Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38758146

ABSTRACT

Objective: The current study was performed to assess the effectiveness of detailed operating room quality care on the quality of operating room care and patient satisfaction. Methods: A total of 102 patients who underwent surgery in Union Hospital, Tongji Medical College, Huazhong University of Science and Technology between October 2020 and April 2022 were recruited and assigned to receive either conventional operating room care (conventional group) or detailed operating room quality care (quality group), with 51 cases in each group. Outcome measures for the evaluation of the detailed quality care included quality of operating room care, safe operation, incidence of errors in instrument preparation, loss of parts, incidence of intraoperative adverse reactions, and patient satisfaction. Results: Patients who received quality care showed higher scores for information acquisition ability, communication ability, standardization of nursing process, and professionalism of nursing service than those who received conventional care (P = .021, .032, .003, .043). Detailed operating room quality care resulted in significantly higher standardization of anesthesia disinfection, promptness of instrument preparation, instrument and equipment management, effectiveness of auxiliary cooperation, and standardization of medical records scores versus conventional care (P = .004, .022, .036, .004, .002). Detailed operating room quality care was associated with a lower incidence of instrument preparation errors, lost parts, and intraoperative adverse reactions than conventional care (P < .05). Patients were more satisfied with quality care (49/51, 96.1%) than with conventional care (39/51, 76.5%) (P = .004). Conclusion: Detailed operating room quality care can significantly improve patient satisfaction, enhance the quality of operating room care and safe operation, and reduce the risk of instrument preparation errors, lost parts, and intraoperative complications in the operating room.

2.
Adv Sci (Weinh) ; 11(15): e2305701, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38348590

ABSTRACT

Phenylketonuria (PKU) is the most common inherited metabolic disease in humans. Clinical screening of newborn heel blood samples for PKU is costly and time-consuming because it requires multiple procedures, like isotope labeling and derivatization, and PKU subtype identification requires an additional urine sample. Delayed diagnosis of PKU, or subtype identification can result in mental disability. Here, plasmonic silver nanoshells are used for laser desorption/ionization mass spectrometry (MS) detection of PKU with label-free assay by recognizing metabolic profile in dried blood spot (DBS) samples. A total of 1100 subjects are recruited and each DBS sample can be processed in seconds. This platform achieves PKU screening with a sensitivity of 0.985 and specificity of 0.995, which is comparable to existing clinical liquid chromatography MS (LC-MS) methods. This method can process 360 samples per hour, compared with the LC-MS method which processes only 30 samples per hour. Moreover, this assay enables precise identification of PKU subtypes without the need for a urine sample. It is demonstrated that this platform enables high-performance and fast, low-cost PKU screening and subtype identification. This approach might be suitable for the detection of other clinically relevant biomarkers in blood or other clinical samples.


Subject(s)
Phenylketonurias , Infant, Newborn , Humans , Phenylketonurias/diagnosis , Phenylketonurias/metabolism , Liquid Chromatography-Mass Spectrometry , Metabolome
3.
Adv Sci (Weinh) ; 11(2): e2304610, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37953381

ABSTRACT

Metabolic fingerprints in serum characterize diverse diseases for diagnostics and biomarker discovery. The identification of systemic lupus erythematosus (SLE) by serum metabolic fingerprints (SMFs) will facilitate precision medicine in SLE in an early and designed manner. Here, a discovery cohort of 731 individuals including 357 SLE patients and 374 healthy controls (HCs), and a validation cohort of 184 individuals (SLE/HC, 91/93) are constructed. Each SMF is directly recorded by nano-assisted laser desorption/ionization mass spectrometry (LDI MS) within 1 minute using 1 µL of native serum, which contains 908 mass to charge features. Sparse learning of SMFs achieves the SLE identification with sensitivity/specificity and area-under-the-curve (AUC) up to 86.0%/92.0% and 0.950 for the discovery cohort. For the independent validation cohort, it exhibits no performance loss by affording the sensitivity/specificity and AUC of 89.0%/100.0% and 0.992. Notably, a metabolic biomarker panel is screened out from the SMFs, demonstrating the unique metabolic pattern of SLE patients different from both HCs and rheumatoid arthritis patients. In conclusion, SMFs characterize SLE by revealing its unique metabolic pattern. Different regulation of small molecule metabolites contributes to the precise diagnosis of autoimmune disease and further exploration of the pathogenic mechanisms.


Subject(s)
Arthritis, Rheumatoid , Lupus Erythematosus, Systemic , Humans , Biomarkers , Lupus Erythematosus, Systemic/diagnosis , Sensitivity and Specificity
4.
Small ; 19(7): e2206349, 2023 02.
Article in English | MEDLINE | ID: mdl-36470664

ABSTRACT

Infection classification is the key for choosing the proper treatment plans. Early determination of the causative agents is critical for disease control. Host responses analysis can detect variform and sensitive host inflammatory responses to ascertain the presence and type of the infection. However, traditional host-derived inflammatory indicators are insufficient for clinical infection classification. Fingerprints-based omic analysis has attracted increasing attention globally for analyzing the complex host systemic immune response. A single type of fingerprints is not applicable for infection classification (area under curve (AUC) of 0.550-0.617). Herein, an infection classification platform based on deep learning of dual plasma fingerprints (DPFs-DL) is developed. The DPFs with high reproducibility (coefficient of variation <15%) are obtained at low sample consumption (550 nL native plasma) using inorganic nanoparticle and organic matrix assisted laser desorption/ionization mass spectrometry. A classifier (DPFs-DL) for viral versus bacterial infection discrimination (AUC of 0.775) and coronavirus disease 2019 (COVID-2019) diagnosis (AUC of 0.917) is also built. Furthermore, a metabolic biomarker panel of two differentially regulated metabolites, which may serve as potential biomarkers for COVID-19 management (AUC of 0.677-0.883), is constructed. This study will contribute to the development of precision clinical care for infectious diseases.


Subject(s)
COVID-19 , Deep Learning , Humans , Reproducibility of Results , COVID-19/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Biomarkers
5.
Adv Sci (Weinh) ; 9(34): e2203786, 2022 12.
Article in English | MEDLINE | ID: mdl-36257825

ABSTRACT

Identification of novel non-invasive biomarkers is critical for the early diagnosis of lung adenocarcinoma (LUAD), especially for the accurate classification of pulmonary nodule. Here, a multiplexed assay is developed on an optimized nanoparticle-based laser desorption/ionization mass spectrometry platform for the sensitive and selective detection of serum metabolic fingerprints (SMFs). Integrative SMFs based multi-modal platforms are constructed for the early detection of LUAD and the classification of pulmonary nodule. The dual modal model, metabolic fingerprints with protein tumor marker neural network (MP-NN), integrating SMFs with protein tumor marker carcinoembryonic antigen (CEA) via deep learning, shows superior performance compared with the single modal model Met-NN (p < 0.001). Based on MP-NN, the tri modal model MPI-RF integrating SMFs, tumor marker CEA, and image features via random forest demonstrates significantly higher performance than the clinical models (Mayo Clinic and Veterans Affairs) and the image artificial intelligence in pulmonary nodule classification (p < 0.001). The developed platforms would be promising tools for LUAD screening and pulmonary nodule management, paving the conceptual and practical foundation for the clinical application of omics tools.


Subject(s)
Adenocarcinoma of Lung , Artificial Intelligence , United States , Humans , United States Government Agencies , Adenocarcinoma of Lung/diagnosis , Early Diagnosis , Biomarkers, Tumor
6.
Proc Natl Acad Sci U S A ; 119(12): e2122245119, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35302894

ABSTRACT

High-performance metabolic analysis is emerging in the diagnosis and prognosis of breast cancer (BrCa). Still, advanced tools are in demand to deliver the application potentials of metabolic analysis. Here, we used fast nanoparticle-enhanced laser desorption/ionization mass spectrometry (NPELDI-MS) to record serum metabolic fingerprints (SMFs) of BrCa in seconds, achieving high reproducibility and low consumption of direct serum detection without treatment. Subsequently, machine learning of SMFs generated by NPELDI-MS functioned as an efficient readout to distinguish BrCa from non-BrCa with an area under the curve of 0.948. Furthermore, a metabolic prognosis scoring system was constructed using SMFs with effective prediction performance toward BrCa (P < 0.005). Finally, we identified a biomarker panel of seven metabolites that were differentially enriched in BrCa serum and their related pathways. Together, our findings provide an efficient serum metabolic tool to characterize BrCa and highlight certain metabolic signatures as potential diagnostic and prognostic factors of diseases including but not limited to BrCa.


Subject(s)
Breast Neoplasms , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Female , Humans , Mass Spectrometry/methods , Prognosis , Reproducibility of Results
7.
ACS Nano ; 16(2): 2852-2865, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35099942

ABSTRACT

Chemotherapy is a primary cancer treatment strategy, the monitoring of which is critical to enhancing the survival rate and quality of life of cancer patients. However, current chemotherapy monitoring mainly relies on imaging tools with inefficient sensitivity and radiation invasiveness. Herein, we develop the bowl-shaped submicroreactor chip of Au-loaded 3-aminophenol formaldehyde resin (denoted as APF-bowl&Au) with a specifically designed structure and Au loading content. The obtained APF-bowl&Au, used as the matrix of laser desorption/ionization mass spectrometry (LDI MS), possesses an enhanced localized electromagnetic field for strengthened small metabolite detection. The APF-bowl&Au enables the extraction of serum metabolic fingerprints (SMFs), and machine learning of the SMFs achieves chemotherapy monitoring of ovarian cancer with area-under-the-curve (AUC) of 0.81-0.98. Furthermore, a serum metabolic biomarker panel is preliminarily identified, exhibiting gradual changes as the chemotherapy cycles proceed. This work provides insights into the development of nanochips and contributes to a universal detection platform for chemotherapy monitoring.


Subject(s)
Quality of Life , Serum , Humans , Lasers , Polymers , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
8.
Adv Sci (Weinh) ; 8(18): e2101333, 2021 09.
Article in English | MEDLINE | ID: mdl-34323397

ABSTRACT

Although mass spectrometry (MS) of metabolites has the potential to provide real-time monitoring of patient status for diagnostic purposes, the diagnostic application of MS is limited due to sample treatment and data quality/reproducibility. Here, the generation of a deep stabilizer for ultra-fast, label-free MS detection and the application of this method for serum metabolic diagnosis of coronary heart disease (CHD) are reported. Nanoparticle-assisted laser desorption/ionization-MS is used to achieve direct metabolic analysis of trace unprocessed serum in seconds. Furthermore, a deep stabilizer is constructed to map native MS results to high-quality results obtained by established methods. Finally, using the newly developed protocol and diagnosis variation characteristic surface to characterize sensitivity/specificity and variation, CHD is diagnosed with advanced accuracy in a high-throughput/speed manner. This work advances design of metabolic analysis tools for disease detection as it provides a direct label-free, ultra-fast, and stabilized platform for future protocol development in clinics.


Subject(s)
Coronary Disease/blood , Coronary Disease/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Coronary Disease/metabolism , Humans , Nanoparticles , Reproducibility of Results , Sensitivity and Specificity , Time
9.
Adv Mater ; 33(17): e2007978, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33742513

ABSTRACT

Gastric cancer (GC) is a multifactorial process, accompanied by alterations in metabolic pathways. Non-invasive metabolic profiling facilitates GC diagnosis at early stage leading to an improved prognostic outcome. Herein, mesoporous PdPtAu alloys are designed to characterize the metabolic profiles in human blood. The elemental composition is optimized with heterogeneous surface plasmonic resonance, offering preferred charge transfer for photoinduced desorption/ionization and enhanced photothermal conversion for thermally driven desorption. The surface structure of PdPtAu is further tuned with controlled mesopores, accommodating metabolites only, rather than large interfering compounds. Consequently, the optimized PdPtAu alloy yields direct metabolic fingerprints by laser desorption/ionization mass spectrometry in seconds, consuming 500 nL of native plasma. A distinct metabolic phenotype is revealed for early GC by sparse learning, resulting in precise GC diagnosis with an area under the curve of 0.942. It is envisioned that the plasmonic alloy will open up a new era of minimally invasive blood analysis to improve the surveillance of cancer patients in the clinical setting.


Subject(s)
Alloys , Phenotype , Stomach Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Adv Mater ; 32(23): e2000906, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32342553

ABSTRACT

Diagnostics is the key in screening and treatment of cancer. As an emerging tool in precision medicine, metabolic analysis detects end products of pathways, and thus is more distal than proteomic/genetic analysis. However, metabolic analysis is far from ideal in clinical diagnosis due to the sample complexity and metabolite abundance in patient specimens. A further challenge is real-time and accurate tracking of treatment effect, e.g., radiotherapy. Here, Pd-Au synthetic alloys are reported for mass-spectrometry-based metabolic fingerprinting and analysis, toward medulloblastoma diagnosis and radiotherapy evaluation. A core-shell structure is designed using magnetic core particles to support Pd-Au alloys on the surface. Optimized synthetic alloys enhance the laser desorption/ionization efficacy and achieve direct detection of 100 nL of biofluids in seconds. Medulloblastoma patients are differentiated from healthy controls with average diagnostic sensitivity of 94.0%, specificity of 85.7%, and accuracy of 89.9%, by machine learning of metabolic fingerprinting. Furthermore, the radiotherapy process of patients is monitored and a preliminary panel of serum metabolite biomarkers is identified with gradual changes. This work will lead to the application-driven development of novel materials with tailored structural design and establishment of new protocols for precision medicine in near future.


Subject(s)
Alloys/metabolism , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/radiotherapy , Medulloblastoma/diagnosis , Medulloblastoma/radiotherapy , Metabolomics , Alloys/chemistry , Cell Line, Tumor , Cerebellar Neoplasms/blood , Cerebellar Neoplasms/metabolism , Gold/chemistry , Humans , Machine Learning , Medulloblastoma/blood , Medulloblastoma/metabolism , Palladium/chemistry , Treatment Outcome
11.
Angew Chem Int Ed Engl ; 59(4): 1703-1710, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31829520

ABSTRACT

Metabolic fingerprints of biofluids encode diverse diseases and particularly urine detection offers complete non-invasiveness for diagnostics of the future. Present urine detection affords unsatisfactory performance and requires advanced materials to extract molecular information, due to the limited biomarkers and high sample complexity. Herein, we report plasmonic polymer@Ag for laser desorption/ionization mass spectrometry (LDI-MS) and sparse-learning-based metabolic diagnosis of kidney diseases. Using only 1 µL of urine without enrichment or purification, polymer@Ag afforded urine metabolic fingerprints (UMFs) by LDI-MS in seconds. Analysis by sparse learning discriminated lupus nephritis from various other non-lupus nephropathies and controls. We combined UMFs with urine protein levels (UPLs) and constructed a new diagnostic model to characterize subtypes of kidney diseases. Our work guides urine-based diagnosis and leads to new personalized analytical tools for other diseases.


Subject(s)
Biomarkers, Tumor/urine , Kidney Diseases/urine , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...