Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Innovation (Camb) ; 5(4): 100648, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39021525

RESUMEN

Pulmonary infections pose formidable challenges in clinical settings with high mortality rates across all age groups worldwide. Accurate diagnosis and early intervention are crucial to improve patient outcomes. Artificial intelligence (AI) has the capability to mine imaging features specific to different pathogens and fuse multimodal features to reach a synergistic diagnosis, enabling more precise investigation and individualized clinical management. In this study, we successfully developed a multimodal integration (MMI) pipeline to differentiate among bacterial, fungal, and viral pneumonia and pulmonary tuberculosis based on a real-world dataset of 24,107 patients. The area under the curve (AUC) of the MMI system comprising clinical text and computed tomography (CT) image scans yielded 0.910 (95% confidence interval [CI]: 0.904-0.916) and 0.887 (95% CI: 0.867-0.909) in the internal and external testing datasets respectively, which were comparable to those of experienced physicians. Furthermore, the MMI system was utilized to rapidly differentiate between viral subtypes with a mean AUC of 0.822 (95% CI: 0.805-0.837) and bacterial subtypes with a mean AUC of 0.803 (95% CI: 0.775-0.830). Here, the MMI system harbors the potential to guide tailored medication recommendations, thus mitigating the risk of antibiotic misuse. Additionally, the integration of multimodal factors in the AI-driven system also provided an evident advantage in predicting risks of developing critical illness, contributing to more informed clinical decision-making. To revolutionize medical care, embracing multimodal AI tools in pulmonary infections will pave the way to further facilitate early intervention and precise management in the foreseeable future.

2.
Int J Mol Sci ; 25(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38928008

RESUMEN

Mitochondrial one-carbon metabolism provides carbon units to several pathways, including nucleic acid synthesis, mitochondrial metabolism, amino acid metabolism, and methylation reactions. Late-onset Alzheimer's disease is the most common age-related neurodegenerative disease, characterised by impaired energy metabolism, and is potentially linked to mitochondrial bioenergetics. Here, we discuss the intersection between the molecular pathways linked to both mitochondrial one-carbon metabolism and Alzheimer's disease. We propose that enhancing one-carbon metabolism could promote the metabolic processes that help brain cells cope with Alzheimer's disease-related injuries. We also highlight potential therapeutic avenues to leverage one-carbon metabolism to delay Alzheimer's disease pathology.


Asunto(s)
Enfermedad de Alzheimer , Carbono , Metabolismo Energético , Mitocondrias , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Mitocondrias/metabolismo , Carbono/metabolismo , Animales
3.
Trials ; 25(1): 358, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38835091

RESUMEN

BACKGROUND: This multicenter, double-blinded, randomized controlled trial (RCT) aims to assess the impact of an artificial intelligence (AI)-based model on the efficacy of intracranial aneurysm detection in CT angiography (CTA) and its influence on patients' short-term and long-term outcomes. METHODS: Study design: Prospective, multicenter, double-blinded RCT. SETTINGS: The model was designed for the automatic detection of intracranial aneurysms from original CTA images. PARTICIPANTS: Adult inpatients and outpatients who are scheduled for head CTA scanning. Randomization groups: (1) Experimental Group: Head CTA interpreted by radiologists with the assistance of the True-AI-integrated intracranial aneurysm diagnosis strategy (True-AI arm). (2) Control Group: Head CTA interpreted by radiologists with the assistance of the Sham-AI-integrated intracranial aneurysm diagnosis strategy (Sham-AI arm). RANDOMIZATION: Block randomization, stratified by center, gender, and age group. PRIMARY OUTCOMES: Coprimary outcomes of superiority in patient-level sensitivity and noninferiority in specificity for the True-AI arm to the Sham-AI arm in intracranial aneurysms. SECONDARY OUTCOMES: Diagnostic performance for other intracranial lesions, detection rates, workload of CTA interpretation, resource utilization, treatment-related clinical events, aneurysm-related events, quality of life, and cost-effectiveness analysis. BLINDING: Study participants and participating radiologists will be blinded to the intervention. SAMPLE SIZE: Based on our pilot study, the patient-level sensitivity is assumed to be 0.65 for the Sham-AI arm and 0.75 for the True-AI arm, with specificities of 0.90 and 0.88, respectively. The prevalence of intracranial aneurysms for patients undergoing head CTA in the hospital is approximately 12%. To establish superiority in sensitivity and noninferiority in specificity with a margin of 5% using a one-sided α = 0.025 to ensure that the power of coprimary endpoint testing reached 0.80 and a 5% attrition rate, the sample size was determined to be 6450 in a 1:1 allocation to True-AI or Sham-AI arm. DISCUSSION: The study will determine the precise impact of the AI system on the detection performance for intracranial aneurysms in a double-blinded design and following the real-world effects on patients' short-term and long-term outcomes. TRIAL REGISTRATION: This trial has been registered with the NIH, U.S. National Library of Medicine at ClinicalTrials.gov, ID: NCT06118840 . Registered 11 November 2023.


Asunto(s)
Inteligencia Artificial , Angiografía por Tomografía Computarizada , Aneurisma Intracraneal , Humanos , Aneurisma Intracraneal/diagnóstico por imagen , Método Doble Ciego , Estudios Prospectivos , Valor Predictivo de las Pruebas , Estudios Multicéntricos como Asunto , Angiografía Cerebral/métodos , Masculino , Femenino , Factores de Tiempo , Ensayos Clínicos Controlados Aleatorios como Asunto , Adulto
4.
Invest Ophthalmol Vis Sci ; 65(5): 31, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38771572

RESUMEN

Purpose: Although effective amblyopia treatments are available, treatment outcome is unpredictable, and the condition recurs in up to 25% of the patients. We aimed to evaluate whether a large-scale quantitative contrast sensitivity function (CSF) data source, coupled with machine learning (ML) algorithms, can predict amblyopia treatment response and recurrence in individuals. Methods: Visual function measures from traditional chart vision acuity (VA) and novel CSF assessments were used as the main predictive variables in the models. Information from 58 potential predictors was extracted to predict treatment response and recurrence. Six ML methods were applied to construct models. The SHapley Additive exPlanations was used to explain the predictions. Results: A total of 2559 consecutive records of 643 patients with amblyopia were eligible for modeling. Combining variables from VA and CSF assessments gave the highest accuracy for treatment response prediction, with the area under the receiver operating characteristic curve (AUC) of 0.863 and 0.815 for outcome predictions after 3 and 6 months, respectively. Variables from the VA assessment alone predicted the treatment response, with AUC values of 0.723 and 0.675 after 3 and 6 months, respectively. Variables from the CSF assessment gave rise to an AUC of 0.909 for recurrence prediction compared to 0.539 for VA assessment alone, and adding VA variables did not improve predictive performance. The interocular differences in CSF features are significant contributors to recurrence risk. Conclusions: Our models showed CSF data could enhance treatment response prediction and accurately predict amblyopia recurrence, which has the potential to guide amblyopia management by enabling patient-tailored decision making.


Asunto(s)
Ambliopía , Sensibilidad de Contraste , Recurrencia , Agudeza Visual , Humanos , Ambliopía/terapia , Ambliopía/fisiopatología , Ambliopía/diagnóstico , Agudeza Visual/fisiología , Masculino , Femenino , Sensibilidad de Contraste/fisiología , Niño , Resultado del Tratamiento , Preescolar , Curva ROC , Aprendizaje Automático , Estudios Retrospectivos , Adolescente , Privación Sensorial , Algoritmos
5.
Health Informatics J ; 30(2): 14604582241255818, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779978

RESUMEN

Mycoplasma pneumonia may lead to hospitalizations and pose life-threatening risks in children. The automated identification of mycoplasma pneumonia from electronic medical records holds significant potential for improving the efficiency of hospital resource allocation. In this study, we proposed a novel method for identifying mycoplasma pneumonia by integrating multi-modal features derived from both free-text descriptions and structured test data in electronic medical records. Our approach begins with the extraction of free-text and structured data from clinical records through a systematic preprocessing pipeline. Subsequently, we employ a pre-trained transformer language model to extract features from the free-text, while multiple additive regression trees are used to transform features from the structured data. An attention-based fusion mechanism is then applied to integrate these multi-modal features for effective classification. We validated our method using clinic records of 7157 patients, retrospectively collected for training and testing purposes. The experimental results demonstrate that our proposed multi-modal fusion approach achieves significant improvements over other methods across four key performance metrics.


Asunto(s)
Registros Electrónicos de Salud , Neumonía por Mycoplasma , Humanos , Neumonía por Mycoplasma/diagnóstico , Registros Electrónicos de Salud/estadística & datos numéricos , Niño , Estudios Retrospectivos , Mycoplasma pneumoniae/patogenicidad , Femenino , Masculino , Preescolar
6.
Radiology ; 311(1): e232057, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38591974

RESUMEN

Background Preoperative discrimination of preinvasive, minimally invasive, and invasive adenocarcinoma at CT informs clinical management decisions but may be challenging for classifying pure ground-glass nodules (pGGNs). Deep learning (DL) may improve ternary classification. Purpose To determine whether a strategy that includes an adjudication approach can enhance the performance of DL ternary classification models in predicting the invasiveness of adenocarcinoma at chest CT and maintain performance in classifying pGGNs. Materials and Methods In this retrospective study, six ternary models for classifying preinvasive, minimally invasive, and invasive adenocarcinoma were developed using a multicenter data set of lung nodules. The DL-based models were progressively modified through framework optimization, joint learning, and an adjudication strategy (simulating a multireader approach to resolving discordant nodule classifications), integrating two binary classification models with a ternary classification model to resolve discordant classifications sequentially. The six ternary models were then tested on an external data set of pGGNs imaged between December 2019 and January 2021. Diagnostic performance including accuracy, specificity, and sensitivity was assessed. The χ2 test was used to compare model performance in different subgroups stratified by clinical confounders. Results A total of 4929 nodules from 4483 patients (mean age, 50.1 years ± 9.5 [SD]; 2806 female) were divided into training (n = 3384), validation (n = 579), and internal (n = 966) test sets. A total of 361 pGGNs from 281 patients (mean age, 55.2 years ± 11.1 [SD]; 186 female) formed the external test set. The proposed strategy improved DL model performance in external testing (P < .001). For classifying minimally invasive adenocarcinoma, the accuracy was 85% and 79%, sensitivity was 75% and 63%, and specificity was 89% and 85% for the model with adjudication (model 6) and the model without (model 3), respectively. Model 6 showed a relatively narrow range (maximum minus minimum) across diagnostic indexes (accuracy, 1.7%; sensitivity, 7.3%; specificity, 0.9%) compared with the other models (accuracy, 0.6%-10.8%; sensitivity, 14%-39.1%; specificity, 5.5%-17.9%). Conclusion Combining framework optimization, joint learning, and an adjudication approach improved DL classification of adenocarcinoma invasiveness at chest CT. Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Sohn and Fields in this issue.


Asunto(s)
Adenocarcinoma del Pulmón , Adenocarcinoma , Aprendizaje Profundo , Neoplasias Pulmonares , Humanos , Femenino , Persona de Mediana Edad , Estudios Retrospectivos , Adenocarcinoma del Pulmón/diagnóstico por imagen , Adenocarcinoma/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Neoplasias Pulmonares/diagnóstico por imagen
7.
Cell Death Dis ; 15(3): 203, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467609

RESUMEN

Organelles form membrane contact sites between each other, allowing for the transfer of molecules and signals. Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) are cellular subdomains characterized by close apposition of mitochondria and ER membranes. They have been implicated in many diseases, including neurodegenerative, metabolic, and cardiac diseases. Although MERCS have been extensively studied, much remains to be explored. To uncover novel regulators of MERCS, we conducted a genome-wide, flow cytometry-based screen using an engineered MERCS reporter cell line. We found 410 genes whose downregulation promotes MERCS and 230 genes whose downregulation decreases MERCS. From these, 29 genes were selected from each population for arrayed screening and 25 were validated from the high population and 13 from the low population. GET4 and BAG6 were highlighted as the top 2 genes that upon suppression increased MERCS from both the pooled and arrayed screens, and these were subjected to further investigation. Multiple microscopy analyses confirmed that loss of GET4 or BAG6 increased MERCS. GET4 and BAG6 were also observed to interact with the known MERCS proteins, inositol 1,4,5-trisphosphate receptors (IP3R) and glucose-regulated protein 75 (GRP75). In addition, we found that loss of GET4 increased mitochondrial calcium uptake upon ER-Ca2+ release and mitochondrial respiration. Finally, we show that loss of GET4 rescues motor ability, improves lifespan and prevents neurodegeneration in a Drosophila model of Alzheimer's disease (Aß42Arc). Together, these results suggest that GET4 is involved in decreasing MERCS and that its loss is neuroprotective.


Asunto(s)
Sistemas CRISPR-Cas , Membranas Asociadas a Mitocondrias , Sistemas CRISPR-Cas/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Calcio/metabolismo
8.
Lancet Digit Health ; 6(4): e261-e271, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38519154

RESUMEN

BACKGROUND: Artificial intelligence (AI) models in real-world implementation are scarce. Our study aimed to develop a CT angiography (CTA)-based AI model for intracranial aneurysm detection, assess how it helps clinicians improve diagnostic performance, and validate its application in real-world clinical implementation. METHODS: We developed a deep-learning model using 16 546 head and neck CTA examination images from 14 517 patients at eight Chinese hospitals. Using an adapted, stepwise implementation and evaluation, 120 certified clinicians from 15 geographically different hospitals were recruited. Initially, the AI model was externally validated with images of 900 digital subtraction angiography-verified CTA cases (examinations) and compared with the performance of 24 clinicians who each viewed 300 of these cases (stage 1). Next, as a further external validation a multi-reader multi-case study enrolled 48 clinicians to individually review 298 digital subtraction angiography-verified CTA cases (stage 2). The clinicians reviewed each CTA examination twice (ie, with and without the AI model), separated by a 4-week washout period. Then, a randomised open-label comparison study enrolled 48 clinicians to assess the acceptance and performance of this AI model (stage 3). Finally, the model was prospectively deployed and validated in 1562 real-world clinical CTA cases. FINDINGS: The AI model in the internal dataset achieved a patient-level diagnostic sensitivity of 0·957 (95% CI 0·939-0·971) and a higher patient-level diagnostic sensitivity than clinicians (0·943 [0·921-0·961] vs 0·658 [0·644-0·672]; p<0·0001) in the external dataset. In the multi-reader multi-case study, the AI-assisted strategy improved clinicians' diagnostic performance both on a per-patient basis (the area under the receiver operating characteristic curves [AUCs]; 0·795 [0·761-0·830] without AI vs 0·878 [0·850-0·906] with AI; p<0·0001) and a per-aneurysm basis (the area under the weighted alternative free-response receiver operating characteristic curves; 0·765 [0·732-0·799] vs 0·865 [0·839-0·891]; p<0·0001). Reading time decreased with the aid of the AI model (87·5 s vs 82·7 s, p<0·0001). In the randomised open-label comparison study, clinicians in the AI-assisted group had a high acceptance of the AI model (92·6% adoption rate), and a higher AUC when compared with the control group (0·858 [95% CI 0·850-0·866] vs 0·789 [0·780-0·799]; p<0·0001). In the prospective study, the AI model had a 0·51% (8/1570) error rate due to poor-quality CTA images and recognition failure. The model had a high negative predictive value of 0·998 (0·994-1·000) and significantly improved the diagnostic performance of clinicians; AUC improved from 0·787 (95% CI 0·766-0·808) to 0·909 (0·894-0·923; p<0·0001) and patient-level sensitivity improved from 0·590 (0·511-0·666) to 0·825 (0·759-0·880; p<0·0001). INTERPRETATION: This AI model demonstrated strong clinical potential for intracranial aneurysm detection with improved clinician diagnostic performance, high acceptance, and practical implementation in real-world clinical cases. FUNDING: National Natural Science Foundation of China. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Asunto(s)
Aprendizaje Profundo , Aneurisma Intracraneal , Humanos , Aneurisma Intracraneal/diagnóstico por imagen , Angiografía por Tomografía Computarizada , Inteligencia Artificial , Estudios Prospectivos , Angiografía Cerebral/métodos
9.
J Imaging Inform Med ; 37(3): 922-934, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38332402

RESUMEN

This study aimed to assess the performance of a deep learning algorithm in helping radiologist achieve improved efficiency and accuracy in chest radiograph diagnosis. We adopted a deep learning algorithm to concurrently detect the presence of normal findings and 13 different abnormalities in chest radiographs and evaluated its performance in assisting radiologists. Each competing radiologist had to determine the presence or absence of these signs based on the label provided by the AI. The 100 radiographs were randomly divided into two sets for evaluation: one without AI assistance (control group) and one with AI assistance (test group). The accuracy, false-positive rate, false-negative rate, and analysis time of 111 radiologists (29 senior, 32 intermediate, and 50 junior) were evaluated. A radiologist was given an initial score of 14 points for each image read, with 1 point deducted for an incorrect answer and 0 points given for a correct answer. The final score for each doctor was automatically calculated by the backend calculator. We calculated the mean scores of each radiologist in the two groups (the control group and the test group) and calculated the mean scores to evaluate the performance of the radiologists with and without AI assistance. The average score of the 111 radiologists was 597 (587-605) in the control group and 619 (612-626) in the test group (P < 0.001). The time spent by the 111 radiologists on the control and test groups was 3279 (2972-3941) and 1926 (1710-2432) s, respectively (P < 0.001). The performance of the 111 radiologists in the two groups was evaluated by the area under the receiver operating characteristic curve (AUC). The radiologists showed better performance on the test group of radiographs in terms of normal findings, pulmonary fibrosis, heart shadow enlargement, mass, pleural effusion, and pulmonary consolidation recognition, with AUCs of 1.0, 0.950, 0.991, 1.0, 0.993, and 0.982, respectively. The radiologists alone showed better performance in aortic calcification (0.993), calcification (0.933), cavity (0.963), nodule (0.923), pleural thickening (0.957), and rib fracture (0.987) recognition. This competition verified the positive effects of deep learning methods in assisting radiologists in interpreting chest X-rays. AI assistance can help to improve both the efficacy and efficiency of radiologists.


Asunto(s)
Inteligencia Artificial , Aprendizaje Profundo , Radiografía Torácica , Radiólogos , Humanos , Radiografía Torácica/métodos , Masculino , Interpretación de Imagen Radiográfica Asistida por Computador/métodos , Algoritmos , Femenino , Persona de Mediana Edad , Adulto
10.
J Xray Sci Technol ; 32(3): 583-596, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38306089

RESUMEN

PURPOSE: The explore the added value of peri-calcification regions on contrast-enhanced mammography (CEM) in the differential diagnosis of breast lesions presenting as only calcification on routine mammogram. METHODS: Patients who underwent CEM because of suspicious calcification-only lesions were included. The test set included patients between March 2017 and March 2019, while the validation set was collected between April 2019 and October 2019. The calcifications were automatically detected and grouped by a machine learning-based computer-aided system. In addition to extracting radiomic features on both low-energy (LE) and recombined (RC) images from the calcification areas, the peri-calcification regions, which is generated by extending the annotation margin radially with gradients from 1 mm to 9 mm, were attempted. Machine learning (ML) models were built to classify calcifications into malignant and benign groups. The diagnostic matrices were also evaluated by combing ML models with subjective reading. RESULTS: Models for LE (significant features: wavelet-LLL_glcm_Imc2_MLO; wavelet-HLL_firstorder_Entropy_MLO; wavelet-LHH_glcm_DifferenceVariance_CC; wavelet-HLL_glcm_SumEntropy_MLO;wavelet-HLH_glrlm_ShortRunLowGray LevelEmphasis_MLO; original_firstorder_Entropy_MLO; original_shape_Elongation_MLO) and RC (significant features: wavelet-HLH_glszm_GrayLevelNonUniformityNormalized_MLO; wavelet-LLH_firstorder_10Percentile_CC; original_firstorder_Maximum_MLO; wavelet-HHH_glcm_Autocorrelation_MLO; original_shape_Elongation_MLO; wavelet-LHL_glszm_GrayLevelNonUniformityNormalized_MLO; wavelet-LLH_firstorder_RootMeanSquared_MLO) images were set up with 7 features. Areas under the curve (AUCs) of RC models are significantly better than those of LE models with compact and expanded boundary (RC v.s. LE, compact: 0.81 v.s. 0.73, p < 0.05; expanded: 0.89 v.s. 0.81, p < 0.05) and RC models with 3 mm boundary extension yielded the best performance compared to those with other sizes (AUC = 0.89). Combining with radiologists' reading, the 3mm-boundary RC model achieved a sensitivity of 0.871 and negative predictive value of 0.937 with similar accuracy of 0.843 in predicting malignancy. CONCLUSIONS: The machine learning model integrating intra- and peri-calcification regions on CEM has the potential to aid radiologists' performance in predicting malignancy of suspicious breast calcifications.


Asunto(s)
Neoplasias de la Mama , Mama , Calcinosis , Medios de Contraste , Aprendizaje Automático , Mamografía , Humanos , Mamografía/métodos , Femenino , Calcinosis/diagnóstico por imagen , Neoplasias de la Mama/diagnóstico por imagen , Persona de Mediana Edad , Diagnóstico Diferencial , Mama/diagnóstico por imagen , Adulto , Anciano , Interpretación de Imagen Radiográfica Asistida por Computador/métodos
11.
Nat Commun ; 15(1): 1131, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326351

RESUMEN

Early and accurate diagnosis of focal liver lesions is crucial for effective treatment and prognosis. We developed and validated a fully automated diagnostic system named Liver Artificial Intelligence Diagnosis System (LiAIDS) based on a diverse sample of 12,610 patients from 18 hospitals, both retrospectively and prospectively. In this study, LiAIDS achieved an F1-score of 0.940 for benign and 0.692 for malignant lesions, outperforming junior radiologists (benign: 0.830-0.890, malignant: 0.230-0.360) and being on par with senior radiologists (benign: 0.920-0.950, malignant: 0.550-0.650). Furthermore, with the assistance of LiAIDS, the diagnostic accuracy of all radiologists improved. For benign and malignant lesions, junior radiologists' F1-scores improved to 0.936-0.946 and 0.667-0.680 respectively, while seniors improved to 0.950-0.961 and 0.679-0.753. Additionally, in a triage study of 13,192 consecutive patients, LiAIDS automatically classified 76.46% of patients as low risk with a high NPV of 99.0%. The evidence suggests that LiAIDS can serve as a routine diagnostic tool and enhance the diagnostic capabilities of radiologists for liver lesions.


Asunto(s)
Inteligencia Artificial , Neoplasias Hepáticas , Humanos , Estudios Retrospectivos , Radiólogos , Neoplasias Hepáticas/diagnóstico por imagen
12.
Life Sci ; 336: 122347, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38103728

RESUMEN

AIMS: The increasing resistance to anti-seizure medications (ASMs) and the ambiguous mechanisms of epilepsy highlight the pressing demand for the discovery of pioneering lead compounds. Berberine (BBR) has received significant attention in recent years within the field of chronic metabolic disorders. However, the reports on the treatment of epilepsy with BBR are not systematic and the mechanism remains unclear. MAIN METHODS: In this study, the seizure behaviors of mice were recorded following subcutaneous injection of pentetrazol (PTZ). Non-targeted metabolomics was used to analyze the serum metabolites based on ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). Meanwhile, multivariate statistical methods were used for metabolite identification and pathway analysis. Furthermore, network pharmacology, molecular docking, and quantitative real-time PCR assay were used for the target identification. KEY FINDINGS: BBR had anti-seizure effects on PTZ-induced seizure mice after long-term treatment. Tryptophan metabolism and phenylalanine metabolism were involved in regulating the therapeutic effects of BBR. SIGNIFICANCE: This study reveals the potential mechanism of BBR for epilepsy treatment based on non-targeted metabolomics and network pharmacology, which provides evidence for uncovering the pathogenesis of epilepsy, suggesting that BBR is a potential lead compound for anti-epileptic treatment.


Asunto(s)
Berberina , Epilepsia , Ratones , Animales , Berberina/farmacología , Berberina/uso terapéutico , Berberina/metabolismo , Farmacología en Red , Simulación del Acoplamiento Molecular , Metabolómica/métodos , Pentilenotetrazol/toxicidad , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico
13.
Radiol Med ; 129(2): 229-238, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38108979

RESUMEN

BACKGROUND: The accurate identification and evaluation of lymph nodes by CT images is of great significance for disease diagnosis, treatment, and prognosis. PURPOSE: To assess the lymph nodes' segmentation, size, and station by artificial intelligence (AI) for unenhanced chest CT images and evaluate its value in clinical scenarios. MATERIAL AND METHODS: This retrospective study proposed an end-to-end Lymph Nodes Analysis System (LNAS) consisting of three models: the Lymph Node Segmentation model (LNS), the Mediastinal Organ Segmentation model (MOS), and the Lymph Node Station Registration model (LNR). We selected a healthy chest CT image as the template image and annotated 14 lymph node station masks according to the IASLC to build the lymph node station mapping template. The exact contours and stations of the lymph nodes were annotated by two junior radiologists and reviewed by a senior radiologist. Patients aged 18 and above, who had undergone unenhanced chest CT and had at least one suspicious enlarged mediastinal lymph node in imaging reports, were included. Exclusions were patients who had thoracic surgeries in the past 2 weeks or artifacts on CT images affecting lymph node observation by radiologists. The system was trained on 6725 consecutive chest CTs that from Tianjin Medical University General Hospital, among which 6249 patients had suspicious enlarged mediastinal lymph nodes. A total of 519 consecutive chest CTs from Qilu Hospital of Shandong University (Qingdao) were used for external validation. The gold standard for each CT was determined by two radiologists and reviewed by one senior radiologist. RESULTS: The patient-level sensitivity of the LNAS system reached of 93.94% and 92.89% in internal and external test dataset, respectively. And the lesion-level sensitivity (recall) reached 89.48% and 85.97% in internal and external test dataset. For man-machine comparison, AI significantly apparently shortened the average reading time (p < 0.001) and had better lesion-level and patient-level sensitivities. CONCLUSION: AI improved the sensitivity lymph node segmentation by radiologists with an advantage in reading time.


Asunto(s)
Inteligencia Artificial , Aprendizaje Profundo , Humanos , Estudios Retrospectivos , Ganglios Linfáticos/diagnóstico por imagen , Ganglios Linfáticos/patología , Tomografía Computarizada por Rayos X/métodos
14.
J Thorac Dis ; 15(10): 5475-5484, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37969262

RESUMEN

Background: This study assessed the diagnostic performance of a deep learning (DL)-based model for differentiating malignant subcentimeter (≤10 mm) solid pulmonary nodules (SSPNs) from benign ones in computed tomography (CT) images compared against radiologists with 10 and 15 years of experience in thoracic imaging (medium-senior seniority). Methods: Overall, 200 SSPNs (100 benign and 100 malignant) were retrospectively collected. Malignancy was confirmed by pathology, and benignity was confirmed by follow-up or pathology. CT images were fed into the DL model to obtain the probability of malignancy (range, 0-100%) for each nodule. According to the diagnostic results, enrolled nodules were classified into benign, malignant, or indeterminate. The accuracy and diagnostic composition of the model were compared with those of the radiologists using the McNemar-Bowker test. Enrolled nodules were divided into 3-6-, 6-8-, and 8-10-mm subgroups. For each subgroup, the diagnostic results of the model were compared with those of the radiologists. Results: The accuracy of the DL model, in differentiating malignant and benign SSPNs, was significantly higher than that of the radiologists (71.5% vs. 38.5%, P<0.001). The DL model reported more benign or malignant deterministic results and fewer indeterminate results. In subgroup analysis of nodule size, the DL model also yielded higher performance in comparison with that of the radiologists, providing fewer indeterminate results. The accuracy of the two methods in the 3-6-, 6-8-, and 8-10-mm subgroups was 75.5% vs. 28.3% (P<0.001), 62.0% vs. 28.2% (P<0.001), and 77.6% vs. 55.3% (P=0.001), respectively, and the indeterminate results were 3.8% vs. 66.0%, 8.5% vs. 66.2%, and 2.6% vs. 35.5% (all P<0.001), respectively. Conclusions: The DL-based method yielded higher performance in comparison with that of the radiologists in differentiating malignant and benign SSPNs. This DL model may reduce uncertainty in diagnosis and improve diagnostic accuracy, especially for SSPNs smaller than 8 mm.

15.
Signal Transduct Target Ther ; 8(1): 416, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37907497

RESUMEN

There have been hundreds of millions of cases of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the growing population of recovered patients, it is crucial to understand the long-term consequences of the disease and management strategies. Although COVID-19 was initially considered an acute respiratory illness, recent evidence suggests that manifestations including but not limited to those of the cardiovascular, respiratory, neuropsychiatric, gastrointestinal, reproductive, and musculoskeletal systems may persist long after the acute phase. These persistent manifestations, also referred to as long COVID, could impact all patients with COVID-19 across the full spectrum of illness severity. Herein, we comprehensively review the current literature on long COVID, highlighting its epidemiological understanding, the impact of vaccinations, organ-specific sequelae, pathophysiological mechanisms, and multidisciplinary management strategies. In addition, the impact of psychological and psychosomatic factors is also underscored. Despite these crucial findings on long COVID, the current diagnostic and therapeutic strategies based on previous experience and pilot studies remain inadequate, and well-designed clinical trials should be prioritized to validate existing hypotheses. Thus, we propose the primary challenges concerning biological knowledge gaps and efficient remedies as well as discuss the corresponding recommendations.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Síndrome Post Agudo de COVID-19 , Evaluación de Resultado en la Atención de Salud
16.
Quant Imaging Med Surg ; 13(10): 6424-6433, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37869340

RESUMEN

Background: Extremities fractures are a leading cause of death and disability, especially in the elderly. Avulsion fracture are also the most commonly missed diagnosis, and delayed diagnosis leads to higher litigation rates. Therefore, this study evaluates the diagnostic efficiency of the artificial intelligence (AI) model before and after optimization based on computed tomography (CT) images and then compares it with that of radiologists, especially for avulsion fractures. Methods: The digital X-ray photography [digital radiography (DR)] and CT images of adult limb trauma in our hospital from 2017 to 2020 were retrospectively collected, with or without 1 or more fractures of the shoulder, elbow, wrist, hand, hip, knee, ankle, and foot. Labeling of the fracture referred to the visualization of the fracture on the corresponding CT images. After training the pre-optimized AI model, the diagnostic performance of the pre-optimized AI, optimized AI model, and the initial radiological reports were evaluated. For the lesion level, the detection rate of avulsion and non-avulsion fractures was analyzed, whereas for the case level, the accuracy, sensitivity, and specificity were compared among them. Results: The total datasets (1,035 cases) were divided into a training set (n=675), a validation set (n=169), and a test set (n=191) in a balanced joint distribution. At the lesion level, the detection rates of avulsion fracture (57.89% vs. 35.09%, P=0.004) and non-avulsion fracture (85.64% vs. 71.29%, P<0.001) by the optimized AI were significantly higher than that by pre-optimized AI. The average precision (AP) of the optimized AI model for all lesions was higher than that of pre-optimized AI model (0.582 vs. 0.425). The detection rate of avulsion fracture by the optimized AI model was significantly higher than that by radiologists (57.89% vs. 29.82%, P=0.002). For the non-avulsion fracture, there was no significant difference of detection rate between the optimized AI model and radiologists (P=0.853). At the case level, the accuracy (86.40% vs. 71.93%, P<0.001) and sensitivity (87.29% vs. 73.48%, P<0.001) of the optimized AI were significantly higher than those of the pre-optimized AI model. There was no statistical difference in accuracy, sensitivity, and specificity between the optimized AI model and the radiologists (P>0.05). Conclusions: The optimized AI model improves the diagnostic efficacy in detecting extremity fractures on radiographs, and the optimized AI model is significantly better than radiologists in detecting avulsion fractures, which may be helpful in the clinical practice of orthopedic emergency.

17.
IEEE Trans Image Process ; 32: 5580-5594, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37782617

RESUMEN

Compared to unsupervised domain adaptation, semi-supervised domain adaptation (SSDA) aims to significantly improve the classification performance and generalization capability of the model by leveraging the presence of a small amount of labeled data from the target domain. Several SSDA approaches have been developed to enable semantic-aligned feature confusion between labeled (or pseudo labeled) samples across domains; nevertheless, owing to the scarcity of semantic label information of the target domain, they were arduous to fully realize their potential. In this study, we propose a novel SSDA approach named Graph-based Adaptive Betweenness Clustering (G-ABC) for achieving categorical domain alignment, which enables cross-domain semantic alignment by mandating semantic transfer from labeled data of both the source and target domains to unlabeled target samples. In particular, a heterogeneous graph is initially constructed to reflect the pairwise relationships between labeled samples from both domains and unlabeled ones of the target domain. Then, to degrade the noisy connectivity in the graph, connectivity refinement is conducted by introducing two strategies, namely Confidence Uncertainty based Node Removal and Prediction Dissimilarity based Edge Pruning. Once the graph has been refined, Adaptive Betweenness Clustering is introduced to facilitate semantic transfer by using across-domain betweenness clustering and within-domain betweenness clustering, thereby propagating semantic label information from labeled samples across domains to unlabeled target data. Extensive experiments on three standard benchmark datasets, namely DomainNet, Office-Home, and Office-31, indicated that our method outperforms previous state-of-the-art SSDA approaches, demonstrating the superiority of the proposed G-ABC algorithm.

18.
Clocks Sleep ; 5(3): 552-565, 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37754354

RESUMEN

Poor sleep is a major public health problem with implications for a wide range of critical health outcomes. Insomnia and sleep apnoea are the two most common causes of poor sleep, and recent studies have shown that these disorders frequently co-occur. Comorbid insomnia and sleep apnoea can substantially impair quality of life and increase the overall risk of mortality. However, the causal and physiological links between sleep apnoea and insomnia are unclear. It is also unknown whether having a higher risk for one condition can increase the risk of developing the other. Here, we investigated links between sleep apnoea and insomnia in a British population using a combination of self-reported questionnaires and causal inference. We found that 54.3% of the cohort had moderate insomnia, 9.4% had moderate sleep apnoea, and that 6.2% scored high for both conditions. Importantly, having a higher risk of sleep apnoea was associated with a higher risk of insomnia and vice versa. To determine the causal directionality between sleep apnoea and insomnia, we used Mendelian randomisation and found evidence that sleep apnoea could cause insomnia, but not the reverse. To elucidate how both sleep apnoea and insomnia were linked to each other, we looked at the behavioural markers of poor sleep. We found that feeling fatigued after sleeping and having noticeable sleep problems were linked to a higher burden of both sleep apnoea and insomnia. In conclusion, our results show that sleep apnoea increases the risk of developing insomnia, and both conditions can result in fatigue. We highlight the importance of considering and treating the symptoms of both conditions.

19.
Biol Direct ; 18(1): 43, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37528429

RESUMEN

Antipsychotic drugs are the mainstay of treatment for schizophrenia and provide adjunct therapies for other prevalent psychiatric conditions, including bipolar disorder and major depressive disorder. However, they also induce debilitating extrapyramidal syndromes (EPS), such as Parkinsonism, in a significant minority of patients. The majority of antipsychotic drugs function as dopamine receptor antagonists in the brain while the most recent 'third'-generation, such as aripiprazole, act as partial agonists. Despite showing good clinical efficacy, these newer agents are still associated with EPS in ~ 5 to 15% of patients. However, it is not fully understood how these movement disorders develop. Here, we combine clinically-relevant drug concentrations with mutliscale model systems to show that aripiprazole and its primary active metabolite induce mitochondrial toxicity inducing robust declines in cellular ATP and viability. Aripiprazole, brexpiprazole and cariprazine were shown to directly inhibit respiratory complex I through its ubiquinone-binding channel. Importantly, all three drugs induced mitochondrial toxicity in primary embryonic mouse neurons, with greater bioenergetic inhibition in ventral midbrain neurons than forebrain neurons. Finally, chronic feeding with aripiprazole resulted in structural damage to mitochondria in the brain and thoracic muscle of adult Drosophila melanogaster consistent with locomotor dysfunction. Taken together, we show that antipsychotic drugs acting as partial dopamine receptor agonists exhibit off-target mitochondrial liabilities targeting complex I.


Asunto(s)
Antipsicóticos , Trastorno Depresivo Mayor , Animales , Ratones , Aripiprazol/farmacología , Aripiprazol/uso terapéutico , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Drosophila melanogaster , Transporte de Electrón
20.
Artículo en Inglés | MEDLINE | ID: mdl-37467083

RESUMEN

Modeling 3D avatars benefits various application scenarios such as AR/VR, gaming, and filming. Character faces contribute significant diversity and vividity as a vital component of avatars. However, building 3D character face models usually requires a heavy workload with commercial tools, even for experienced artists. Various existing sketch-based tools fail to support amateurs in modeling diverse facial shapes and rich geometric details. In this paper, we present SketchMetaFace - a sketching system targeting amateur users to model high-fidelity 3D faces in minutes. We carefully design both the user interface and the underlying algorithm. First, curvature-aware strokes are adopted to better support the controllability of carving facial details. Second, considering the key problem of mapping a 2D sketch map to a 3D model, we develop a novel learning-based method termed "Implicit and Depth Guided Mesh Modeling" (IDGMM). It fuses the advantages of mesh, implicit, and depth representations to achieve high-quality results with high efficiency. In addition, to further support usability, we present a coarse-to-fine 2D sketching interface design and a data-driven stroke suggestion tool. User studies demonstrate the superiority of our system over existing modeling tools in terms of the ease to use and visual quality of results. Experimental analyses also show that IDGMM reaches a better trade-off between accuracy and efficiency.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA