Search Results Heading

MBRLSearchResults

mbrl.module.common.modules.added.book.to.shelf
Title added to your shelf!
View what I already have on My Shelf.
Oops! Something went wrong.
Oops! Something went wrong.
While trying to add the title to your shelf something went wrong :( Kindly try again later!
Are you sure you want to remove the book from the shelf?
Oops! Something went wrong.
Oops! Something went wrong.
While trying to remove the title from your shelf something went wrong :( Kindly try again later!
    Done
    Filters
    Reset
  • Discipline
      Discipline
      Clear All
      Discipline
  • Is Peer Reviewed
      Is Peer Reviewed
      Clear All
      Is Peer Reviewed
  • Series Title
      Series Title
      Clear All
      Series Title
  • Reading Level
      Reading Level
      Clear All
      Reading Level
  • Year
      Year
      Clear All
      From:
      -
      To:
  • More Filters
      More Filters
      Clear All
      More Filters
      Content Type
    • Item Type
    • Is Full-Text Available
    • Subject
    • Country Of Publication
    • Publisher
    • Source
    • Target Audience
    • Donor
    • Language
    • Place of Publication
    • Contributors
    • Location
23,417 result(s) for "Gao, Yan"
Sort by:
Smart IoT with the hybrid evolutionary method and image processing for tumor detection
The primary objective of modern healthcare systems is to enhance public health by providing efficient, reliable, and well-structured solutions. Improving patient satisfaction through tailored medical services has driven rapid advancements in healthcare, leading to increased competition and system complexity. However, the expansion of healthcare services introduces challenges such as high data volume, latency, response time constraints, and security vulnerabilities. To address these issues, fog computing offers an effective solution by processing data closer to end devices, reducing latency, and enabling real-time responses. This research proposes a robust brain tumor detection framework within a fog-based smart healthcare infrastructure. The process begins with data placement leveraging an improved evolutionary technique for Image Processing (HETS-IP) to optimize fog node placement based on key parameters such as energy efficiency and latency. Specifically, the Particle Swarm Optimization (PSO) algorithm is enhanced with a direct binary encoding technique, in which solutions are represented as binary strings, making it suitable for problems where decisions are discrete. This approach allows efficient optimization in binary decision spaces and improves adaptability for complex placement problems. Once data placement is committed, the tumor detection framework is performed directly at fog nodes to enhance real-time processing. This phase will begin with preprocessing, where a bilateral filter is applied to reduce noise while preserving critical edge details. Next, feature extraction is utilized to derive statistical texture features, which capture diagnostic information essential for distinguishing between tumor types. The process continues by classification using a deep Convolutional Neural Network (CNN) with sequential architecture to classify tumors. Simulation results demonstrate that HETS-IP outperforms traditional evolutionary algorithms, including Ant Colony Optimization (ACO), Genetic Algorithm-Simulated Annealing (GASA), and Genetic Algorithm (GA). On average, HETS-IP reduces energy consumption by 5%, 9%, and 14% and decreases makespan by 4%, 6%, and 11%, respectively. Additionally, the proposed approach achieves an accuracy of 97% and a precision of 96%, ensuring highly reliable brain tumor detection.
Deep transfer learning for reducing health care disparities arising from biomedical data inequality
As artificial intelligence (AI) is increasingly applied to biomedical research and clinical decisions, developing unbiased AI models that work equally well for all ethnic groups is of crucial importance to health disparity prevention and reduction. However, the biomedical data inequality between different ethnic groups is set to generate new health care disparities through data-driven, algorithm-based biomedical research and clinical decisions. Using an extensive set of machine learning experiments on cancer omics data, we find that current prevalent schemes of multiethnic machine learning are prone to generating significant model performance disparities between ethnic groups. We show that these performance disparities are caused by data inequality and data distribution discrepancies between ethnic groups. We also find that transfer learning can improve machine learning model performance for data-disadvantaged ethnic groups, and thus provides an effective approach to reduce health care disparities arising from data inequality among ethnic groups. Developing machine learning models that work equally well for all ethnic groups is of crucial importance to health disparity prevention and reduction. Here, using an extensive set of machine learning experiments on cancer omics data, the authors find that transfer learning can improve model performance for data-disadvantaged ethnic groups.
Antibiotics for cancer treatment: A double-edged sword
Various antibiotics have been used in the treatment of cancers, via their anti-proliferative, pro-apoptotic and anti-epithelial-mesenchymal-transition (EMT) capabilities. However, increasingly studies have indicated that antibiotics may also induce cancer generation by disrupting intestinal microbiota, which further promotes chronic inflammation, alters normal tissue metabolism, leads to genotoxicity and weakens the immune response to bacterial malnutrition, thereby adversely impacting cancer treatment. Despite the advent of high-throughput sequencing technology in recent years, the potential adverse effects of antibiotics on cancer treatments via causing microbial imbalance has been largely ignored. In this review, we discuss the double-edged sword of antibiotics in the field of cancer treatments, explore their potential mechanisms and provide solutions to reduce the potential negative effects of antibiotics.
Metal-organic framework membranes with single-atomic centers for photocatalytic CO2 and O2 reduction
The demand for sustainable energy has motivated the development of artificial photosynthesis. Yet the catalyst and reaction interface designs for directly fixing permanent gases (e.g. CO 2 , O 2 , N 2 ) into liquid fuels are still challenged by slow mass transfer and sluggish catalytic kinetics at the gas-liquid-solid boundary. Here, we report that gas-permeable metal-organic framework (MOF) membranes can modify the electronic structures and catalytic properties of metal single-atoms (SAs) to promote the diffusion, activation, and reduction of gas molecules (e.g. CO 2, O 2 ) and produce liquid fuels under visible light and mild conditions. With Ir SAs as active centers, the defect-engineered MOF (e.g. activated NH 2 -UiO-66) particles can reduce CO 2 to HCOOH with an apparent quantum efficiency (AQE) of 2.51% at 420 nm on the gas-liquid-solid reaction interface. With promoted gas diffusion at the porous gas-solid interfaces, the gas-permeable SA/MOF membranes can directly convert humid CO 2 gas into HCOOH with a near-unity selectivity and a significantly increased AQE of 15.76% at 420 nm. A similar strategy can be applied to the photocatalytic O 2 -to-H 2 O 2 conversions, suggesting the wide applicability of our catalyst and reaction interface designs. Photoreduction of permanent gas faces challenges in reactant diffusion and activation at the three-phase interface. Here the authors showed porous metal-organic framework membranes decorated by metal single atoms can boost the photoreduction of CO 2 and O 2 at the high-throughput gas-solid interface.
Lentivirus-mediated downregulation of α-synuclein reduces neuroinflammation and promotes functional recovery in rats with spinal cord injury
Background The prognosis of spinal cord injury (SCI) is closely related to secondary injury, which is dominated by neuroinflammation. There is evidence that α-synuclein aggregates after SCI and that inhibition of α-synuclein aggregation can improve the survival of neurons after SCI, but the mechanism is still unclear. This study was designed to investigate the effects of α-synuclein on neuroinflammation after SCI and to determine the underlying mechanisms. Method A T3 spinal cord contusion model was established in adult male Sprague-Dawley rats. An SNCA-shRNA-carrying lentivirus (LV-SNCA-shRNA) was injected into the injury site to block the expression of α-synuclein (forming the SCI+KD group), and the SCI and sham groups were injected with an empty vector. Basso-Beattie-Bresnahan (BBB) behavioural scores and footprint analysis were used to detect motor function. Inflammatory infiltration and myelin loss were measured in the spinal cord tissues of each group by haematoxylin-eosin (HE) and Luxol Fast Blue (LFB) staining, respectively. Immunohistochemistry, Western blot analysis, and RT-qPCR were used to analyse protein expression and transcription levels in the tissues. Immunofluorescence was used to determine the morphology and function of glial cells and the expression of matrix metalloproteinase-9 in the central canal of the spinal cord. Finally, peripheral serum cytokine levels were determined by enzyme-linked immunosorbent assay. Results Compared with the SCI group, the SCI+KD group exhibited reduced inflammatory infiltration, preserved myelin, and functional recovery. Specifically, the early arrest of α-synuclein inhibited the pro-inflammatory factors IL-1β, TNF-α, and IL-2 and increased the expression of the anti-inflammatory factors IL-10, TGF-β, and IL-4. The neuroinflammatory response was regulated by reduced proliferation of Iba1+ microglia/macrophages and promotion of the shift of M1-polarized Iba1+/iNOS+ microglia/macrophages to M2-polarized Iba1+/Arg1+ microglia/macrophages after injury. In addition, compared with the SCI group, the SCI+KD group also exhibited a smaller microglia/astrocyte (Iba1/GFAP) immunostaining area in the central canal, lower MMP-9 expression, and improved cerebrospinal barrier function. Conclusion Lentivirus-mediated downregulation of α-synuclein reduces neuroinflammation, improves blood-cerebrospinal barrier function, promotes functional recovery, reduces microglial activation, and promotes the polarization of M1 microglia/macrophages to an M2 phenotype to confer a neuroprotective immune microenvironment in rats with SCI.
Advances of Electrochemical and Electrochemiluminescent Sensors Based on Covalent Organic Frameworks
HighlightsCovalent organic frameworks (COFs) show enormous potential for building high-performance electrochemical sensors due to their high porosity, large specific surface areas, stable rigid topology, ordered structures, and tunable pore microenvironments.The basic properties, monomers, and general synthesis methods of COFs in the electroanalytical chemistry field are introduced, with special emphasis on their usages in the fabrication of chemical sensors, ions sensors, immunosensors, and aptasensors.The emerged COFs in the electrochemiluminescence realm are thoroughly covered along with their preliminary applications.Covalent organic frameworks (COFs), a rapidly developing category of crystalline conjugated organic polymers, possess highly ordered structures, large specific surface areas, stable chemical properties, and tunable pore microenvironments. Since the first report of boroxine/boronate ester-linked COFs in 2005, COFs have rapidly gained popularity, showing important application prospects in various fields, such as sensing, catalysis, separation, and energy storage. Among them, COFs-based electrochemical (EC) sensors with upgraded analytical performance are arousing extensive interest. In this review, therefore, we summarize the basic properties and the general synthesis methods of COFs used in the field of electroanalytical chemistry, with special emphasis on their usages in the fabrication of chemical sensors, ions sensors, immunosensors, and aptasensors. Notably, the emerged COFs in the electrochemiluminescence (ECL) realm are thoroughly covered along with their preliminary applications. Additionally, final conclusions on state-of-the-art COFs are provided in terms of EC and ECL sensors, as well as challenges and prospects for extending and improving the research and applications of COFs in electroanalytical chemistry.
Sulfur Homeostasis in Plants
Sulfur (S) is an essential macronutrient for plant growth and development. S is majorly absorbed as sulfate from soil, and is then translocated to plastids in leaves, where it is assimilated into organic products. Cysteine (Cys) is the first organic product generated from S, and it is used as a precursor to synthesize many S-containing metabolites with important biological functions, such as glutathione (GSH) and methionine (Met). The reduction of sulfate takes place in a two-step reaction involving a variety of enzymes. Sulfate transporters (SULTRs) are responsible for the absorption of SO42− from the soil and the transport of SO42− in plants. There are 12–16 members in the S transporter family, which is divided into five categories based on coding sequence homology and biochemical functions. When exposed to S deficiency, plants will alter a series of morphological and physiological processes. Adaptive strategies, including cis-acting elements, transcription factors, non-coding microRNAs, and phytohormones, have evolved in plants to respond to S deficiency. In addition, there is crosstalk between S and other nutrients in plants. In this review, we summarize the recent progress in understanding the mechanisms underlying S homeostasis in plants.
c-MYC mediates the crosstalk between breast cancer cells and tumor microenvironment
The MYC oncogenic family is dysregulated in diverse tumors which is generally linked to the poor prognosis of tumors. The members in MYC family are transcription factors which are responsible for the regulation of various genes expression. Among them, c-MYC is closely related to the progression of tumors. Furthermore, c-MYC aberrations is tightly associated with the prevalence of breast cancer. Tumor microenvironment (TME) is composed of many different types of cellular and non-cellular factors, mainly including cancer-associated fibroblasts, tumor-associated macrophages, vascular endothelial cells, myeloid-derived suppressor cells and immune cells, all of which can affect the diagnosis, prognosis, and therapeutic efficacy of breast cancer. Importantly, the biological processes occurred in TME, such as angiogenesis, immune evasion, invasion, migration, and the recruition of stromal and tumor-infiltrating cells are under the modulation of c-MYC. These findings indicated that c-MYC serves as a critical regulator of TME. Here, we aimed to summarize and review the relevant research, thus to clarify c-MYC is a key mediator between breast cancer cells and TME. DVLPdmoXAPbqrJeJ-uvGmg Video Abstract
Sirtuin5 contributes to colorectal carcinogenesis by enhancing glutaminolysis in a deglutarylation-dependent manner
Reversible post-translational modifications represent a mechanism to control tumor metabolism. Here we show that mitochondrial Sirtuin5 (SIRT5), which mediates lysine desuccinylation, deglutarylation, and demalonylation, plays a role in colorectal cancer (CRC) glutamine metabolic rewiring. Metabolic profiling identifies that deletion of SIRT5 causes a marked decrease in 13 C-glutamine incorporation into tricarboxylic-acid (TCA) cycle intermediates and glutamine-derived non-essential amino acids. This reduces the building blocks required for rapid growth. Mechanistically, the direct interaction between SIRT5 and glutamate dehydrogenase 1 (GLUD1) causes deglutarylation and functional activation of GLUD1, a critical regulator of cellular glutaminolysis. Consistently, GLUD1 knockdown diminishes SIRT5-induced proliferation, both in vivo and in vitro. Clinically, overexpression of SIRT5 is significantly correlated with poor prognosis in CRC. Thus, SIRT5 supports the anaplerotic entry of glutamine into the TCA cycle in malignant phenotypes of CRC via activating GLUD1. Tumour metabolism can be controlled through post-translational modifications. Here the authors show that Sirtuin5 promotes glutaminolysis in colorectal cancer cells via glutamate dehydrogenase-1, a critical regulator of glutaminolysis, inducing its deglutarylation and functional activation.