Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Neurosurg Rev ; 47(1): 198, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722430

ABSTRACT

Achieving a pear-shaped balloon holds pivotal significance in the context of successful percutaneous microcompression procedures for trigeminal neuralgia. However, inflated balloons may assume various configurations, whether it is inserted into Meckel's cave or not. The absence of an objective evaluation metric has become apparent. To investigate the relationship between the morphology of Meckel's Cave and the balloon used in percutaneous microcompression for trigeminal neuralgia and establish objective criteria for assessing balloon shape in percutaneous microcompression procedures. This retrospective study included 58 consecutive patients with primary trigeminal neuralgia. Data included demographic, clinical outcomes, and morphological features of Meckel's cave and the balloon obtained from MRI and Dyna-CT imaging. MRI of Meckel's cave and Dyna-CT of intraoperative balloon were modeled, and the morphological characteristics and correlation were analyzed. The reconstructed balloon presented a fuller morphology expanding outward and upward on the basis of Meckel's cave. The projected area of balloon was strongly positively correlated with the projected area of Meckel's cave. The Pearson correlation coefficients were 0.812 (P<0.001) for axial view, 0.898 (P<0.001) for sagittal view and 0.813 (P<0.001) for coronal view. Similarity analysis showed that the sagittal projection image of Meckel's cave and that of the balloon had good similarity. This study reveals that the balloon in percutaneous microcompression essentially represents an expanded morphology of Meckel's cave, extending outward and upward. There is a strong positive correlation between the volume and projected area of the balloon and that of Meckel's cave. Notably, the sagittal projection image of Meckel's cave serves as a reliable predictor of the intraoperative balloon shape. This method has a certain generalizability and can help providing objective criteria for judging balloon shape during percutaneous microcompression procedures.


Subject(s)
Magnetic Resonance Imaging , Trigeminal Neuralgia , Humans , Female , Male , Middle Aged , Aged , Retrospective Studies , Trigeminal Neuralgia/surgery , Trigeminal Neuralgia/diagnostic imaging , Magnetic Resonance Imaging/methods , Adult , Tomography, X-Ray Computed/methods , Neurosurgical Procedures/methods , Treatment Outcome , Aged, 80 and over
2.
Acta Neurol Belg ; 123(4): 1321-1329, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37020131

ABSTRACT

OBJECTIVE: The abnormal expression of matrix metalloproteinase 9 (MMP9) and Aquaporin 4 (AQP4) closely associates with the traumatic brain injury (TBI) development. METHODS: Here, we investigated the relationship between miR-211-5p and MMP9/AQP4 axis in TBI patients and astrocyte cells. Demographics, clinical features, and cerebrospinal fluid (CSF) samples were collected from traumatic brain injury (TBI) patients (n = 96) and controls (n = 30) for pathological and gene expression analyses. Luciferase activity assay and gene expression analyses were performed to dissect the regulatory mechanism of miR-211-5p on MMP9/AQP4 in human astrocyte cells. RESULTS: miR-211-5p mRNA was significantly decreased in the CSF of TBI patients, which positively correlated with the expression of both MMP9 and AQP4. miR-211-5p could target MMP9 directly in SVG P12 cells. Overexpression of miR-211-5p decreased the expression of MMP9, on the contrary, knockdown miR-211-5p through inhibitors increased the expression of both MMP9 and AQP4. CONCLUSION: miR-211-5p inhibits the MMP9/AQP4 axis in human astrocyte cells, which represents a promising approach for the TBI treatment.


Subject(s)
Brain Injuries, Traumatic , MicroRNAs , Humans , Aquaporin 4/genetics , Brain Injuries, Traumatic/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Int J Neurosci ; 133(6): 604-611, 2023 Jun.
Article in English | MEDLINE | ID: mdl-34219583

ABSTRACT

BACKGROUND: Aquaporin 4 (AQP4), usually expressed at astrocytes end-feet, is a main component of the lymph-lymphatic system and promotes paravascular cerebrospinal fluid-interstitial fluid exchange. Moreover, angiotensin II type 1 (AT1) receptor affects amyloid ß (Aß) levels. This study aimed to detect the effect of AT1 receptor deficiency on the blood-brain barrier (BBB) of traumatic brain injury (TBI) mice and the effect on Aß level and glial lymphatic circulation. METHODS: TBI model was built using AT1 receptor knockout mice (AT1-KO) and C57BL/6 mice (wild type, WT). BBB integrity was detected by Evans blue extravasation. The expression of the astrocytic water channel AQP4 and astrocyte activation were evaluated with immunofluorescence. The expressions of amyloid precursor protein (APP), junction protein zonula occludens protein-1 (ZO-1) and occludin in mice brain were detected by Western blot (WB). Aß levels were assayed by enzyme-linked immunosorbent assay (ELISA). RESULTS: AT1 receptor deficiency defended BBB integrity and rescued occludin and ZO-1 decrease in mice brain induced by TBI. AT1-KO mice had less increase of APP expression and Aß 1-42, Aß 1-40 levels compared to WT mice under TBI. Moreover, AT1 receptor deficiency was found to significantly inhibit AQP4 depolarization after TBI. CONCLUSION: T1 receptor deficiency attenuated TBI-induced impairments of BBB by rescuing tight junction proteins and inhibited AQP4 polarization, thus improving the function of glymphatic system to enhance interstitial Aß clearance in TBI mice brain.


Subject(s)
Blood-Brain Barrier , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 1/metabolism , Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Occludin/metabolism , Occludin/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aquaporin 4/metabolism , Animals , Mice
4.
Neurol Res ; 44(8): 692-699, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35189787

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a common clinical condition caused by external force. Aquaporin-4 (AQP4) in astrocytes participates in the generation of cell swelling in TBI. METHODS: This research explored the effect of AQP4 gene silencing in a TBI rat model. A hydraulic craniocerebral trauma instrument was employed for establishing the TBI rat model. AQP4 expression in the brain was inhibited by the injection of AQP4 shRNA-lentiviral vector. The expression of relative genes was evaluated by Western blot and qRT-PCR. Neuronal apoptosis was analyzed by TUNEL assay. RESULTS: AQP4 shRNA treatment inhibited AQP4 expression in the brain of rats with TBI. AQP4 shRNA alleviated TBI-induced brain edema and neurological deficit in rats. Neuronal apoptosis and astrocyte activation in TBI rats were reduced by AQP4 silencing. CONCLUSION: This research demonstrated that AQP4 shRNA-induced silencing of AQP4 in the TBI rat model reduced the expression of AQP4 and GFAP, alleviated brain edema, neurological deficit, neuronal apoptosis and inhibited astrocyte activation.


Subject(s)
Aquaporin 4 , Brain Edema , Brain Injuries, Traumatic , Animals , Aquaporin 4/genetics , Astrocytes , Brain Edema/etiology , Brain Edema/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/therapy , Gene Silencing , Lentivirus/genetics , Lentivirus/metabolism , Neuroprotection , RNA, Small Interfering , Rats
5.
Bioeng Transl Med ; 6(3): e10203, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34589592

ABSTRACT

Treatments of glioblastoma (GBM) have not been very effective, largely due to the inefficiency of drugs in penetrating the blood brain barrier (BBB). In this study, we investigated the potential of exosome-coated doxorubicin (DOX)-loaded nanoparticles (ENPDOX) in BBB penetration, inducing immunogenic cell death (ICD) and promoting survival of GBM-bearing mice. DOX-loaded nanoparticles (NPDOX) were coated with exosomes prepared from mouse brain endothelial bEnd.3 cells. ENPDOX cellular uptake was examined. Penetration of ENPDOX through the BBB was tested in an in vitro transwell system and a GBM mouse model. The effects of ENPDOX in inducing apoptosis and ICD were assessed. Finally, the efficacy of ENPDOX in the treatment of GBM-bearing mice was assessed. ENPDOX was taken up by bEnd.3 cells and could penetrate the BBB both in vitro and in vivo. In vitro, ENDDOX induced apoptosis and ICD of glioma GL261 cells. Systemic administration of ENPDOX resulted in maturation of dendritic cells, activation of cytotoxic cells, altered production of cytokines, suppressed proliferation and increased apoptosis of GBM cells in vivo and prolonged survival of GBM-bearing mice. Our findings indicate that ENPDOX may be a potent therapeutic strategy for GBM which warrants further investigation in clinical application.

6.
Neurochem Res ; 46(6): 1380-1389, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33651262

ABSTRACT

Traumatic brain injury (TBI) has been a crucial health problem, with more than 50 million patients worldwide each year. Glymphatic system is a fluid exchange system that relies on the polarized water channel aquaporin-4 (AQP4) at the astrocytes, accounting for the clearance of abnormal proteins and metabolites from brain tissues. However, the dysfunction of glymphatic system and alteration of AQP4 polarization during the progression of TBI remain unclear. AQP4-/- and Wild Type (WT) mice were used to establish the TBI mouse model respectively. Brain edema and Evans blue extravasation were conducted 24 h post-injury to evaluate the acute TBI. Morris water maze (MWM) was used to establish the long-term cognitive functions of AQP4-/- and WT mice post TBI. Western-blot and qRT-PCR assays were performed to demonstrate protective effects of AQP4 deficiency to blood-brain barrier (BBB) integrity and amyloid-ß clearance. The inflammation of cerebral tissues post TBI was estimated by ELISA assay. AQP4 deficiency alleviated the brain edema and neurological deficit in TBI mice. AQP4-knockout led to improved cognitive outcomes in mice post TBI. The BBB integrity and cerebral amyloid-ß clearance were protected by AQP4 deficiency in TBI mice. AQP4 deficiency ameliorated the TBI-induced inflammation. AQP4 deficiency improved longer-term neurological outcomes in a mouse model of TBI.


Subject(s)
Aquaporin 4/deficiency , Brain Injuries, Traumatic/metabolism , Neuroprotection/physiology , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain Edema/etiology , Brain Edema/metabolism , Brain Injuries, Traumatic/complications , Disease Progression , Glymphatic System/metabolism , Inflammation/etiology , Inflammation/metabolism , Male , Mice, Knockout , Morris Water Maze Test/physiology
7.
Neuroreport ; 32(3): 188-197, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33470761

ABSTRACT

Pyroptosis has been reported to contribute to the traumatic brain injury (TBI) process. Ac-FLTD-CMK is a newly synthesized pyroptosis inhibitor. However, whether Ac-FLTD-CMK inhibits pyroptosis and plays a neuroprotective role after TBI is unknown. The present study aimed to determine the effects of Ac-FLTD-CMK on TBI in a mouse model. Male C57BL/6 mice were randomly divided into sham, TBI + vehicle, and TBI + Ac-FLTD-CMK groups. TBI was induced using a weight-drop apparatus. Intraventricular injection of Ac-FLTD-CMK was performed 30 min after TBI. Caspase-1, caspase-11, gasdermin-D (GSDMD), and caspase-3 expression in the peri-contusional cortex were assessed by western blotting. Interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) expression in the peri-contusional cortex were measured using ELISA. Behavioral experiments, brain water content, Evans blue extravasation, lactate dehydrogenase (LDH) release, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining were also performed. The results showed that Ac-FLTD-CMK administration significantly downregulated caspase-1 p20, caspase-11 p20, GSDMD N-terminal, IL-1ß, and IL-18 expression; reduced LDH release; alleviated neuronal death; attenuated brain edema and blood-brain barrier damage; and improved neurobehavioral function. These findings indicate that Ac-FLTD-CMK treatment suppresses pyroptosis and protects mice against TBI.


Subject(s)
Brain Contusion/metabolism , Brain/drug effects , Caspase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Caspase 1/drug effects , Caspase 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspases, Initiator/drug effects , Caspases, Initiator/metabolism , Disease Models, Animal , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Open Field Test , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Rotarod Performance Test
8.
Neurochem Res ; 45(12): 2926-2938, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33063244

ABSTRACT

With millions of traumatic brain injury (TBI) patients every year, TBI is regarded as one of the leading causes of human death and disability. Calcitonin gene-related peptide (CGRP) has been domenstrated to be a potential therapeutic target for TBI. However, the detailed effect and underlying mechanism of CGRP on the injured brain after TBI has hardly been investigated. In this work, we established TBI models of mice and injected CGRP before and after modelling to study its effects on the brain lesion, neurological functions and behaviours, neuron apoptosis and autophagy after TBI. Impacts of introduced CGRP on the activation of Akt/mTOR signalling in the cortical tissues surrounding injured areas after TBI were also evaluated. It was found that CGRP was reduced after TBI, and gradually restored over time. CGRP administration significantly restored the brain lesion induced by TBI. The permeability of blood-brain barrier and brain edema was increased dramatically after TBI, which was ameliorated by exogenous CGRP. Moreover, several neurological behaviour tests were performed, showing that CGRP introduction also relieved the cognitive abilities of mice which were impaired after TBI. Enhancing apoptosis and autophagy of neurons in the cortical tissues of injury sites following TBI were also alleviated by CGRP administration. Besides, CGRP-treated brain cortical tissues showed increased activation of Akt/mTOR signalling after TBI. Therefore, the results suggest that exogenous CGRP plays a neuroprotective role in the injuryed brain after TBI, to relieve cell apoptosis and autophagy, at least partially through Akt/mTOR signalling pathway. This finding also provides more evidence for the treatment of TBI through introducing exogenous CGRP or its related drugs.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Brain Injuries, Traumatic/drug therapy , Calcitonin Gene-Related Peptide/therapeutic use , Neuroprotective Agents/therapeutic use , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/drug effects , Brain/pathology , Brain Edema/drug therapy , Brain Edema/etiology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Humans , Male , Mice, Inbred BALB C , Morris Water Maze Test/drug effects , Neurons/drug effects , Open Field Test/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
9.
Front Neurol ; 11: 707, 2020.
Article in English | MEDLINE | ID: mdl-32765412

ABSTRACT

Background: The glymphatic pathway has been shown to be impaired in traumatic brain injury (TBI). Omega-3 polysaturated fatty acids (Omega-3, PUFAs) are involved in the clearance of amyloid-ß through the glymphatic system and this effect is Aquaporin-4 (AQP4) dependent. We hypothesize that Omega-3 PUFAs can alleviate neurological impairment in TBI by protecting the glymphatic pathway. Methods: We pretreated mice with Omega-3 PUFAs rich fish oil and introduced TBI in the mice. Neurological functions were assessed through the modified neurological severity score (mNSS) system and Rota-rod test. Aß42 levels and radioisotope clearance were examined to determine the function of glymphatic system. AQP4 protein and mRNA expressions and its polarity were examined in fish oil treated TBI mice or control mice. Finally, the integrity of blood-brain barrier was determined by Evans blue extravasation and measurement of tight junction proteins (ZO-1 and Occludin) levels. Results: TBI surgery induced significant neurological functional impairment, Omega-3 PUFAs attenuated TBI-induced neurological impairment, as evidenced by reduced mNSS, improved performance in the Rota-rod test. Furthermore, Omega-3 PUFAs improved glymphatic clearance after induction of TBI in mice, reduced Aß42 accumulation, partially restored the clearance of both 3H-mannitol and 14C-Inulin. Omega-3 PUFAs also suppressed AQP4 expression and partially prevented loss of AQP4 polarity in mice undergoing TBI. Finally, Omega-3 PUFAs protected mice from TBI induced blood-brain barrier disruption. Conclusion: Omaga-3 PUFAs attenuate neurological function by partially restoring the AQP4 dependent glymphatic system in mice with TBI.

10.
Cell Physiol Biochem ; 46(6): 2532-2542, 2018.
Article in English | MEDLINE | ID: mdl-29742510

ABSTRACT

BACKGROUND/AIMS: Traumatic brain injury (TBI) is a complex neurological injury in young adults lacking effective treatment. Emerging evidences suggest that inflammation contributes to the secondary brain injury following TBI, including breakdown of the blood brain barrier (BBB), subsequent edema and neurological deterioration. High mobility group box-1 (HMGB1) has been identified as a key cytokine in the inflammation reaction following TBI. Here, we investigated the therapeutic efficacy of HMGB1 A-box fragment, an antagonist competing with full-length HMGB1 for receptor binding, against TBI. METHODS: TBI was induced by controlled cortical impact (CCI) in adult male mice. HMGB1 A-box fragment was given intravenously at 2 mg/kg/day for 3 days after CCI. HMGB1 A-box-treated CCI mice were compared with saline-treated CCI mice and sham mice in terms of BBB disruption evaluated by Evan's blue extravasation, brain edema by brain water content, cell death by propidium iodide staining, inflammation by Western blot and ELISA assay for cytokine productions, as well as neurological functions by the modified Neurological Severity Score, wire grip and beam walking tests. RESULTS: HMGB1 A-box reversed brain damages in the mice following TBI. It significantly reduced brain edema by protecting integrity of the BBB, ameliorated cell degeneration, and decreased expression of pro-inflammatory cytokines released in injured brain after TBI. These cellular and molecular effects were accompanied by improved behavioral performance in TBI mice. Notably, HMGB1 A-box blocked IL-1ß-induced HMGB1 release, and preferentially attenuated TLR4, Myd88 and P65 in astrocyte cultures. CONCLUSION: Our data suggest that HMGB1 is involved in CCI-induced TBI, which can be inhibited by HMGB1 A-box fragment. Therefore, HMGB1 A-box fragment may have therapeutic potential for the secondary brain damages in TBI.


Subject(s)
Brain Edema/drug therapy , Brain Injuries, Traumatic/drug therapy , Brain/drug effects , HMGB1 Protein/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , Brain Edema/immunology , Brain Edema/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Cells, Cultured , Disease Models, Animal , HMGB1 Protein/chemistry , HMGB1 Protein/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
11.
J Clin Lab Anal ; 31(5)2017 Sep.
Article in English | MEDLINE | ID: mdl-28000287

ABSTRACT

BACKGROUND: We studied the association between high serum levels of vascular endothelial growth factor (VEGF) and clinical outcomes of intracerebral hemorrhage (ICH) patients. METHODS: Patients were divided into group A (<20 mL), group B (20-30 mL), and group C (>30 mL) based on the bleeding amount. ICH patients were also categorized into the mild group, moderate group (16-30), and severe group (31-45) based on the National Institutes of Health Stroke Scale (NIHSS). The serum levels of VEGF in acute ICH patients detected at 24, 48, and 72 hours were obtained using ELISA kit, and then compared with control group. Main clinical outcomes were evaluated using the modified Rankin scale at 90 days. RESULTS: The serum levels of VEGF were significantly higher than those in the control group. The serum levels of VEGF in group C were specifically higher compared with those in other two groups. The severe group exhibited higher levels of VEGF than the other two groups. NIHSS scores in patients with good outcomes were lower than those with poor outcomes. Besides, VEGF levels in patients with good outcomes were much higher than those in patients with poor outcomes. ROC results indicated that the optimal cut-off value of VEGF at 72 hours for predicting good outcomes was 111.17 pg/mL with 91.5 sensitivity, 98.7 specificity, and an AUC of 0.952 Our results showed that higher serum levels of VEGF were associated with process of ICH. CONCLUSION: VEGF could be a new marker in ICH for severity.


Subject(s)
Biomarkers/blood , Cerebral Hemorrhage , Vascular Endothelial Growth Factor A/blood , Aged , Case-Control Studies , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/diagnosis , Cerebral Hemorrhage/epidemiology , Female , Humans , Male , Middle Aged , Treatment Outcome
12.
Behav Brain Res ; 320: 412-419, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27818237

ABSTRACT

Intracranial hemorrhage (ICH) is a common subtype of stroke with high morbidity and mortality. However, few studies have examined the effects of diabetes on the recovery from ICH-induced brain injury. Therefore, we examined the effects of diabetes on protein levels of aquaporins, neuronal loss, angiogenesis, blood brain barrier (BBB) integrity, and neurological deficits following intra-DH collagenase-induced ICH in the hippocampus. We found that diabetic rats exhibited enhanced AQP9 expression in the hippocampus relative to non-diabetic rats, which was associated with increased behavioral deficits. Additionally, ICH induced neovascularization, proliferation of brain microvascular endothelial cells, and hippocampal neuronal loss. However, ICH-induced neovascularization and proliferation of brain microvascular endothelial cells was severely impaired in diabetic rats. Furthermore, ICH-induced hippocampal neuronal loss was exaggerated in diabetic rats. Finally, ICH impaired BBB integrity in the ipsilateral hemisphere, which was increased in diabetic rats. Taken together, the attenuated brain angiogenesis, increased hippocampal neuronal loss, and impaired BBB integrity in diabetic rats after ICH were associated with enhanced AQP9 expression. This may suggest that AQP9 is one of the underlying mechanisms that can mitigate the recovery from ICH in diabetic populations.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Gene Expression Regulation/physiology , Hippocampus/metabolism , Intracranial Hemorrhages/physiopathology , Recovery of Function/physiology , Animals , Aquaporins/metabolism , Blood-Brain Barrier/pathology , Collagenases/toxicity , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Gene Expression Regulation/drug effects , In Situ Nick-End Labeling , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/pathology , Male , Occludin/genetics , Occludin/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , von Willebrand Factor/metabolism
13.
Colloids Surf B Biointerfaces ; 129: 21-9, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25819362

ABSTRACT

Subarachnoid hemorrhage (SAH) is a life-threatening disease that causes high morbidity and mortality. Pirfenidone is a SAH drug that prevents secondary bleeding and cerebral infarction. To improve its therapeutic efficacy, this study aimed to employ a functionalized graphene oxide nanosheet (FGO) as a drug carrier loading pirfenidone to treat SAH. The graphene oxide nanosheet was introduced with transcription activator peptide (Tat), followed by functionalization with methoxy polyethylene glycol (mPEG) and loading with pirfenidone. The pirfenidone-loaded FGO (pirfenidone-FGO) exhibits better treatment efficacy than the single pirfenidone due to more effective loading and controlled release of the drug in tissue. The introduction of Tat and mPEG onto GO nanosheet contributes to the ability to cross the blood-brain barrier and the stability in blood circulation of the drug. At lower pH values, the highly efficient release of the drug from the pirfenidone-FGO exerts effective treatment to acidic inflammatory lesion after severe SAH. Besides its treatment function, FGO is also shown as a strong near infrared absorbing material which can be applied in photoacoustic imaging, allowing rapid real-time monitoring with deep resolution of brain tissues after SAH. The treatment efficacy of pirfenidone-FGO for central nervous system injuries is further demonstrated by hematoxylin and eosin staining of coronal brain slices, as well as measurements of brain water content and blood-brain barrier permeability. Our study supports the potential of FGO in clinical application in treatment of SAH.


Subject(s)
Brain/drug effects , Drug Carriers , Graphite/chemistry , Oxides/chemistry , Pyridones/pharmacology , Subarachnoid Hemorrhage/drug therapy , Animals , Blood-Brain Barrier/drug effects , Brain/pathology , Cell Survival/drug effects , Disease Models, Animal , Mice , PC12 Cells , Pyridones/administration & dosage , Rats , Subarachnoid Hemorrhage/pathology , Treatment Outcome
14.
J Craniofac Surg ; 24(5): 1646-9, 2013.
Article in English | MEDLINE | ID: mdl-24036713

ABSTRACT

In this study, we examined the effectiveness of percutaneous injected fibrin glue as a treatment for frontal sinus cerebrospinal fluid (CSF) rhinorrhea in a series of 4 cases. All 4 patients had fracture in the posterior wall of the frontal sinus. The anterior wall of the frontal sinus was punctured following high-resolution computed tomography imaging. In 3 out of 4 patients with defective skull due to prior frontal craniotomy, direct percutaneous puncture of the frontal sinus was used. Fibrin glue was injected to close the fistula and to seal the rhinorrhea. Surgery procedures lasted for 15-35 minutes (average 27.6 min). Rhinorrhea was stopped in all patients after the surgery, with no recurrence at a 10-month follow-up visit. In 1 case, the glue was expelled by coughing at 2 days after the surgery but was completely stopped with no recurrence after a second attempt. One patient with no recurrence at a 10-month follow-up died of tumor relapse at 12 months. In summary, fibrin glue could be used as a novel treatment for frontal sinus CSF rhinorrhea.


Subject(s)
Cerebrospinal Fluid Rhinorrhea/etiology , Cerebrospinal Fluid Rhinorrhea/surgery , Fibrin Tissue Adhesive/therapeutic use , Frontal Sinus/injuries , Skull Fractures/complications , Accidents, Traffic , Adult , Cerebrospinal Fluid Rhinorrhea/diagnostic imaging , Craniotomy , Female , Frontal Sinus/diagnostic imaging , Frontal Sinus/surgery , Humans , Male , Middle Aged , Skull Fractures/diagnostic imaging , Skull Fractures/surgery , Tomography, X-Ray Computed , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...