Proteins/therapeutic use , Thrombophilia/drug therapy , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Male , Mice, Inbred C57BL , Thrombophilia/etiology , Thrombophilia/metabolism , Thrombophilia/pathology
15.
Respir Res ; 24(1): 182, 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37434162

ABSTRACT

Alveolar epithelial barrier is a potential therapeutic target for acute respiratory distress syndrome (ARDS). However, an effective intervention against alveolar epithelial barrier has not been developed. Here, based on single-cell RNA and mRNA sequencing results, death receptor 3 (DR3) and its only known ligand tumor necrosis factor ligand-associated molecule 1A (TL1A) were significantly reduced in epithelium from an ARDS mice and cell models. The apparent reduction in the TL1A/DR3 axis in lungs from septic-ARDS patients was correlated with the severity of the disease. The examination of knockout (KO) and alveolar epithelium conditional KO (CKO) mice showed that TL1A deficiency exacerbated alveolar inflammation and permeability in lipopolysaccharide (LPS)-induced ARDS. Mechanistically, TL1A deficiency decreased glycocalyx syndecan-1 and tight junction-associated zonula occludens 3 by increasing cathepsin E level for strengthening cell-to-cell permeability. Additionally, DR3 deletion aggravated barrier dysfunction and pulmonary edema in LPS-induced ARDS through the above mechanisms based on the analyses of DR3 CKO mice and DR3 overexpression cells. Therefore, the TL1A/DR3 axis has a potential value as a key therapeutic signaling for the protection of alveolar epithelial barrier.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 25 , Respiratory Distress Syndrome , Tumor Necrosis Factor Ligand Superfamily Member 15 , Animals , Mice , Epithelium , Ligands , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/genetics , Tumor Necrosis Factor-alpha , Receptors, Tumor Necrosis Factor, Member 25/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
16.
J Magn Reson Imaging ; 58(3): 732-738, 2023 09.
Article in English | MEDLINE | ID: mdl-36594577

ABSTRACT

BACKGROUND: Glioma genotypes are of importance for clinical decision-making. This data can only be acquired through histopathological analysis based on resection or biopsy. Consequently, there is a need for alternative biomarkers that noninvasively provide reliable information for preoperatively identifying molecular characteristics. PURPOSE: To investigate apparent diffusion coefficient (ADC) as imaging biomarker for preoperatively identifying glioma genotypes based on the 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors. STUDY TYPE: Retrospective. SUBJECTS: One hundred and fifty-nine patients (47.6 ± 14.4 years) diagnosed with WHO grade 2-4 glioma including 93 males and 66 females. FIELD STRENGTH/SEQUENCE: A 3 T/spin echo echo planner imaging. ASSESSMENT: The ADC measurements were assessed by two neuroradiologists (both with 6 years of experience). Three different lowest portions inside the tumors without overlap were manually drawn on the ADC maps as regions of interest (ROIs). The mean ADC value of the three ROIs was defined as the minimum ADC value (ADCmin ). An ROI was placed in the contralateral normal appearing white matter (NAWM) to obtain the ADC value (ADCNAWM ). The ADCmin to ADCNAWM ratio (ADCratio ) was calculated. Genetics results were retrospectively recorded from pathologic and genetic test reports. STATISTICAL TESTS: Two-sample independent t-tests, receiver operating characteristic curve analysis, and intraclass correlation coefficient analysis were used. Statistical significance was set at P < 0.05. RESULTS: Isocitrate dehydrogenase (IDH)-mutated glioma showed higher ADCmin and ADCratio than IDH wild-type glioma. Among IDH-mutated glioma, higher ADCmin and ADCratio were found in 1p19q intact glioma than in 1p19q codeletion glioma. ADC parameters enabled differentiation of IDH mutation status with area under the curve (AUC) of 0.84 and 0.86. DATA CONCLUSION: ADC has potential discriminative value for IDH mutation and 1p19q codeletion status. EVIDENCE LEVEL: 3. TECHNICAL EFFICACY: Stage 2.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Humans , Glioma/diagnostic imaging , Glioma/genetics , Male , Female , Adult , Middle Aged , Isocitrate Dehydrogenase/genetics , Mutation , Promoter Regions, Genetic , Biomarkers, Tumor , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Diffusion Magnetic Resonance Imaging , Retrospective Studies , Aged
17.
Cell Commun Signal ; 21(1): 211, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596642

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of injury-related disability and death around the world, but the clinical stratification, diagnosis, and treatment of complex TBI are limited. Due to their unique properties, extracellular vesicles (EVs) are emerging candidates for being biomarkers of traumatic brain injury as well as serving as potential therapeutic targets. However, the effects of different extracellular vesicle subtypes on the pathophysiology of traumatic brain injury are very different, or potentially even opposite. Before extracellular vesicles can be used as targets for TBI therapy, it is necessary to classify different extracellular vesicle subtypes according to their functions to clarify different strategies for EV-based TBI therapy. The purpose of this review is to discuss contradictory effects of different EV subtypes on TBI, and to propose treatment ideas based on different EV subtypes to maximize their benefits for the recovery of TBI patients. Video Abstract.


Subject(s)
Brain Injuries, Traumatic , Extracellular Vesicles , Humans , Brain Injuries, Traumatic/therapy
18.
Pharmacol Res ; 192: 106791, 2023 06.
Article in English | MEDLINE | ID: mdl-37156450

ABSTRACT

Endothelial dysfunction is a key proponent of pathophysiological process of traumatic brain injury (TBI). We previously demonstrated that extracellular vesicles (EVs) released from injured brains led to endothelial barrier disruption and vascular leakage. However, the molecular mechanisms of this EV-induced endothelial dysfunction (endotheliopathy) remain unclear. Here, we enriched plasma EVs from TBI patients (TEVs), and detected high mobility group box 1 (HMGB1) exposure to 50.33 ± 10.17% of TEVs and the number of HMGB1+TEVs correlated with injury severity. We then investigated for the first time the impact of TEVs on endothelial function using adoptive transfer models. We found that TEVs induced dysfunction of cultured human umbilical vein endothelial cells and mediated endothelial dysfunction in both normal and TBI mice, which were propagated through the HMGB1-activated receptor for advanced glycation end products (RAGE)/Cathepsin B signaling, and the resultant NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and canonical caspase-1/gasdermin D (GSDMD)-dependent pyroptosis. Finally, von Willebrand factor (VWF) was detected on the surface of 77.01 ± 7.51% of HMGB1+TEVs. The TEV-mediated endotheliopathy was reversed by a polyclonal VWF antibody, indicating that VWF might serve a coupling factor that tethered TEVs to ECs, thus facilitating HMGB1-induced endotheliopathy. These results suggest that circulating EVs isolated from patients with TBI alone are sufficient to induce endothelial dysfunction and contribute to secondary brain injury that are dependent on immunologically active HMGB1 exposed on their surface. This finding provided new insight for the development of potential therapeutic targets and diagnostic biomarkers for TBI.


Subject(s)
Brain Injuries, Traumatic , Extracellular Vesicles , HMGB1 Protein , Vascular Diseases , Humans , Mice , Animals , von Willebrand Factor , Brain Injuries, Traumatic/complications , Human Umbilical Vein Endothelial Cells
19.
Inorg Chem ; 62(25): 10014-10020, 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37307143

ABSTRACT

The high-temperature durability of SiBCN ceramics is significantly influenced by the boron concentration and synthesis methods. Although single-source synthetic routes allow one to obtain homogeneous ceramics at the atomic level, the content of the boron element therein is limited by borane (BH3). In this study, the synthesis of carborane-substituted polyborosilazanes was performed via a simple one-pot reaction of polysilazanes with alkyne bonds on the main chain and decaborododecahydrodiacetonitrile complexes with different molar ratios. This enabled one to vary the boron content from ∼0 to 40.00 wt %. The ceramic yields were in the range of 50.92-90.81 wt %. Independently of the borane concentration, SiBCN ceramics started to crystallize at 1200 °C, and B4C appeared as a new crystalline phase with increasing boron content. The introduction of boron inhibited the crystallization of Si3N4 and increased the crystallization temperature of SiC. The presence of the B4C phase improved both thermal stability and functional properties such as neutron-shielding characteristics of the ceramics. Therefore, this research opens up new prospects for the design of novel polyborosilanzes with great application potential.

20.
J Integr Neurosci ; 22(1): 6, 2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36722226

ABSTRACT

BACKGROUND: Deviation of electrode array from the midline of spinal cords affects the therapeutic outcomes of C2-4 cervical spinal cord stimulation (SCS) in patients with disorders of consciousness (DOC). This study proposed the implementation of a novel C2-3 dural exposure procedure and investigated its efficacy compared to conventional surgery. METHODS: Surgical and postoperative imaging data from 69 patients with DOC who underwent SCS in the lateral decubitus position were retrospectively assessed. The C2-3 dural exposure procedure was performed in 16 patients while the rest underwent conventional surgery. The incidence of electrode deviation was compared, and factors associated with the deviation were investigated. RESULTS: The rate of complete midline coverage by the electrodes in the C2-3 dural exposure group was significantly higher than the conventional group (93.8% vs. 54.7%, p = 0.004). Exposure of the dura between C2-3 was a significant favorable factor for complete midline coverage by the electrode array (odds ratio [OR]: 0.091; 95% confidence interval [CI]: 0.011-0.757; p = 0.027). Electrode positioned ≥5 cm above the lower edge of the C2 vertebra was a significant risk factor for incomplete midline coverage (OR: 1.126; 95% CI: 1.016-1.248; p = 0.023). No difference in operation time, intraoperative bleeding, or surgical site infection was observed between the 2 groups. CONCLUSIONS: The C2-3 dural exposure procedure, performed in the lateral decubitus position, was safe and had higher complete midline coverage than conventional surgery.


Subject(s)
Cervical Cord , Humans , Retrospective Studies , Consciousness Disorders/therapy , Spinal Cord , Electrodes
SELECTION OF CITATIONS
SEARCH DETAIL