Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Bioinformatics ; 40(3)2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38527901

RESUMEN

MOTIVATION: Many diseases, particularly cardiometabolic disorders, exhibit complex multimorbidities with one another. An intuitive way to model the connections between phenotypes is with a disease-disease network (DDN), where nodes represent diseases and edges represent associations, such as shared single-nucleotide polymorphisms (SNPs), between pairs of diseases. To gain further genetic understanding of molecular contributors to disease associations, we propose a novel version of the shared-SNP DDN (ssDDN), denoted as ssDDN+, which includes connections between diseases derived from genetic correlations with intermediate endophenotypes. We hypothesize that a ssDDN+ can provide complementary information to the disease connections in a ssDDN, yielding insight into the role of clinical laboratory measurements in disease interactions. RESULTS: Using PheWAS summary statistics from the UK Biobank, we constructed a ssDDN+ revealing hundreds of genetic correlations between diseases and quantitative traits. Our augmented network uncovers genetic associations across different disease categories, connects relevant cardiometabolic diseases, and highlights specific biomarkers that are associated with cross-phenotype associations. Out of the 31 clinical measurements under consideration, HDL-C connects the greatest number of diseases and is strongly associated with both type 2 diabetes and heart failure. Triglycerides, another blood lipid with known genetic causes in non-mendelian diseases, also adds a substantial number of edges to the ssDDN. This work demonstrates how association with clinical biomarkers can better explain the shared genetics between cardiometabolic disorders. Our study can facilitate future network-based investigations of cross-phenotype associations involving pleiotropy and genetic heterogeneity, potentially uncovering sources of missing heritability in multimorbidities. AVAILABILITY AND IMPLEMENTATION: The generated ssDDN+ can be explored at https://hdpm.biomedinfolab.com/ddn/biomarkerDDN.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/genética , Endofenotipos , Estudio de Asociación del Genoma Completo , Fenotipo , Enfermedades Cardiovasculares/genética , Biomarcadores , Polimorfismo de Nucleótido Simple , Predisposición Genética a la Enfermedad
2.
Bioinformatics ; 39(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36571484

RESUMEN

MOTIVATION: Understanding comorbidity is essential for disease prevention, treatment and prognosis. In particular, insight into which pairs of diseases are likely or unlikely to co-occur may help elucidate the potential relationships between complex diseases. Here, we introduce the use of an inter-disease interactivity network to discover/prioritize comorbidities. Specifically, we determine disease associations by accounting for the direction of effects of genetic components shared between diseases, and categorize those associations as synergistic or antagonistic. We further develop a comorbidity scoring algorithm to predict whether diseases are more or less likely to co-occur in the presence of a given index disease. This algorithm can handle networks that incorporate relationships with opposite signs. RESULTS: We finally investigate inter-disease associations among 427 phenotypes in UK Biobank PheWAS data and predict the priority of comorbid diseases. The predicted comorbidities were verified using the UK Biobank inpatient electronic health records. Our findings demonstrate that considering the interaction of phenotype associations might be helpful in better predicting comorbidity. AVAILABILITY AND IMPLEMENTATION: The source code and data of this study are available at https://github.com/dokyoonkimlab/DiseaseInteractiveNetwork. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Bancos de Muestras Biológicas , Programas Informáticos , Comorbilidad , Fenotipo
3.
Sensors (Basel) ; 24(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39001125

RESUMEN

In this paper, two orthogonally placed Vivaldi antennas with a septum-like polarizer to generate circular polarized (CP) waves are presented. Septum polarizers have garnered attention due to their simple structure and high quality of CP waves. While a typical septum polarizer has been applied to various types of waveguides, its applicability to the substrate integrated Vivaldi antenna is demonstrated here for the first time. A pulse train-shaped polarizer is used, which is placed on one of the two Vivaldi antennas. The contours of the polarizer are optimized using a genetic algorithm to provide an equal amplitude and 90° phase difference between the two orthogonal electric fields. In contrast to typical feed networks with a 90° phase shifter, any unwanted loss caused by an electronic circuit can be greatly mitigated. The antenna prototype was fabricated, and its radiation pattern and impedance matching were measured and compared to the simulated results.

4.
J Transl Med ; 21(1): 415, 2023 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-37365631

RESUMEN

BACKGROUND: Computational drug repurposing is crucial for identifying candidate therapeutic medications to address the urgent need for developing treatments for newly emerging infectious diseases. The recent COVID-19 pandemic has taught us the importance of rapidly discovering candidate drugs and providing them to medical and pharmaceutical experts for further investigation. Network-based approaches can provide repurposable drugs quickly by leveraging comprehensive relationships among biological components. However, in a case of newly emerging disease, applying a repurposing methods with only pre-existing knowledge networks may prove inadequate due to the insufficiency of information flow caused by the novel nature of the disease. METHODS: We proposed a network-based complementary linkage method for drug repurposing to solve the lack of incoming new disease-specific information in knowledge networks. We simulate our method under the controlled repurposing scenario that we faced in the early stage of the COVID-19 pandemic. First, the disease-gene-drug multi-layered network was constructed as the backbone network by fusing comprehensive knowledge database. Then, complementary information for COVID-19, containing data on 18 comorbid diseases and 17 relevant proteins, was collected from publications or preprint servers as of May 2020. We estimated connections between the novel COVID-19 node and the backbone network to construct a complemented network. Network-based drug scoring for COVID-19 was performed by applying graph-based semi-supervised learning, and the resulting scores were used to validate prioritized drugs for population-scale electronic health records-based medication analyses. RESULTS: The backbone networks consisted of 591 diseases, 26,681 proteins, and 2,173 drug nodes based on pre-pandemic knowledge. After incorporating the 35 entities comprised of complemented information into the backbone network, drug scoring screened top 30 potential repurposable drugs for COVID-19. The prioritized drugs were subsequently analyzed in electronic health records obtained from patients in the Penn Medicine COVID-19 Registry as of October 2021 and 8 of these were found to be statistically associated with a COVID-19 phenotype. CONCLUSION: We found that 8 of the 30 drugs identified by graph-based scoring on complemented networks as potential candidates for COVID-19 repurposing were additionally supported by real-world patient data in follow-up analyses. These results show that our network-based complementary linkage method and drug scoring algorithm are promising strategies for identifying candidate repurposable drugs when new emerging disease outbreaks.


Asunto(s)
COVID-19 , Humanos , COVID-19/epidemiología , Pandemias , Algoritmos , Proteínas , Reposicionamiento de Medicamentos/métodos
5.
Am J Obstet Gynecol ; 229(3): 298.e1-298.e19, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36933686

RESUMEN

BACKGROUND: Hypertensive disorders during pregnancy are associated with the risk of long-term cardiovascular disease after pregnancy, but it has not yet been determined whether genetic predisposition for hypertensive disorders during pregnancy can predict the risk for long-term cardiovascular disease. OBJECTIVE: This study aimed to evaluate the risk for long-term atherosclerotic cardiovascular disease according to polygenic risk scores for hypertensive disorders during pregnancy. STUDY DESIGN: Among UK Biobank participants, we included European-descent women (n=164,575) with at least 1 live birth. Participants were divided according to genetic risk categorized by polygenic risk scores for hypertensive disorders during pregnancy (low risk, score ≤25th percentile; medium risk, score 25th∼75th percentile; high risk, score >75th percentile), and were evaluated for incident atherosclerotic cardiovascular disease, defined as the new occurrence of one of the following: coronary artery disease, myocardial infarction, ischemic stroke, or peripheral artery disease. RESULTS: Among the study population, 2427 (1.5%) had a history of hypertensive disorders during pregnancy, and 8942 (5.6%) developed incident atherosclerotic cardiovascular disease after enrollment. Women with high genetic risk for hypertensive disorders during pregnancy had a higher prevalence of hypertension at enrollment. After enrollment, women with high genetic risk for hypertensive disorders during pregnancy had an increased risk for incident atherosclerotic cardiovascular disease, including coronary artery disease, myocardial infarction, and peripheral artery disease, compared with those with low genetic risk, even after adjustment for history of hypertensive disorders during pregnancy. CONCLUSION: High genetic risk for hypertensive disorders during pregnancy was associated with increased risk for atherosclerotic cardiovascular disease. This study provides evidence on the informative value of polygenic risk scores for hypertensive disorders during pregnancy in prediction of long-term cardiovascular outcomes later in life.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedad de la Arteria Coronaria , Hipertensión Inducida en el Embarazo , Infarto del Miocardio , Enfermedad Arterial Periférica , Embarazo , Humanos , Femenino , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Hipertensión Inducida en el Embarazo/epidemiología , Hipertensión Inducida en el Embarazo/genética , Factores de Riesgo , Infarto del Miocardio/epidemiología
6.
Br J Cancer ; 126(11): 1539-1547, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35249104

RESUMEN

BACKGROUND: Systemic inflammation is associated with survival outcomes in colon cancer. However, it is not well-known which systemic inflammatory marker is a powerful prognostic marker in patients with colon cancer. METHODS: A total of 4535 colon cancer patients were included in this study. We developed a novel prognostic index using a robust combination of seven systemic inflammation-associated blood features of the discovery set. The predictability and generality of the novel prognostic index were evaluated in the discovery, validation and replication sets. RESULTS: Among all combinations, the combination of albumin and monocyte count was the best candidate expression. The final formula of the proposed novel index is named the Prognostic Immune and Nutritional Index (PINI). The concordance index of PINI for overall and progression-free survival was the highest in the discovery, validation and replication sets compared to existing prognostic inflammatory markers. PINI was found to be a significant independent prognostic factor for both overall and progression-free survival. CONCLUSIONS: PINI is a novel prognostic index that has improved discriminatory power in colon cancer patients and appears to be superior to existing prognostic inflammatory markers. PINI can be utilised for decision-making regarding personalised treatment as the complement of the TNM staging system.


Asunto(s)
Neoplasias del Colon , Evaluación Nutricional , Humanos , Inflamación , Estadificación de Neoplasias , Pronóstico
7.
Sensors (Basel) ; 21(4)2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670691

RESUMEN

The development of biomedical devices benefits patients by offering real-time healthcare. In particular, pacemakers have gained a great deal of attention because they offer opportunities for monitoring the patient's vitals and biological statics in real time. One of the important factors in realizing real-time body-centric sensing is to establish a robust wireless communication link among the medical devices. In this paper, radio transmission and the optimal characteristics for impedance matching the medical telemetry of an implant are investigated. For radio transmission, an integral coupling formula based on 3D vector far-field patterns was firstly applied to compute the antenna coupling between two antennas placed inside and outside of the body. The formula provides the capability for computing the antenna coupling in the near-field and far-field region. In order to include the effects of human implantation, the far-field pattern was characterized taking into account a sphere enclosing an antenna made of human tissue. Furthermore, the characteristics of impedance matching inside the human body were studied by means of inherent wave impedances of electrical and magnetic dipoles. Here, we demonstrate that the implantation of a magnetic dipole is advantageous because it provides similar impedance characteristics to those of the human body.


Asunto(s)
Prótesis e Implantes , Telemetría , Impedancia Eléctrica , Electricidad , Humanos
8.
Bioinformatics ; 35(11): 1923-1930, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30335143

RESUMEN

SUMMARY: Immune diseases have a strong genetic component with Mendelian patterns of inheritance. While the tight association has been a major understanding in the underlying pathophysiology for the category of immune diseases, the common features of these diseases remain unclear. Based on the potential commonality among immune genes, we design Gene Ranker for key gene identification. Gene Ranker is a network-based gene scoring algorithm that initially constructs a backbone network based on protein interactions. Patient gene expression networks are added into the network. An add-on process screens the networks of weighted gene co-expression network analysis (WGCNA) on the samples of immune patients. Gene Ranker is disease-specific; however, any WGCNA network that passes the screening procedure can be added on. With the constructed network, it employs the semi-supervised learning for gene scoring. RESULTS: The proposed method was applied to immune diseases. Based on the resulting scores, Gene Ranker identified potential key genes in immune diseases. In scoring validation, an average area under the receiver operating characteristic curve of 0.82 was achieved, which is a significant increase from the reference average of 0.76. Highly ranked genes were verified through retrieval and review of 27 million PubMed literatures. As a typical case, 20 potential key genes in rheumatoid arthritis were identified: 10 were de facto genes and the remaining were novel. AVAILABILITY AND IMPLEMENTATION: Gene Ranker is available at http://www.alphaminers.net/GeneRanker/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Bases de Datos Genéticas , Redes Reguladoras de Genes , Genoma , Humanos , Curva ROC
9.
BMC Bioinformatics ; 20(Suppl 13): 383, 2019 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31337333

RESUMEN

BACKGROUND: Drug repurposing has been motivated to ameliorate low probability of success in drug discovery. For the recent decade, many in silico attempts have received primary attention as a first step to alleviate the high cost and longevity. Such study has taken benefits of abundance, variety, and easy accessibility of pharmaceutical and biomedical data. Utilizing the research friendly environment, in this study, we propose a network-based machine learning algorithm for drug repurposing. Particularly, we show a framework on how to construct a drug network, and how to strengthen the network by employing multiple/heterogeneous types of data. RESULTS: The proposed method consists of three steps. First, we construct a drug network from drug-target protein information. Then, the drug network is reinforced by utilizing drug-drug interaction knowledge on bioactivity and/or medication from literature databases. Through the enhancement, the number of connected nodes and the number of edges between them become more abundant and informative, which can lead to a higher probability of success of in silico drug repurposing. The enhanced network recommends candidate drugs for repurposing through drug scoring. The scoring process utilizes graph-based semi-supervised learning to determine the priority of recommendations. CONCLUSIONS: The drug network is reinforced in terms of the coverage and connections of drugs: the drug coverage increases from 4738 to 5442, and the drug-drug associations as well from 808,752 to 982,361. Along with the network enhancement, drug recommendation becomes more reliable: AUC of 0.89 was achieved lifted from 0.79. For typical cases, 11 recommended drugs were shown for vascular dementia: amantadine, conotoxin GV, tenocyclidine, cycloeucine, etc.


Asunto(s)
Reposicionamiento de Medicamentos/métodos , Preparaciones Farmacéuticas/química , Área Bajo la Curva , Interacciones Farmacológicas , Humanos , Preparaciones Farmacéuticas/metabolismo , Proteínas/química , Proteínas/metabolismo , Curva ROC , Aprendizaje Automático Supervisado
10.
BMC Bioinformatics ; 20(1): 576, 2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31722666

RESUMEN

BACKGROUND: The recent advances in human disease network have provided insights into establishing the relationships between the genotypes and phenotypes of diseases. In spite of the great progress, it yet remains as only a map of topologies between diseases, but not being able to be a pragmatic diagnostic/prognostic tool in medicine. It can further evolve from a map to a translational tool if it equips with a function of scoring that measures the likelihoods of the association between diseases. Then, a physician, when practicing on a patient, can suggest several diseases that are highly likely to co-occur with a primary disease according to the scores. In this study, we propose a method of implementing 'n-of-1 utility' (n potential diseases of one patient) to human disease network-the translational disease network. RESULTS: We first construct a disease network by introducing the notion of walk in graph theory to protein-protein interaction network, and then provide a scoring algorithm quantifying the likelihoods of disease co-occurrence given a primary disease. Metabolic diseases, that are highly prevalent but have found only a few associations in previous studies, are chosen as entries of the network. CONCLUSIONS: The proposed method substantially increased connectivity between metabolic diseases and provided scores of co-occurring diseases. The increase in connectivity turned the disease network info-richer. The result lifted the AUC of random guessing up to 0.72 and appeared to be concordant with the existing literatures on disease comorbidity.


Asunto(s)
Enfermedades Metabólicas/metabolismo , Mapas de Interacción de Proteínas , Investigación Biomédica Traslacional , Algoritmos , Área Bajo la Curva , Comorbilidad , Humanos , Probabilidad
11.
BMC Med Inform Decis Mak ; 17(Suppl 1): 52, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28539122

RESUMEN

BACKGROUND: Biological system is a multi-layered structure of omics with genome, epigenome, transcriptome, metabolome, proteome, etc., and can be further stretched to clinical/medical layers such as diseasome, drugs, and symptoms. One advantage of omics is that we can figure out an unknown component or its trait by inferring from known omics components. The component can be inferred by the ones in the same level of omics or the ones in different levels. METHODS: To implement the inference process, an algorithm that can be applied to the multi-layered complex system is required. In this study, we develop a semi-supervised learning algorithm that can be applied to the multi-layered complex system. In order to verify the validity of the inference, it was applied to the prediction problem of disease co-occurrence with a two-layered network composed of symptom-layer and disease-layer. RESULTS: The symptom-disease layered network obtained a fairly high value of AUC, 0.74, which is regarded as noticeable improvement when comparing 0.59 AUC of single-layered disease network. If further stretched to whole layered structure of omics, the proposed method is expected to produce more promising results. CONCLUSION: This research has novelty in that it is a new integrative algorithm that incorporates the vertical structure of omics data, on contrary to other existing methods that integrate the data in parallel fashion. The results can provide enhanced guideline for disease co-occurrence prediction, thereby serve as a valuable tool for inference process of multi-layered biological system.


Asunto(s)
Comorbilidad , Biología Computacional/métodos , Diagnóstico , Aprendizaje Automático Supervisado , Algoritmos , Disciplinas de las Ciencias Biológicas , Humanos
12.
BMC Med Inform Decis Mak ; 17(Suppl 1): 56, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28539112

RESUMEN

BACKGROUND: Hearing Aids amplify sounds at certain frequencies to help patients, who have hearing loss, to improve the quality of life. Variables affecting hearing improvement include the characteristics of the patients' hearing loss, the characteristics of the hearing aids, and the characteristics of the frequencies. Although the two former characteristics have been studied, there are only limited studies predicting hearing gain, after wearing Hearing Aids, with utilizing all three characteristics. Therefore, we propose a new machine learning algorithm that can present the degree of hearing improvement expected from the wearing of hearing aids. METHODS: The proposed algorithm consists of cascade structure, recurrent structure and deep network structure. For cascade structure, it reflects correlations between frequency bands. For recurrent structure, output variables in one particular network of frequency bands are reused as input variables for other networks. Furthermore, it is of deep network structure with many hidden layers. We denote such networks as cascade recurring deep network where training consists of two phases; cascade phase and tuning phase. RESULTS: When applied to medical records of 2,182 patients treated for hearing loss, the proposed algorithm reduced the error rate by 58% from the other neural networks. CONCLUSIONS: The proposed algorithm is a novel algorithm that can be utilized for signal or sequential data. Clinically, the proposed algorithm can serve as a medical assistance tool that fulfill the patients' satisfaction.


Asunto(s)
Audífonos , Pérdida Auditiva/terapia , Aprendizaje Automático , Redes Neurales de la Computación , Algoritmos , Humanos , Estudios Retrospectivos
13.
BMC Med Inform Decis Mak ; 16 Suppl 3: 72, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27454118

RESUMEN

BACKGROUND: The study on disease-disease association has been increasingly viewed and analyzed as a network, in which the connections between diseases are configured using the source information on interactome maps of biomolecules such as genes, proteins, metabolites, etc. Although abundance in source information leads to tighter connections between diseases in the network, for a certain group of diseases, such as metabolic diseases, the connections do not occur much due to insufficient source information; a large proportion of their associated genes are still unknown. One way to circumvent the difficulties in the lack of source information is to integrate available external information by using one of up-to-date integration or fusion methods. However, if one wants a disease network placing huge emphasis on the original source of data but still utilizing external sources only to complement it, integration may not be pertinent. Interpretation on the integrated network would be ambiguous: meanings conferred on edges would be vague due to fused information. METHODS: In this study, we propose a network based algorithm that complements the original network by utilizing external information while preserving the network's originality. The proposed algorithm links the disconnected node to the disease network by using complementary information from external data source through four steps: anchoring, connecting, scoring, and stopping. RESULTS: When applied to the network of metabolic diseases that is sourced from protein-protein interaction data, the proposed algorithm recovered connections by 97%, and improved the AUC performance up to 0.71 (lifted from 0.55) by using the external information outsourced from text mining results on PubMed comorbidity literatures. Experimental results also show that the proposed algorithm is robust to noisy external information. CONCLUSION: This research has novelty in which the proposed algorithm preserves the network's originality, but at the same time, complements it by utilizing external information. Furthermore it can be utilized for original association recovery and novel association discovery for disease network.


Asunto(s)
Minería de Datos , Enfermedades Metabólicas , Redes Neurales de la Computación , Mapas de Interacción de Proteínas , PubMed , Algoritmos , Humanos
14.
Small Methods ; : e2301735, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38529746

RESUMEN

GaAs thin-film solar cells have high efficiency, reliability, and operational stability, making them a promising solution for self-powered skin-conformal biosensors. However, inherent device thickness limits suitability for such applications, making them uncomfortable and unreliable in flexural environments. Therefore, reducing the flexural rigidity becomes crucial for integration with skin-compatible electronic devices. Herein, this study demonstrated a novel one-step surface modification bonding methodology, allowing a streamlined transfer process of ultra-thin (2.3 µm thick) GaAs solar cells on flexible polymer substrates. This reproducible technique enables strong bonding between dissimilar materials (GaAs-polydimethylsiloxane, PDMS) without high external pressures and temperatures. The fabricated solar cell showed exceptional performance with an open-circuit voltage of 1.018 V, short-circuit current density of 20.641 mA cm-2, fill factor of 79.83%, and power conversion efficiency of 16.77%. To prove the concept, the solar cell is integrated with a skin-compatible organic electrochemical transistor (OECT). Competitive electrical outputs of GaAs solar cells enabled high current levels of OECT under subtle light intensities lower than 50 mW cm-2, which demonstrates a self-powered electrocardiogram sensor with low noise (signal-to-noise ratio of 32.68 dB). Overall, this study presents a promising solution for the development of free-form and comfortable device structures that can continuously power wearable devices and biosensors.

15.
Artículo en Inglés | MEDLINE | ID: mdl-38768397

RESUMEN

The integration of multiomics data with detailed phenotypic insights from electronic health records marks a paradigm shift in biomedical research, offering unparalleled holistic views into health and disease pathways. This review delineates the current landscape of multimodal omics data integration, emphasizing its transformative potential in generating a comprehensive understanding of complex biological systems. We explore robust methodologies for data integration, ranging from concatenation-based to transformation-based and network-based strategies, designed to harness the intricate nuances of diverse data types. Our discussion extends from incorporating large-scale population biobanks to dissecting high-dimensional omics layers at the single-cell level. The review underscores the emerging role of large language models in artificial intelligence, anticipating their influence as a near-future pivot in data integration approaches. Highlighting both achievements and hurdles, we advocate for a concerted effort toward sophisticated integration models, fortifying the foundation for groundbreaking discoveries in precision medicine.

16.
medRxiv ; 2023 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-37293013

RESUMEN

Many diseases exhibit complex multimorbidities with one another. An intuitive way to model the connections between phenotypes is with a disease-disease network (DDN), where nodes represent diseases and edges represent associations, such as shared single-nucleotide polymorphisms (SNPs), between pairs of diseases. To gain further genetic understanding of molecular contributors to disease associations, we propose a novel version of the shared-SNP DDN (ssDDN), denoted as ssDDN+, which includes connections between diseases derived from genetic correlations with endophenotypes. We hypothesize that a ssDDN+ can provide complementary information to the disease connections in a ssDDN, yielding insight into the role of clinical laboratory measurements in disease interactions. Using PheWAS summary statistics from the UK Biobank, we constructed a ssDDN+ revealing hundreds of genetic correlations between disease phenotypes and quantitative traits. Our augmented network uncovers genetic associations across different disease categories, connects relevant cardiometabolic diseases, and highlights specific biomarkers that are associated with cross-phenotype associations. Out of the 31 clinical measurements under consideration, HDL-C connects the greatest number of diseases and is strongly associated with both type 2 diabetes and diabetic retinopathy. Triglycerides, another blood lipid with known genetics causes in non-mendelian diseases, also adds a substantial number of edges to the ssDDN. Our study can facilitate future network-based investigations of cross-phenotype associations involving pleiotropy and genetic heterogeneity, potentially uncovering sources of missing heritability in multimorbidities.

17.
Sci Rep ; 13(1): 571, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36631519

RESUMEN

Recently, biocompatible optical sources have been surfacing for new-rising biomedical applications, allowing them to be used for multi-purpose technologies such as biological sensing, optogenetic modulation, and phototherapy. Especially, vertical-cavity surface-emitting laser (VCSEL) is in the spotlight as a prospective candidate for optical sources owing to its low-driving current performance, low-cost, and package easiness in accordance with two-dimensional (2D) arrays structure. In this study, we successfully demonstrated the actualization of biocompatible thin-film 930 nm VCSELs transferred onto a Polydimethylsiloxane (PDMS) carrier. The PDMS feature with biocompatibility as well as biostability makes the thin-film VCSELs well-suited for biomedical applications. In order to integrate the conventional VCSEL onto the PDMS carrier, we utilized a double-transfer technique that transferred the thin-film VCSELs onto foreign substrates twice, enabling it to maintain the p-on-n polarity of the conventional VCSEL. Additionally, we employed a surface modification-assisted bonding (SMB) using an oxygen plasma in conjunction with silane treatment when bonding the PDMS carrier with the substrate-removed conventional VCSELs. The threshold current and maximum output power of the fabricated 930 nm thin-film VCSELs are 1.08 mA and 7.52 mW at an injection current of 13.9 mA, respectively.


Asunto(s)
Dimetilpolisiloxanos , Rayos Láser , Fototerapia
18.
Aging (Albany NY) ; 16(2): 985-1001, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38154113

RESUMEN

The impact of the senescence related microenvironment on cancer prognosis and therapeutic response remains poorly understood. In this study, we investigated the prognostic significance of senescence related tumor microenvironment genes (PSTGs) and their potential implications for immunotherapy response. Using the Cancer Genome Atlas- head and neck squamous cell carcinoma (HNSC) data, we identified two subtypes based on the expression of PSTGs, acquired from tumor-associated senescence genes, tumor microenvironment (TME)-related genes, and immune-related genes, using consensus clustering. Using the LASSO, we constructed a risk model consisting of senescence related TME core genes (STCGs). The two subtypes exhibited significant differences in prognosis, genetic alterations, methylation patterns, and enriched pathways, and immune infiltration. Our risk model stratified patients into high-risk and low-risk groups and validated in independent cohorts. The high-risk group showed poorer prognosis and immune inactivation, suggesting reduced responsiveness to immunotherapy. Additionally, we observed a significant enrichment of STCGs in stromal cells using single-cell RNA transcriptome data. Our findings highlight the importance of the senescence related TME in HNSC prognosis and response to immunotherapy. This study contributes to a deeper understanding of the complex interplay between senescence and the TME, with potential implications for precision medicine and personalized treatment approaches in HNSC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Microambiente Tumoral , Humanos , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Microambiente Tumoral/genética , Análisis por Conglomerados , Neoplasias de Cabeza y Cuello/genética
19.
Gigascience ; 112022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35166337

RESUMEN

BACKGROUND: Disease complications, the onset of secondary phenotypes given a primary condition, can exacerbate the long-term severity of outcomes. However, the exact cause of many of these cross-phenotype associations is still unknown. One potential reason is shared genetic etiology-common genetic drivers may lead to the onset of multiple phenotypes. Disease-disease networks (DDNs), where nodes represent diseases and edges represent associations between diseases, can provide an intuitive way of understanding the relationships between phenotypes. Using summary statistics from a phenome-wide association study (PheWAS), we can generate a corresponding DDN where edges represent shared genetic variants between diseases. Such a network can help us analyze genetic associations across the diseasome, the landscape of all human diseases, and identify potential genetic influences for disease complications. RESULTS: To improve the ease of network-based analysis of shared genetic components across phenotypes, we developed the humaN disEase phenoType MAp GEnerator (NETMAGE), a web-based tool that produces interactive DDN visualizations from PheWAS summary statistics. Users can search the map by various attributes and select nodes to view related phenotypes, associated variants, and various network statistics. As a test case, we used NETMAGE to construct a network from UK BioBank (UKBB) PheWAS summary statistic data. Our map correctly displayed previously identified disease comorbidities from the UKBB and identified concentrations of hub diseases in the endocrine/metabolic and circulatory disease categories. By examining the associations between phenotypes in our map, we can identify potential genetic explanations for the relationships between diseases and better understand the underlying architecture of the human diseasome. Our tool thus provides researchers with a means to identify prospective genetic targets for drug design, using network medicine to contribute to the exploration of personalized medicine.


Asunto(s)
Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Humanos , Fenotipo , Estudios Prospectivos
20.
Pac Symp Biocomput ; 27: 325-336, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34890160

RESUMEN

The polygenic risk score (PRS) can help to identify individuals' genetic susceptibility for various diseases by combining patient genetic profiles and identified single-nucleotide polymorphisms (SNPs) from genome-wide association studies. Although multiple diseases will usually afflict patients at once or in succession, conventional PRSs fail to consider genetic relationships across multiple diseases. Even multi-trait PRSs, which take into account genetic effects for more than one disease at a time, fail to consider a sufficient number of phenotypes to accurately reflect the state of disease comorbidity in a patient, or are biased in terms of the traits that are selected. Thus, we developed novel network-based comorbidity risk scores to quantify associations among multiple phenotypes from phenome-wide association studies (PheWAS). We first constructed a disease-SNP heterogeneous multi-layered network (DS-Net), which consists of a disease network (disease-layer) and SNP network (SNP-layer). The disease-layer describes the population-level interactome from PheWAS data. The SNP-layer was constructed according to linkage disequilibrium. Both layers were attached to transform the information from a population-level interactome to individual-level inferences. Then, graph-based semi-supervised learning was applied to predict possible comorbidity scores on disease-layer for each subject. The SNP-layer serves as receiving individual genotyping data in the scoring process, and the disease-layer serves as the propagated output for an individual's multiple disease comorbidity scores. The possible comorbidity scores were combined by logistic regression, and it is denoted as netCRS. The DS-Net was constructed from UK Biobank PheWAS data, and the individual genetic profiles were collected from the Penn Medicine Biobank. As a proof-of-concept study, myocardial infarction (MI) was selected to compare netCRS with the PRS with pruning and thresholding (PRS-PT). The combined model (netCRS + PRS-PT + covariates) achieved an AUC improvement of 6.26% compared to the (PRS-PT + covariates) model. In terms of risk stratification, the combined model was able to capture the risk of MI up to approximately eight-fold higher than that of the low-risk group. The netCRS and PRS-PT complement each other in predicting high-risk groups of patients with MI. We expect that using these risk prediction models will allow for the development of prevention strategies and reduction of MI morbidity and mortality.


Asunto(s)
Estudio de Asociación del Genoma Completo , Infarto del Miocardio , Bancos de Muestras Biológicas , Biología Computacional , Predisposición Genética a la Enfermedad , Humanos , Herencia Multifactorial , Infarto del Miocardio/epidemiología , Infarto del Miocardio/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA