Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Aging (Albany NY) ; 162024 May 06.
Article in English | MEDLINE | ID: mdl-38713160

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and emerging evidence indicates that trigeminal nerve electrical stimulation (TNS) is a promising therapeutic intervention for neurological impairment following TBI. However, the precise mechanisms underlying the neuroprotective effects of TNS in TBI are poorly understood. Thus, the objective of this study was to investigate the potential involvement of the orexin-A (OX-A)/orexin receptor 1 (OX1R) mediated TLR4/NF-κB/NLRP3 signaling pathway in the neuroprotective effects of TNS in rats with TBI. METHODS: Sprague-Dawley rats were randomly assigned to four groups: sham, TBI, TBI+TNS+SB334867, and TBI+TNS. TBI was induced using a modified Feeney's method, and subsequent behavioral assessments were conducted to evaluate neurological function. The trigeminal nerve trunk was isolated, and TNS was administered following the establishment of the TBI model. The levels of neuroinflammation, brain tissue damage, and proteins associated with the OX1R/TLR4/NF-κB/NLRP3 signaling pathway were assessed using hematoxylin-eosin staining, Nissl staining, western blot analysis, quantitative real-time polymerase chain reaction, and immunofluorescence techniques. RESULTS: The findings of our study indicate that TNS effectively mitigated tissue damage, reduced brain edema, and alleviated neurological deficits in rats with TBI. Furthermore, TNS demonstrated the ability to attenuate neuroinflammation levels and inhibit the expression of proteins associated with the TLR4/NF-κB/NLRP3 signaling pathway. However, it is important to note that the aforementioned effects of TNS were reversible upon intracerebroventricular injection of an OX1R antagonist. CONCLUSION: TNS may prevent brain damage and relieve neurological deficits after a TBI by inhibiting inflammation, possibly via the TLR4/NF-κB/NLRP3 signaling pathway mediated by OX-A/OX1R.

2.
Aging (Albany NY) ; 16(7): 6566-6587, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38604164

ABSTRACT

Traumatic brain injury (TBI) and its resulting complications pose a major challenge to global public health, resulting in increased rates of disability and mortality. Cerebrovascular dysfunction is nearly universal in TBI cases and is closely associated with secondary injury after TBI. Transcranial direct current stimulation (tDCS) shows great potential in the treatment of TBI; however, the exact mechanism remains elusive. In this study, we performed in vivo and in vitro experiments to explore the effects and mechanisms of tDCS in a controlled cortical impact (CCI) rat model simulating TBI. In vivo experiments show that tDCS can effectively reduce brain tissue damage, cerebral edema and neurological deficits. The potential mechanism may be that tDCS improves the neurological function of rats by increasing orexin A (OXA) secretion, upregulating the TF-AKT/ERK signaling pathway, and promoting angiogenesis at the injury site. Cellular experiments showed that OXA promoted HUVEC migration and angiogenesis, and these effects were counteracted by the ERK1/2 inhibitor LY3214996. The results of Matrigel experiment in vivo showed that TNF-a significantly reduced the ability of HUVEC to form blood vessels, but OXA could rescue the effect of TNF-a on the ability of HUVEC to form blood vessels. However, LY3214996 could inhibit the therapeutic effect of OXA. In summary, our preliminary study demonstrates that tDCS can induce angiogenesis through the OXA-TF-AKT/ERK signaling pathway, thereby improving neurological function in rats with TBI.


Subject(s)
Brain Injuries, Traumatic , MAP Kinase Signaling System , Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt , Transcranial Direct Current Stimulation , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Male , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Humans , Human Umbilical Vein Endothelial Cells , Disease Models, Animal , Signal Transduction , Angiogenesis
3.
Aging (Albany NY) ; 16(4): 3404-3419, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38349868

ABSTRACT

BACKGROUND: Traumatic Brain Injury (TBI) has high disability and mortality rate. Oxidative stress and ferroptosis are important pathophysiological characteristics after TBI. Orexin-A (OXA) can alleviate neuronal damage in diverse neurological disorders. Nevertheless, the role and mechanism of OXA in TBI stay unknown. OBJECTIVES: The research investigated protection influence of OXA on TBI and its potential mechanisms. METHODS: Male Sprague-Dawley rats were randomly grouped into: sham, TBI, TBI + normal saline (NS) and TBI+OXA groups. TBI model was constructed in rat via modified Feeney's approach, and OXA treatment was administered following construction of TBI model. RESULTS: Relative to TBI+NS group, TBI+OXA group displayed greatly recovered tissue damage and neurological deficits. Additionally, OXA eased oxidative stress as well as ferroptosis in cerebral cortex of rats following TBI. Furthermore, OXA increased Nrf2 expression and regulating factors HO-1 and NQO1 in cerebral cortex of TBI rats. CONCLUSIONS: Our research found OXA may restrain ferroptosis via Nrf2/HO-1 signaling pathway activation, thereby reducing brain injury after TBI.


Subject(s)
Brain Injuries, Traumatic , Ferroptosis , Rats , Male , Animals , NF-E2-Related Factor 2/metabolism , Orexins/metabolism , Rats, Sprague-Dawley , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology
4.
Medicine (Baltimore) ; 103(2): e36701, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38215152

ABSTRACT

This study aimed to develop and validate a nomogram to predict awakening at 1 year in patients with prolonged disorders of consciousness (pDOC). We retrospectively analyzed the data of 381 patients with pDOC at 2 centers. The data were randomly divided into training and validation sets using a ratio of 6:4. For the training set, univariate and multivariate logical regression analyses were used to identify the predictive variables. Receiver operating characteristic curves, calibration curves, and a decision curve analysis were utilized to assess the predictive accuracy, discriminative ability, and clinical utility of the model, respectively. The final model included age, Glasgow Coma Scale score, serum albumin level, and computed tomography midline shift, all of which had a significant effect on awakening after pDOC. For the 1-year awakening in the training set, the model had good discriminative power, with an area under the curve of 0.733 (95% confidence interval: 0.667-0.789). For the validation set, the area under the curve for 1-year awakening was 0.721 (95% confidence interval: 0.617-0.826). Model performance was good for both the training and validation sets according to calibration plots and decision curve analysis. We developed a precise, effective nomogram to assist clinicians in better assessing patients' outcomes, guiding clinical judgment, and personalizing the therapeutic process.


Subject(s)
Consciousness Disorders , Craniocerebral Trauma , Humans , Consciousness Disorders/diagnosis , Consciousness Disorders/etiology , Retrospective Studies , Calibration , Glasgow Coma Scale , Nomograms
5.
Front Neurosci ; 17: 1323079, 2023.
Article in English | MEDLINE | ID: mdl-38156271

ABSTRACT

Background: Transcutaneous auricular vagus nerve stimulation (taVNS) has emerged as a potentially effective neuromodulation technique for addressing neurological disorders, including disorders of consciousness. Expanding upon our prior clinical study, which demonstrated the superior effectiveness of a 4-week taVNS treatment in patients with minimally conscious state (MCS) compared to those in a vegetative state/unresponsive wakefulness state, the aim of this investigation was to evaluate the safety and therapeutic efficacy of taVNS in individuals with MCS through a sham-controlled randomized double-blind clinical trial. Methods: A cohort of 50 adult patients (male = 33, female = 17) diagnosed with a MCS were randomly assigned to either the active taVNS (N = 25) or sham taVNS (N = 25) groups. The treatment period lasted for 4 weeks, followed by an 8-week follow-up period. The Coma Recovery Scale-Revised (CRS-R) and Glasgow Coma Scale (GCS) were administered at baseline and weekly during the initial 4 weeks. Additionally, the Disability Rating Scale (DRS) was used to assess the patients' functional abilities via telephone at week 12. Furthermore, various neurophysiological measures, including electroencephalogram (EEG), upper-limb somatosensory evoked potentials (USEP), brainstem auditory evoked potentials (BAEP), and P300 event-related potentials (P300), were employed to monitor changes in brain activity and neural conduction pathways. Results: The scores for the active taVNS group in the CRS-R and GCS showed greater improvement over time compared to the sham taVNS group (CRS-R: 1-week, Z = -1.248, p = 0.212; 2-week, Z = -1.090, p = 0.276; 3-week, Z = -2.017, p = 0.044; 4-week, Z = -2.267, p = 0.023. GCS: 1-week, Z = -1.325, p = 0.185; 2-week, Z = -1.245, p = 0.213; 3-week, Z = -1.848, p = 0.065; 4-week, Z = -1.990, p = 0.047). Additionally, the EEG, USEP, BAEP, and P300 also demonstrated significant improvement in the active taVNS group compared to the sham taVNS group at week 4 (EEG, Z = -2.086, p = 0.037; USEP, Z = -2.014, p = 0.044; BAEP, Z = -2.298, p = 0.022; P300 amplitude, Z = -1.974, p = 0.049; P300 latency, t = 2.275, p = 0.027). Subgroup analysis revealed that patients with MCS derived greater benefits from receiving taVNS treatment earlier (CRS-R, Disease duration ≤ 1-month, mean difference = 8.50, 95% CI = [2.22, 14.78], p = 0.027; GCS, Disease duration ≤ 1-month, mean difference = 3.58, 95% CI = [0.14, 7.03], p = 0.044). By week 12, the active taVNS group exhibited lower Disability Rating Scale (DRS) scores compared to the sham taVNS group (Z = -2.105, p = 0.035), indicating a more favorable prognosis for MCS patients who underwent taVNS. Furthermore, no significant adverse events related to taVNS were observed during treatment. Conclusion: The findings of this study suggest that taVNS may serve as a potentially effective and safe intervention for facilitating the restoration of consciousness in individuals diagnosed with MCS. This therapeutic approach appears to enhance cerebral functioning and optimize neural conduction pathways. Clinical trial registration: http://www.chictr.org.cn, Identifier ChiCTR2200066629.

6.
Ann Clin Transl Neurol ; 10(9): 1569-1577, 2023 09.
Article in English | MEDLINE | ID: mdl-37424159

ABSTRACT

OBJECTIVE: Aneurysmal subarachnoid hemorrhage (aSAH) is a major cause of death and disability worldwide and imposes serious burdens on society and individuals. However, predicting the long-term outcomes in aSAH patients requiring mechanical ventilation remains challenging. We sought to establish a model utilizing the Least Absolute Shrinkage and Selection Operator (LASSO)-penalized Cox regression to estimate the prognosis of aSAH patients requiring mechanical ventilation, based on regularly utilized and easily accessible clinical variables. METHODS: Data were retrieved from the Dryad Digital Repository. Potentially relevant features were selected using LASSO regression analysis. Multiple Cox proportional hazards analyses were performed to develop a model using the training set. Receiver operating characteristics and calibration curves were used to assess its predictive accuracy and discriminative power. Kaplan-Meier and decision curve analyses (DCA) were used to evaluate the clinical utility of the model. RESULTS: Independent prognostic factors, including the Simplified Acute Physiology Score 2, early brain injury, rebleeding, and length of intensive care unit stay, were identified and included in the nomogram. In the training set, the area under the curve values for 1-, 2-, and 4-year survival predictions were 0.82, 0.81, and 0.80, respectively. In the validation set, the nomogram exhibited excellent discrimination ability and good calibration. Moreover, DCA demonstrated that the nomogram was clinically beneficial. Finally, a web-based nomogram was constructed (https://rehablitation.shinyapps.io/aSAH). INTERPRETATION: Our model is a useful tool for accurately predicting long-term outcomes in patients with aSAH who require mechanical ventilation and can assist in making individualized interventions by providing valuable information.


Subject(s)
Brain Injuries , Subarachnoid Hemorrhage , Humans , Prognosis , Respiration, Artificial , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/therapy , ROC Curve
7.
Front Neurol ; 14: 1133893, 2023.
Article in English | MEDLINE | ID: mdl-36937511

ABSTRACT

Introduction: Disorders of consciousness (DoCs) are a frequent complication of brain injury disease, and effective treatments are currently lacking. Transauricular vagus nerve stimulation (tVNS) has been proposed as a promising therapeutic method for neurological disorders such as epilepsy and depression. In our previous study, we demonstrated that vagus nerve stimulation promoted recovery in rats with DoCs caused by traumatic brain injury. However, the clinical effect of vagus nerve stimulation on consciousness disorders is unclear. We aimed to investigate the therapeutic efficacy and safety of tVNS in patients with DoCs. Methods: We conducted a randomized, double-blinded, sham-controlled trial. Patients (N = 60) with DoCs, including minimally conscious state (MCS) and vegetative state/unresponsive wakefulness syndrome, were enrolled and randomized to groups receiving either active or sham tVNS. A frequency of 20 Hz and pulse wave of 200 us was used in the active-tVNS protocol, which was performed in the auricular branch of the vagus nerve in the left outer ear. The sham-tVNS protocol was the same as the active-tVNS protocol although without current input. Both groups of patients also received conventional treatments. Consciousness was evaluated according to the Coma Recovery Scale-Revised before and after the 4-week intervention. We also recorded the type and number of behavioral responses. Safety was primarily assessed according to the incidence of treatment-emergent adverse events. Each patient's heart rate and blood pressure were monitored during all treatment sessions. Results: Ultimately, 57 patients completed the study: 28 patients underwent active tVNS and 29 patients underwent sham tVNS. No significant differences were observed in Coma Recovery Scale-Revised scores between the active- and sham-tVNS groups before the tVNS sessions. Compared with patients in the sham-tVNS group (9.28 ± 4.38), patients with DoCs treated with active tVNS showed improved consciousness (10.93 ± 4.99), although not statistically significant. Further analysis revealed obvious differences between patients with MCS receiving active and sham tVNS, but no significant difference in patients with vegetative state/unresponsive wakefulness syndrome in both groups. All side effects were considered common medical conditions with no obvious correlation to tVNS. Conclusion: These preliminary data provide early evidence that tVNS may be an effective and safe approach for promoting the recovery of consciousness, especially in patients with MCS. Clinical trial registration: https://www.chictr.org.cn/edit.aspx?pid=175938&htm=4, identifier: ChiCTR2200066629.

8.
J Immunother Cancer ; 11(2)2023 02.
Article in English | MEDLINE | ID: mdl-36787938

ABSTRACT

BACKGROUND: GLI-similar 1 (GLIS1) is one of of Krüppel-like zinc finger proteins, which are either stimulators or inhibitors of genetic transcription. Nevertheless, its effects on T cell were elusive. METHODS: In this study, we intend to explore the effects of GLIS1 on modulating the anticancer potency of CD8+ T cells in hepatocellular carcinoma (HCC). The expression of GLIS1 in CD8 peripheral blood mononuclear cell and CD8 tumor-infiltrating lymphocytes of HCC tissues was validated by quantificational real-time-PCR and flow cytometry. The anticancer potency of CD8+ T cells with GLIS1 knock down was confirmed in C57BL/6 mouse model and HCC patient-derived xenograft mice model. GLIS1-/- C57BL/6 mice was applied to explore the effects GLIS1 on tumor immune microenvironment. Chromatin immunoprecipitation and RNA transcriptome sequencing analysis were both performed in GLIS1-knock down of CD8+ T cells. RESULTS: GLIS1 was upregulated in exhausted CD8+ T cells in HCC. GLIS1 downregulation in CD8+ T cells repressed cancer development, elevated the infiltrate ability of CD8+ T cells, mitigated CD8+ T cell exhaustion and ameliorated the anti-PD1 reaction of CD8+ T cells in HCC. The causal link beneath this included transcriptional regulation of SGK1-STAT3-PD1 pathway by GLIS1, thereby maintaining the abundant PD1 expression on the surface of CD8+ T cells. CONCLUSION: Our study revealed that GLIS1 promoted CD8+ T cell exhaustion in HCC through transcriptional regulating SGK1-STAT3-PD1 pathway. Downregulating the expression of GLIS1 in CD8+ T cells exerted an effect with anti-PD1 treatment synergistically, revealing a prospective method for HCC immune therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear/metabolism , Mice, Inbred C57BL , Tumor Microenvironment , STAT3 Transcription Factor/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
9.
Acta Biomater ; 157: 337-351, 2023 02.
Article in English | MEDLINE | ID: mdl-36509402

ABSTRACT

Transarterial chemoembolization (TACE) is an image-guided locoregional therapy used for the treatment of patients with primary hepatocellular carcinoma (HCC). However, conventional TACE formulations such as epirubicin-lipiodol emulsion are rapidly dissociated due to the instability of the emulsion, resulting in insufficient local drug concentrations in the target tumor. To overcome these limitations, we used biodegradable Idarubicin loaded microspheres (BILMs), which were prepared from gelatin and carrageenan and could be loaded with Idarubicin (IDA-MS). The morphology and the ability to load and release IDA of BILMs were characterized in vitro. We evaluated tumor changes and side effects after TACE treatment with IDA-MS in VX2 rabbit and C57BL/6 mice HCC models. In addition, the effect of IDA-MS on the tumor immune microenvironment of HCC tumors was elucidated via mass spectrometry and immunohistochemistry. Result showed that IDA-MS was developed as a new TACE formulation to overcome the poor delivery of drugs due to rapid elimination of the anticancer drug into the systemic circulation. We demonstrated in rabbits and mice HCC models that TACE with IDA-MS resulted in significant tumor shrinkage and no more severe adverse events than those observed in the IDA group. TACE with IDA-MS could also significantly enhance the sensitivity of anti-PD1 immunotherapy, improve the expression of CD8+ T cells, and activate the tumor immune microenvironment in HCC. This study provides a new approach for TACE therapy and immunotherapy and illuminates the future of HCC treatment. STATEMENT OF SIGNIFICANCE: Conventional transarterial chemoembolization (TACE) formulations are rapidly dissociated due to the instability of the emulsion, resulting in insufficient local drug concentrations in hepatocellular carcinoma (HCC). To overcome these limitations, we used biodegradable microspheres called BILMs, which could be loaded with Idarubicin (IDA-MS). We demonstrated in rabbits and mice HCC models that TACE with IDA-MS resulted in significant tumor shrinkage and no more severe adverse events than those observed in the IDA group. TACE with IDA-MS could also significantly enhance the sensitivity of anti-PD1 immunotherapy, improve the expression of CD8+ T cells, and activate the tumor immune microenvironment in HCC. This study provides a new approach for TACE therapy and immunotherapy and illuminates the future of HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Rabbits , Animals , Mice , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Idarubicin/pharmacology , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Microspheres , CD8-Positive T-Lymphocytes/pathology , Emulsions , Treatment Outcome , Chemoembolization, Therapeutic/methods , Mice, Inbred C57BL , Immunotherapy , Tumor Microenvironment
10.
Aging (Albany NY) ; 14(23): 9668-9678, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470667

ABSTRACT

PURPOSE: We assessed the relationship between consciousness level and values of cerebral blood flow (CBF), cerebral blood volume (CBV), mean transit time (MTT), and time to peak (TTP) obtained by whole-brain perfusion computed tomography (pCT) in patients with prolonged disorders of consciousness (pDOC). METHODS: This study included 29 patients in vegetative state (VS), 34 with minimally consciousness state minus (MCS-), and 13 with minimally consciousness state plus (MCS+). All patients were evaluated using the Coma Recovery Scale-Revised (CRS-R), the Glasgow Coma Scale (GCS), and the Full Outline of UnResponsiveness (FOUR). The values of CBF, CBV, MTT, and TTP were obtained from patients who underwent pCT. Differences in CBF, CBV, MTT, and TTP were compared between the three types of pDOC. Correlations between the CRS-R, GCS, and FOUR scores and the pCT results were analyzed. RESULTS: Among the three groups, patients in VS showed a significantly decreased CBF in the bilateral frontal lobe, thalamus, temporal lobe, occipital lobe, brainstem, and damaged part. CBV was significantly reduced in patients with VS in the bilateral frontal lobe, thalamus, temporal lobe, brainstem, and damaged part. The total CRS-R, GCS, and FOUR scores were positively correlated with CBF, CBV, and TTP in almost all regions of interest. CONCLUSION: Reductions in CBF and CBV calculated with pCT are associated with impaired consciousness and perfusion CT could be a promising tool in evaluating the conscious level in patients with pDOC.


Subject(s)
Consciousness Disorders , Consciousness , Humans , Consciousness Disorders/diagnostic imaging , Tomography, X-Ray Computed/methods , Perfusion
11.
Cancer Cell Int ; 22(1): 312, 2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36224624

ABSTRACT

BACKGROUND: Midazolam (MDZ) is an anaesthetic that is widely used for anxiolysis and sedation. More recently, MDZ has also been described to be related to the outcome of various types of carcinomas. However, how MDZ influences the progression of hepatocellular carcinoma (HCC) and its effects on the biological function and tumour immune microenvironment of this type of tumour remain unknown. METHODS: The effects of MDZ on the proliferation, invasion, and migration of HCC cell lines were examined in vitro using the Cell Counting Kit 8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU), Transwell, and wound healing assays. Additionally, western blotting was employed to confirm that PD-L1 was expressed. Chromatin immunoprecipitation-seq (ChIP-seq) analysis was used to pinpoint the transcriptional regulation regions of NF-κB and programmed death-ligand 1 (PD-L1). A C57BL/6 mouse model was used to produce subcutaneous HCC tumors in order to evaluate the in vivo performance of MDZ. Mass spectrometry was also used to assess changes in the tumour immunological microenvironment following MDZ injection. RESULTS: The HCC-LM3 and Hep-3B cell lines' proliferation, invasion, and migration were controlled by MDZ, according to the results of the CCK8, EdU, Transwell, and wound healing assays. PD-L1 expression was shown by ChIP-seq analysis to be boosted by NF-κB, and by Western blotting analysis, it was shown that MDZ downregulated the expression of NF-κB. Additionally, in vivo tests revealed that intraperitoneal MDZ injections reduced HCC tumor development and enhanced the effectiveness of anti-PD-1 therapy. The CD45+ immune cell proportions were higher in the MDZ group than in the PBS group, according to the mass spectrometry results. Injection of MDZ resulted in a decrease in the proportions of CD4+ T cells, CD8+ T cells, natural killer (NK) cells, monocytes, Tregs, and M2 macrophages and a rise in the proportion of dendritic cells. Additionally, the concentrations of the cytokines IFN-g and TNF-a were noticeably raised whereas the concentrations of the CD8+ T-cell fatigue markers ICOS, TIGIT, and TIM3 were noticeably lowered. CONCLUSION: According to this study, MDZ inhibited the progression of HCC by inhibiting the NF-κB pathway and reducing the exhaustion of CD8+ T cells. In clinical practice, MDZ combined with anti-PD-1 therapy might contribute to synergistically improving the antitumor efficacy of HCC treatment.

12.
Aging (Albany NY) ; 14(18): 7455-7469, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36126193

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a serious hazard to human health and is characterized by high rates of disability and mortality. It is necessary to explore new effective treatment methods to reduce the impact of TBI on individuals and society. As an emerging neuromodulation technique, ultrasound is used to treat some neurological diseases, but the neuroprotective mechanism of low-intensity focused ultrasound (LIFUS) in TBI remains unclear. We aimed to investigate the protective effects and potential mechanisms of LIFUS in TBI. METHODS: A rat model of TBI was established using the free-fall method. After establishing the TBI model, the hypothalamus region was covered with LIFUS radiation, and an orexin receptor 1 (OXR1) antagonist (SB334867) was injected intraperitoneally. Neurobehavioral examination, Nissl staining, hematoxylin and eosin staining of the brain tissue, and brain water content, were performed 3 days later. Western blotting, quantitative real-time polymerase chain reaction, immunofluorescence staining, and immunohistochemical staining, were used to evaluate the neuroprotective mechanisms of LIFUS. RESULTS: LIFUS improved tissue damage, neurological deficits, and brain edema. LIFUS can increase the expression of orexin-A (OX-A) and OXR1, significantly inhibit the activation of nuclear factor-κB (NF-κB) protein and nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome after TBI, and reduce the release of pro-inflammatory factors after TBI; however, SB334867 can reverse this effect. CONCLUSIONS: This study suggests that LIFUS may play a neuroprotective role by promoting the release of OX-A from the hypothalamus and inhibiting the inflammatory response after TBI through the OX-A /NF-κB/NLRP3 pathway.


Subject(s)
Brain Injuries, Traumatic , NF-kappa B , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Eosine Yellowish-(YS)/pharmacology , Hematoxylin/pharmacology , Humans , Inflammasomes/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotides , Orexin Receptors , Orexins/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Water
13.
Redox Biol ; 56: 102463, 2022 10.
Article in English | MEDLINE | ID: mdl-36108528

ABSTRACT

Single-cell RNA-sequencing (scRNA-seq) presents better insights into cell behavior in the context of a complex tumor microenvironment by profiling single-cell populations. However, the mechanisms underlying treatment failure in hepatocellular carcinoma (HCC) are poorly understood. In this study, we performed deep scRNA-seq on immune cells under the isolation in peripheral blood, cancer tissues, and nearby common tissues of four HCC cases and two non-cancer controls, and 212,494 cells were included in the analysis. We identified distinct immune cell subtypes, enriched pathways for differential genes, and delineated associated developmentally relevant trajectories. APOC1 was found over-expressed in tumor-associated macrophages (TAMs) of HCC tissues than in normal tissues. Inhibition of APOC1 reversed the M2 phenotype to the M1 phenotype via the ferroptosis pathway in TAMs from HCC. Tumors in APOC1 -/- C57BL/6 mice demonstrated consistent attenuation compared to wild-type (WT) mice. Mass spectrometry results revealed that the relative proportion of M2 macrophages, B cells, and CD4+ T cells in the APOC1 -/- group exhibited a downward expression compared with the WT group, whereas CD8+ T cells, M1 macrophages, and NK cells exhibited an upward trend. Finally, APOC1 was found to be negatively correlated with the expression of PD1/PD-L1 in human HCC samples. In conclusion, the present study demonstrated that inhibiting APOC1 can promote the transformation of M2 macrophages into M1 macrophages via the ferroptosis pathway, thereby reshaping the tumor immune microenvironment and improving the anti-PD1 immunotherapy for HCC, providing a new strategy for improving the therapeutic effect of anti-PD1, and bringing new hope to HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Animals , Apolipoprotein C-I , B7-H1 Antigen , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Ferroptosis/genetics , Humans , Immunotherapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Macrophages/metabolism , Mice , Mice, Inbred C57BL , RNA , Sequence Analysis, RNA , Tumor Microenvironment
14.
World Neurosurg ; 168: e19-e27, 2022 12.
Article in English | MEDLINE | ID: mdl-36064116

ABSTRACT

BACKGROUND: Despite the arousal effect of median nerve stimulation (MNS) being well documented in the clinical treatment of coma patients with traumatic brain injury (TBI), the mechanisms underlying the observed effect are still not completely understood. This study aimed to evaluate the protective effects and potential mechanism of MNS in comatose rats with TBI. METHODS: A total of 60 rats were randomly divided into 5 groups: the control group, sham-stimulated group, MNS group, orexins receptor type 1 (OX1R) antagonist group, and antagonist control group. The free-fall drop method was used to establish a TBI model. After administrating MNS or OX1R antagonist, consciousness was evaluated. Protein levels in the prefrontal cortex were measured using an enzyme-linked immunosorbent assay, Western blotting, and immunofluorescence. RESULTS: In the MNS group, tissue damage and consciousness state was markedly improved compared with that in the sham-stimulated group. Administration of the OX1R antagonist attenuated the beneficial effects of MNS in TBI-induced comatose rats. Additionally, MNS also significantly enhanced the expression of orexin-A/OX1R and the activation of Ras guanine nucleotide-releasing factor 1 (RasGRF1). CONCLUSIONS: These data show that MNS exerts its wake-promoting effect by activating the OX1R-RasGRF1 pathway in TBI-induced comatose rats.


Subject(s)
Brain Injuries, Traumatic , Coma , Orexins , ras-GRF1 , Animals , Rats , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/therapy , Coma/etiology , Coma/therapy , Median Nerve , Orexins/metabolism , ras-GRF1/metabolism , Rats, Sprague-Dawley , Signal Transduction
15.
Front Immunol ; 13: 876048, 2022.
Article in English | MEDLINE | ID: mdl-35812439

ABSTRACT

Immunotherapy treatments, particularly immune checkpoint blockade, can result in benefits in clinical settings. But many pre-clinical and clinical studies have shown that resistance to anti-PD1 therapy frequently occurs, leading to tumor recurrence and treatment failure, including in patients with hepatocellular carcinoma (HCC). In this study, 10 patients with HCC were remedied with anti-PD1, and pre-treatment biopsy samples were sequenced for 289 nanostring panel RNA to compare responsive and non-responsive tumors to identify possible pretreatment biomarkers or targets of anti-PD1 therapeutic responses. Fortunately, the expression of ß-Glucuronidase (GUSB) in the non-responding tumors was found to be remarkably higher than that in responding tumors. Results of the cell counting kit 8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU), transwell, wound healing test, and flow cytometry showed that GUSB facilitated proliferation, invasion, as well as migration of human HCC cells and downregulated PD-L1 expression by promoting miR-513a-5p. Additionally, as a GUSB inhibitor, amoxapine can reduce the progression of human HCC cells, and was an effective treatment for HCC and improved the sensitivity of anti-PD1 therapy. In summary, this study reveals that increased GUSB downregulates PD-L1 expression by promoting miR-513a-5p, leading to primary resistance to anti-PD1 treatment in HCC, and amoxapine enhances the sensitivity of anti-PD1 therapy by inhibiting GUSB, providing a new strategy and method for improving the efficacy of anti-PD1 therapy and bringing new prospects for therapy of HCC.


Subject(s)
Amoxapine , Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Glucuronidase , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , Neoplasm Recurrence, Local , Programmed Cell Death 1 Receptor/metabolism
16.
Front Aging Neurosci ; 14: 934283, 2022.
Article in English | MEDLINE | ID: mdl-35875805

ABSTRACT

Background: This study aimed to develop and validate a nomogram and present it on a website to be used to predict the overall survival at 16, 32, and 48 months in patients with prolonged disorder of consciousness (pDOC). Methods: We retrospectively analyzed the data of 381 patients with pDOC at two centers. The data were randomly divided into training and validation sets using a ratio of 6:4. On the training set, Cox proportional hazard analyses were used to identify the predictive variables. In the training set, two models were screened by COX regression analysis, and based on clinical evidence, model 2 was eventually selected in the nomogram after comparing the receiver operating characteristic (ROC) of the two models. In the training and validation sets, ROC curves, calibration curves, and decision curve analysis (DCA) curves were utilized to measure discrimination, calibration, and clinical efficacy, respectively. Results: The final model included age, Glasgow coma scale (GCS) score, serum albumin level, and computed tomography (CT) midline shift, all of which had a significant effect on survival after DOCs. For the 16-, 32-, and 48-month survival on the training set, the model had good discriminative power, with areas under the curve (AUCs) of 0.791, 0.760, and 0.886, respectively. For the validation set, the AUCs for the 16-, 32-, and 48-month survival predictions were 0.806, 0.789, and 0.867, respectively. Model performance was good for both the training and validation sets according to calibration plots and DCA. Conclusion: We developed an accurate, efficient nomogram, and a corresponding website based on four correlated factors to help clinicians improve their assessment of patient outcomes and help personalize the treatment process and clinical decisions.

17.
Aging (Albany NY) ; 14(2): 789-799, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35045397

ABSTRACT

PURPOSE: It is important to predict the prognosis of patients with prolonged disorders of consciousness (DOC). This study established and validated a nomogram and corresponding web-based calculator to predict outcomes for patients with prolonged DOC. METHODS: All data were obtained from the First Affiliated Hospital of Nanchang University and the Shangrao Hospital of Traditional Chinese Medicine. Predictive variables were identified by univariate and multiple logistic regression analyses. Receiver operating characteristic curves, calibration curves, and a decision curve analysis (DCA) were utilized to assess the predictive accuracy, discriminative ability, and clinical utility of the model, respectively. RESULTS: Independent prognostic factors, such as age, Glasgow coma scale score, state of consciousness, and brainstem auditory-evoked potential grade were integrated into a nomogram. The model demonstrated good discrimination in the training and validation cohorts, with area-under-the-curve values of 0.815 (95% confidence interval [CI]: 0.748-0.882) and 0.805 (95% CI: 0.727-0.883), respectively. The calibration plots and DCA demonstrated good model performance and clear clinical benefits in both cohorts. CONCLUSIONS: Based on our nomogram, we developed an effective, simple, and accurate model of a web-based calculator that may help individualize healthcare decision-making. Further research is warranted to optimize the system and update the predictors.


Subject(s)
Consciousness , Nomograms , Humans , Prognosis , ROC Curve
18.
J Inflamm Res ; 14: 2007-2017, 2021.
Article in English | MEDLINE | ID: mdl-34040413

ABSTRACT

BACKGROUND: Neuronal damage is the main cause of neurological diseases. Neural stem cells (NSCs) have the functions of cell repair and replacement of neurons, secretion of neurotrophic factors, and immune regulation of the neural microenvironment. OBJECTIVE: Previous study found that Orexin-A had a protective effect on neurons in the central nervous system, but it is lacking in making great efforts on the function of Orexin-A on NSCs. This study aimed to investigate the anti-inflammatory responses and signaling mechanisms of Orexin-A on lipopolysaccharide (LPS)-induced NSCs. METHODS: Quantitative real-time polymerase chain reaction was used to detect the mRNA level. Signaling pathway-related protein expression was detected by Western blot. The proliferation and migration of NSCs were investigated by Cell Counting Kit-8 (CCK-8) detection kit and transwell assay. Besides, the staining of hematoxylin and eosin (HE) was performed to study the morphology of cell. RESULTS: Orexin-A decreased the pro-inflammatory cytokines of IL-1ß, TNF-α, and IL-6 induced by LPS by regulating nuclear factor-k-gene binding (NF-kB) and phosphorylation of P38/Erk-mitogen-activated protein kinases (MAPKs) pathways, but not p-JNK signaling. CONCLUSION: Our findings indicate that Orexin-A can alleviate the inflammatory response of NSC. It can provide beneficial help in neural stem cell therapy applications.

19.
Neurorehabil Neural Repair ; 34(9): 831-843, 2020 09.
Article in English | MEDLINE | ID: mdl-32772884

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Oxidative stress, inflammation, and apoptosis are vital pathophysiological features post-TBI. OBJECTIVES: Research has shown that vagus nerve stimulation (VNS) can attenuate oxidative stress in various diseases. However, the critical role of VNS in TBI is still not completely understood. This study investigated the protective effects and potential mechanism of VNS on TBI. METHODS: Male Sprague-Dawley rats were randomized into 3 groups: sham, TBI, and TBI + VNS. The TBI model was induced in rats by the free-fall drop method. The vagal nerve trunk was separated, and VNS was performed after establishing the TBI model. RESULTS: The results showed that VNS significantly ameliorated tissue damage, neurological deficits, and cerebral edema, compared with the sham VNS group. Additionally, VNS alleviated oxidative stress, inflammation, and apoptosis in the pericontusive cortex of rats after TBI. VNS also significantly suppressed expression of the nuclear factor-κB (NF-κB) protein in the nucleus and activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome. CONCLUSIONS: Taken together, the present study indicates that VNS may attenuate brain damage after TBI by inhibiting oxidative stress, inflammation, and apoptosis, possibly through the NF-κB/NLRP3 signaling pathway.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Inflammasomes/metabolism , Inflammation/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Vagus Nerve Stimulation , Animals , Apoptosis/physiology , Brain Injuries, Traumatic/immunology , Disease Models, Animal , Inflammation/immunology , Male , Random Allocation , Rats , Rats, Sprague-Dawley
20.
J Ethnopharmacol ; 156: 175-81, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25219605

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Huang-Lian-Jie-Du-Decotion (HLJDD), an important traditional Chinese medicine formula, has been used for various diseases in clinical practice, and thus has high potential to induce cytochrome P450 (CYP) isoenzymes/P-glycoprotein (P-gp) mediated herb-drug interactions (HDIs) with other co-administered drugs. The purpose of this study was to investigate the in vitro effects of multiple extracts including aqueous extracts, total flavonoids, iridoids, alkaloids from HLJDD on the activities of CYPs in rats (CYP1A2, CYP2C6, CYP2D2, CYP2E1 and CYP3A1) and P-gp, and then to predict potential interactions with co-administered drugs. MATERIALS AND METHODS: The effects of the four extracts from HLJDD on the CYPs activity were evaluated in rat liver microsomes incubation system, and then determined by LC-MS/MS-based CYPs probe substrate assay. Caco-2 cell monolayer was used to investigate the effect of the four extracts on the efflux of Rhodamine 123 to evaluate their influences on P-gp activity. RESULTS: The results show that total flavonoids and alkaloids exibited strong inhibition on rat CYP isoenzymes activities. Total flavonoids exhibited different inhibitory effects on CYPs activities with an order of CYP3A1>CYP2C6>CYP2E1>CYP1A2>CYP2D2, and the values of IC50 were 4.24, 8.16, 17.56, 19.03, 29.51 µg/mL, respectively. Total alkaloids possessed similar inhibition on CYPs and could strongly inhibit the activity of CYP2D2 (IC50=2.38 µg/mL), CYP3A1 (IC50=2.61 µg/mL), CYP2E1 (IC50=22.35 µg/mL), CYP1A2 (IC50=23.2 µg/mL) and CYP2C6 (IC50=43.09 µg/mL). Moderate degree of inhibition on CYPs activities was observed in aqueous and total iridoids extracts. Results from transport assay revealed that total flavonoids and alkaloids exhibited significant inhibitory effect on P-gp activity as evidenced by strong inhibition on the efflux of Rhodamine-123 with IC50 of 104.6 and 82.6 µg/mL. But aqueous extract showed weak and iridoids had negligible effect on P-gp activity. CONCLUSIONS: This study clearly demonstrated that total flavonoids and alkaloids from HLJDD can significantly inhibit the activities of CYPs and P-gp, which should be taken into consideration to predict any potential HDIs when HLJDD and its bioactive components are co-administered with other therapeutic drugs metabolized by CYPs or transported by P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Isoenzymes/antagonists & inhibitors , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Caco-2 Cells , Cell Line, Tumor , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Iridoids/chemistry , Iridoids/pharmacology , Male , Medicine, Chinese Traditional/methods , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...