143 results on '"Guo, Minzhe"'
Search Results
102. Diverse virtual replicas for improving intrusion tolerance in cloud.
- Author
-
Guo, Minzhe and Bhattacharya, Prabir
- Published
- 2014
- Full Text
- View/download PDF
103. Work in progress: Real world relevant security labware for mobile threat analysis and protection experience
- Author
-
Guo, Minzhe, primary, Bhattacharya, Prabir, additional, Yang, Ming, additional, Qian, Kai, additional, and Yang, Li, additional
- Published
- 2012
- Full Text
- View/download PDF
104. Mechanism Design Based Secure Data Object Replication
- Author
-
Guo, Minzhe, primary and Bhattacharya, Prabir, additional
- Published
- 2012
- Full Text
- View/download PDF
105. Mobile security labware with smart devices for cybersecurity education
- Author
-
Qian, Kai, primary, Dan Lo, Chia-Tien, additional, Guo, Minzhe, additional, Bhattacharya, Prabir, additional, and Yang, Li, additional
- Published
- 2012
- Full Text
- View/download PDF
106. A Collaborative Interactive Cyber-learning Platform for Anywhere Anytime Java Programming Learning
- Author
-
Bhattacharya, Prabir, primary, Guo, Minzhe, additional, Tao, Lixin, additional, Fu, Yujian, additional, and Qian, Kai, additional
- Published
- 2011
- Full Text
- View/download PDF
107. Live Programming Learning Objects on Cloud
- Author
-
Wu, Bin, primary, Qian, Kai, additional, Bhattacharya, Prabir, additional, Guo, Minzhe, additional, and Hu, Wanjun, additional
- Published
- 2011
- Full Text
- View/download PDF
108. A Cloud-based Cyberlearning Environment for Introductory Computing Programming Education
- Author
-
Bhattacharya, Prabir, primary, Guo, Minzhe, additional, Tao, Lixin, additional, Wu, Bin, additional, Qian, Kai, additional, and Palmer, E. Kent, additional
- Published
- 2011
- Full Text
- View/download PDF
109. Measuring and ranking attacks based on vulnerability analysis
- Author
-
Wang, Ju An, primary, Guo, Minzhe, additional, Wang, Hao, additional, and Zhou, Linfeng, additional
- Published
- 2011
- Full Text
- View/download PDF
110. Work in progress — A real time Java programming online assessment system
- Author
-
Guo, Minzhe, primary, Qian, Kai, additional, Yang, Li, additional, and Bhattacharya, Prabir, additional
- Published
- 2010
- Full Text
- View/download PDF
111. Vulnerability categorization using Bayesian networks
- Author
-
Wang, Ju An, primary and Guo, Minzhe, additional
- Published
- 2010
- Full Text
- View/download PDF
112. Measuring Similarity for Security Vulnerabilities
- Author
-
Wang, Ju An, primary, Zhou, Linfeng, additional, Guo, Minzhe, additional, Wang, Hao, additional, and Camargo, Jairo, additional
- Published
- 2010
- Full Text
- View/download PDF
113. Ranking Attacks Based on Vulnerability Analysis
- Author
-
Wang, Ju An, primary, Wang, Hao, additional, Guo, Minzhe, additional, Zhou, Linfeng, additional, and Camargo, Jairo, additional
- Published
- 2010
- Full Text
- View/download PDF
114. Design of Online Runtime and Testing Environment for Instant Java Programming Assessment
- Author
-
Guo, Minzhe, primary, Chai, Taolun, additional, and Qian, Kai, additional
- Published
- 2010
- Full Text
- View/download PDF
115. Environmental Metrics for Software Security Based on a Vulnerability Ontology
- Author
-
Wang, Ju An, primary, Guo, Minzhe, additional, Wang, Hao, additional, Xia, Min, additional, and Zhou, Linfeng, additional
- Published
- 2009
- Full Text
- View/download PDF
116. Ontology-based security assessment for software products
- Author
-
Wang, Ju An, primary, Guo, Minzhe, additional, Wang, Hao, additional, Xia, Min, additional, and Zhou, Linfeng, additional
- Published
- 2009
- Full Text
- View/download PDF
117. OVM
- Author
-
Wang, Ju An, primary and Guo, Minzhe, additional
- Published
- 2009
- Full Text
- View/download PDF
118. Security metrics for software systems
- Author
-
Wang, Ju An, primary, Wang, Hao, additional, Guo, Minzhe, additional, and Xia, Min, additional
- Published
- 2009
- Full Text
- View/download PDF
119. Security Data Mining in an Ontology for Vulnerability Management
- Author
-
Wang, Ju An, primary and Guo, Minzhe, additional
- Published
- 2009
- Full Text
- View/download PDF
120. SLICE: determining cell differentiation and lineage based on single cell entropy
- Author
-
Guo, Minzhe, Bao, Erik L., Wagner, Michael, Whitsett, Jeffrey A., and Xu, Yan
- Abstract
A complex organ contains a variety of cell types, each with its own distinct lineage and function. Understanding the lineage and differentiation state of each cell is fundamentally important for the ultimate delineation of organ formation and function. We developed SLICE, a novel algorithm that utilizes single-cell RNA-seq (scRNA-seq) to quantitatively measure cellular differentiation states based on single cell entropy and predict cell differentiation lineages via the construction of entropy directed cell trajectories. We validated our approach using three independent data sets with known lineage and developmental time information from both Homo sapiens and Mus musculus. SLICE successfully measured the differentiation states of single cells and reconstructed cell differentiation trajectories that have been previously experimentally validated. We then applied SLICE to scRNA-seq of embryonic mouse lung at E16.5 to identify lung mesenchymal cell lineage relationships that currently remain poorly defined. A two-branched differentiation pathway of five fibroblastic subtypes was predicted using SLICE. The present study demonstrated the general applicability and high predictive accuracy of SLICE in determining cellular differentiation states and reconstructing cell differentiation lineages in scRNA-seq analysis.
- Published
- 2017
- Full Text
- View/download PDF
121. Vulnerability categorization using Bayesian networks.
- Author
-
Wang, Ju An and Guo, Minzhe
- Published
- 2010
- Full Text
- View/download PDF
122. OVM.
- Author
-
Wang, Ju An and Guo, Minzhe
- Published
- 2009
- Full Text
- View/download PDF
123. Ontology-based security assessment for software products.
- Author
-
Wang, Ju An, Guo, Minzhe, Wang, Hao, Xia, Min, and Zhou, Linfeng
- Published
- 2009
- Full Text
- View/download PDF
124. 'LungGENS': a web-based tool for mapping single-cell gene expression in the developing lung.
- Author
-
Yina Du, Minzhe Guo, Whitsett, Jeffrey A., Yan Xu, Du, Yina, Guo, Minzhe, and Xu, Yan
- Subjects
COMPUTER software ,GENE expression ,GENE mapping ,INTERNET ,LUNGS ,RESEARCH funding ,TRANSCRIPTION factors ,BIOINFORMATICS ,GENE expression profiling - Abstract
We developed LungGENS (Lung Gene Expression iN Single-cell), a web-based bioinformatics resource for querying single-cell gene expression databases by entering a gene symbol or a list of genes or selecting a cell type of their interest. Gene query provides quantitative RNA expression of the gene of interest in each lung cell type. Cell type query returns associated selective gene signatures and genes encoding cell surface markers and transcription factors in interactive heatmap and tables. LungGENS will be broadly applicable in respiratory research, providing a cell-specific RNA expression resource at single-cell resolution. LungGENS is freely available for non-commercial use at https://research.cchmc.org/pbge/lunggens/default.html. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
125. Algorithmic Mechanism Design for Data Replication Problems
- Author
-
Guo, Minzhe
- Subjects
- Computer Science, algorithmic mechanism design, content delivery, data replica placement, malicious, peer-assisted, self-interested
- Abstract
Data replication is an important technique in modern storage-capable distributed systems, such as content delivery networks (CDNs), peer-to-peer networks (P2Ps), and mobile networks, for improving system availability, reliability, and fault-tolerance. Most existing studies on data replication problems assume that all participants in the system fully comply with the designed protocols. Nevertheless, in real-world data replication applications, entities, e.g., servers, data providers, or data consumers, can belong to different stakeholders or administrative domains with different preferences and objectives, exhibiting heterogeneous behaviors that may not be consistent with the expected behavior of the designed protocols. This dissertation studies the problems of data replica placement (DRP), a key component in data replication applications, and utilizes algorithmic mechanism design theory to design algorithms for DRP problems in the settings with heterogeneous behavior models. We first study the DRP problem in CDN in a strategic setting where multiple self-interested players with private preferences own data objects for replication. We design quantitative metrics to measure the content delivery cost associated with specific replica placements and investigate the super-modularity and monotonicity of the cost metrics. We then design DRPMECH, an incentive compatible mechanism that approximates a socially efficient solution to the problem. A detailed set of experiments validates the properties of DRPMECH and shows that it outperforms a state-of-the-art game-theoretical algorithm. Next, we study the DRP problem in peer-assisted content delivery networks (PCDNs) with self-interested seeders. Recently, PCDNs have been proposed for simultaneously obtaining the scalability advantage of P2Ps and the reliability and manageability advantages of CDNs. However, the benefits of peer assistance can be severely affected by the self-interested nature of peers, who in general wish to download more and upload less, unless otherwise motivated. We present DPRP-IC, a decentralized algorithm for DRP in PCDNs, which considers peer contributions and incentives self-interested seeders. We investigate the incentive compatibility of the algorithm and design experiments to evaluate its performance. Results suggest that replica placement algorithms that consider peer contributions have better performance in PCDN; in addition, our approach incentivizes the contribution of self-interested seeders and further improves the performance of replica placement in PCDN.Finally, we study the impact of malicious behaviors on the mechanism design. We identify two types of adversaries in the system, design quantitative metrics to measure the magnitude of malice, and experimentally evaluate the impact of malicious behaviors on the DPRP-IC algorithm. We then integrate the probability of malice of each agent into mechanism design and extend DPRP-IC to a security aware solution, DPRP-IC-SA, which is more resilient to malicious attacks, as demonstrated with a detailed set of experiments.
- Published
- 2016
126. Patho-transcriptomic analysis of invasive mucinous adenocarcinoma of the lung (IMA): comparison with lung adenocarcinoma with signet ring cell features (SRCC).
- Author
-
Stuart WD, Ito M, Baldauf IF, Fukazawa T, Yamatsuji T, Tsuchiya T, Watanabe H, Okada M, Snyder EL, Mino-Kenudson M, Guo M, and Maeda Y
- Abstract
Background: Invasive mucinous adenocarcinoma (IMA) comprises ∼5% of lung adenocarcinoma. There is no effective therapy for IMA when surgical resection is not possible. IMA is sometimes confused with adenocarcinoma with signet ring cell features (SRCC) pathologically since both adenocarcinomas feature tumor cells with abundant intracellular mucin. The molecular mechanisms by which such mucin-producing lung adenocarcinomas develop remain unknown., Methods: Using a Visium spatial transcriptomics approach, we analyzed IMA and compared it with SRCC patho-transcriptomically. Combining spatial transcriptomics data with in vitro studies using RNA-seq and ChIP-seq, we assessed downstream targets of transcription factors HNF4A and SPDEF that are highly expressed in IMA and/or SRCC., Results: Spatial transcriptomics analysis indicated that there are 6 distinct cell clusters in IMA and SRCC. Notably, two clusters (C1 and C3) of mucinous tumor cells exist in both adenocarcinomas albeit at a different ratio. Importantly, a portion of genes (e.g., NKX2-1 , GKN1 , HNF4A and FOXA3 ) are distinctly expressed while some mucous-related genes (e.g., SPDEF and FOXA2 ) are expressed in both adenocarcinomas. We determined that HNF4A induces MUC3A/B and TM4SF4 and that BI 6015, an HNF4A antagonist, suppressed the growth of IMA cells. Using mutant SPDEF that is associated with COVID-19, we also determined that an intact DNA-binding domain of SPDEF is required for SPDEF-mediated induction of mucin genes ( MUC5AC , MUC5B and AGR2 ). Additionally, we found that XMU-MP-1, a SPDEF inhibitor, suppressed the growth of IMA cells., Conclusion: These results revealed that IMA and SRCC contain heterogenous tumor cell types, some of which are targetable.
- Published
- 2024
- Full Text
- View/download PDF
127. Deciphering Endothelial and Mesenchymal Organ Specification in Vascularized Lung and Intestinal Organoids.
- Author
-
Miao Y, Tan C, Pek NM, Yu Z, Iwasawa K, Kechele DO, Sundaram N, Pastrana-Gomez V, Kishimoto K, Yang MC, Jiang C, Tchieu J, Whitsett JA, McCracken KW, Rottier RJ, Kotton DN, Helmrath MA, Wells JM, Takebe T, Zorn AM, Chen YW, Guo M, and Gu M
- Abstract
To investigate the co-development of vasculature, mesenchyme, and epithelium crucial for organogenesis and the acquisition of organ-specific characteristics, we constructed a human pluripotent stem cell-derived organoid system comprising lung or intestinal epithelium surrounded by organotypic mesenchyme and vasculature. We demonstrated the pivotal role of co-differentiating mesoderm and endoderm via precise BMP regulation in generating multilineage organoids and gut tube patterning. Single-cell RNA-seq analysis revealed organ specificity in endothelium and mesenchyme, and uncovered key ligands driving endothelial specification in the lung (e.g., WNT2B and Semaphorins) or intestine (e.g., GDF15). Upon transplantation under the kidney capsule in mice, these organoids further matured and developed perfusable human-specific sub-epithelial capillaries. Additionally, our model recapitulated the abnormal endothelial-epithelial crosstalk in patients with FOXF1 deletion or mutations. Multilineage organoids provide a unique platform to study developmental cues guiding endothelial and mesenchymal cell fate determination, and investigate intricate cell-cell communications in human organogenesis and disease., Highlights: BMP signaling fine-tunes the co-differentiation of mesoderm and endoderm.The cellular composition in multilineage organoids resembles that of human fetal organs.Mesenchyme and endothelium co-developed within the organoids adopt organ-specific characteristics.Multilineage organoids recapitulate abnormal endothelial-epithelial crosstalk in FOXF1-associated disorders.
- Published
- 2024
- Full Text
- View/download PDF
128. LungMAP Portal Ecosystem: Systems-level Exploration of the Lung.
- Author
-
Gaddis N, Fortriede J, Guo M, Bardes EE, Kouril M, Tabar S, Burns K, Ardini-Poleske ME, Loos S, Schnell D, Jin K, Iyer B, Du Y, Huo BX, Bhattacharjee A, Korte J, Munshi R, Smith V, Herbst A, Kitzmiller JA, Clair GC, Carson JP, Adkins J, Morrisey EE, Pryhuber GS, Misra R, Whitsett JA, Sun X, Heathorn T, Paten B, Prasath VBS, Xu Y, Tickle T, Aronow BJ, and Salomonis N
- Subjects
- Animals, Humans, Mice, Lung, Mammals, Organogenesis, Genomics methods
- Abstract
An improved understanding of the human lung necessitates advanced systems models informed by an ever-increasing repertoire of molecular omics, cellular imaging, and pathological datasets. To centralize and standardize information across broad lung research efforts, we expanded the LungMAP.net website into a new gateway portal. This portal connects a broad spectrum of research networks, bulk and single-cell multiomics data, and a diverse collection of image data that span mammalian lung development and disease. The data are standardized across species and technologies using harmonized data and metadata models that leverage recent advances, including those from the Human Cell Atlas, diverse ontologies, and the LungMAP CellCards initiative. To cultivate future discoveries, we have aggregated a diverse collection of single-cell atlases for multiple species (human, rhesus, and mouse) to enable consistent queries across technologies, cohorts, age, disease, and drug treatment. These atlases are provided as independent and integrated queryable datasets, with an emphasis on dynamic visualization, figure generation, reanalysis, cell-type curation, and automated reference-based classification of user-provided single-cell genomics datasets (Azimuth). As this resource grows, we intend to increase the breadth of available interactive interfaces, supported data types, data portals and datasets from LungMAP, and external research efforts.
- Published
- 2024
- Full Text
- View/download PDF
129. PRDM3/16 Regulate Chromatin Accessibility Required for NKX2-1 Mediated Alveolar Epithelial Differentiation and Function.
- Author
-
He H, Bell SM, Davis AK, Zhao S, Sridharan A, Na CL, Guo M, Xu Y, Snowball J, Swarr DT, Zacharias WJ, and Whitsett JA
- Abstract
Differential chromatin accessibility accompanies and mediates transcriptional control of diverse cell fates and their differentiation during embryogenesis. While the critical role of NKX2-1 and its transcriptional targets in lung morphogenesis and pulmonary epithelial cell differentiation is increasingly known, mechanisms by which chromatin accessibility alters the epigenetic landscape and how NKX2-1 interacts with other co-activators required for alveolar epithelial cell differentiation and function are not well understood. Here, we demonstrate that the paired domain zinc finger transcriptional regulators PRDM3 and PRDM16 regulate chromatin accessibility to mediate cell differentiation decisions during lung morphogenesis. Combined deletion of Prdm3 and Prdm16 in early lung endoderm caused perinatal lethality due to respiratory failure from loss of AT2 cell function. Prdm3/16 deletion led to the accumulation of partially differentiated AT1 cells and loss of AT2 cells. Combination of single cell RNA-seq, bulk ATAC-seq, and CUT&RUN demonstrated that PRDM3 and PRDM16 enhanced chromatin accessibility at NKX2-1 transcriptional targets in peripheral epithelial cells, all three factors binding together at a multitude of cell-type specific cis-active DNA elements. Network analysis demonstrated that PRDM3/16 regulated genes critical for perinatal AT2 cell differentiation, surfactant homeostasis, and innate host defense. Lineage specific deletion of PRDM3/16 in AT2 cells led to lineage infidelity, with PRDM3/16 null cells acquiring partial AT1 fate. Together, these data demonstrate that NKX2-1-dependent regulation of alveolar epithelial cell differentiation is mediated by epigenomic modulation via PRDM3/16., Competing Interests: Competing interests: The Authors declare that they have no competing interests for the current work, including patents, financial holdings, advisory positions, or other interests.
- Published
- 2023
- Full Text
- View/download PDF
130. Single Cell Multiomics Identifies Cells and Genetic Networks Underlying Alveolar Capillary Dysplasia.
- Author
-
Guo M, Wikenheiser-Brokamp KA, Kitzmiller JA, Jiang C, Wang G, Wang A, Preissl S, Hou X, Buchanan J, Karolak JA, Miao Y, Frank DB, Zacharias WJ, Sun X, Xu Y, Gu M, Stankiewicz P, Kalinichenko VV, Wambach JA, and Whitsett JA
- Subjects
- Infant, Newborn, Humans, Gene Regulatory Networks genetics, Vascular Endothelial Growth Factor A genetics, Endothelial Cells pathology, Multiomics, Lung pathology, RNA, Forkhead Transcription Factors genetics, Persistent Fetal Circulation Syndrome genetics, Persistent Fetal Circulation Syndrome pathology
- Abstract
Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV ( n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.
- Published
- 2023
- Full Text
- View/download PDF
131. Guided construction of single cell reference for human and mouse lung.
- Author
-
Guo M, Morley MP, Jiang C, Wu Y, Li G, Du Y, Zhao S, Wagner A, Cakar AC, Kouril M, Jin K, Gaddis N, Kitzmiller JA, Stewart K, Basil MC, Lin SM, Ying Y, Babu A, Wikenheiser-Brokamp KA, Mun KS, Naren AP, Clair G, Adkins JN, Pryhuber GS, Misra RS, Aronow BJ, Tickle TL, Salomonis N, Sun X, Morrisey EE, Whitsett JA, and Xu Y
- Subjects
- Animals, Mice, Humans, Single-Cell Analysis, Transcriptome, Gene Expression Profiling, Information Dissemination
- Abstract
Accurate cell type identification is a key and rate-limiting step in single-cell data analysis. Single-cell references with comprehensive cell types, reproducible and functionally validated cell identities, and common nomenclatures are much needed by the research community for automated cell type annotation, data integration, and data sharing. Here, we develop a computational pipeline utilizing the LungMAP CellCards as a dictionary to consolidate single-cell transcriptomic datasets of 104 human lungs and 17 mouse lung samples to construct LungMAP single-cell reference (CellRef) for both normal human and mouse lungs. CellRefs define 48 human and 40 mouse lung cell types catalogued from diverse anatomic locations and developmental time points. We demonstrate the accuracy and stability of LungMAP CellRefs and their utility for automated cell type annotation of both normal and diseased lungs using multiple independent methods and testing data. We develop user-friendly web interfaces for easy access and maximal utilization of the LungMAP CellRefs., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
132. An integrated cell atlas of the lung in health and disease.
- Author
-
Sikkema L, Ramírez-Suástegui C, Strobl DC, Gillett TE, Zappia L, Madissoon E, Markov NS, Zaragosi LE, Ji Y, Ansari M, Arguel MJ, Apperloo L, Banchero M, Bécavin C, Berg M, Chichelnitskiy E, Chung MI, Collin A, Gay ACA, Gote-Schniering J, Hooshiar Kashani B, Inecik K, Jain M, Kapellos TS, Kole TM, Leroy S, Mayr CH, Oliver AJ, von Papen M, Peter L, Taylor CJ, Walzthoeni T, Xu C, Bui LT, De Donno C, Dony L, Faiz A, Guo M, Gutierrez AJ, Heumos L, Huang N, Ibarra IL, Jackson ND, Kadur Lakshminarasimha Murthy P, Lotfollahi M, Tabib T, Talavera-López C, Travaglini KJ, Wilbrey-Clark A, Worlock KB, Yoshida M, van den Berge M, Bossé Y, Desai TJ, Eickelberg O, Kaminski N, Krasnow MA, Lafyatis R, Nikolic MZ, Powell JE, Rajagopal J, Rojas M, Rozenblatt-Rosen O, Seibold MA, Sheppard D, Shepherd DP, Sin DD, Timens W, Tsankov AM, Whitsett J, Xu Y, Banovich NE, Barbry P, Duong TE, Falk CS, Meyer KB, Kropski JA, Pe'er D, Schiller HB, Tata PR, Schultze JL, Teichmann SA, Misharin AV, Nawijn MC, Luecken MD, and Theis FJ
- Subjects
- Humans, Lung, Macrophages, COVID-19, Pulmonary Fibrosis, Lung Neoplasms genetics
- Abstract
Single-cell technologies have transformed our understanding of human tissues. Yet, studies typically capture only a limited number of donors and disagree on cell type definitions. Integrating many single-cell datasets can address these limitations of individual studies and capture the variability present in the population. Here we present the integrated Human Lung Cell Atlas (HLCA), combining 49 datasets of the human respiratory system into a single atlas spanning over 2.4 million cells from 486 individuals. The HLCA presents a consensus cell type re-annotation with matching marker genes, including annotations of rare and previously undescribed cell types. Leveraging the number and diversity of individuals in the HLCA, we identify gene modules that are associated with demographic covariates such as age, sex and body mass index, as well as gene modules changing expression along the proximal-to-distal axis of the bronchial tree. Mapping new data to the HLCA enables rapid data annotation and interpretation. Using the HLCA as a reference for the study of disease, we identify shared cell states across multiple lung diseases, including SPP1
+ profibrotic monocyte-derived macrophages in COVID-19, pulmonary fibrosis and lung carcinoma. Overall, the HLCA serves as an example for the development and use of large-scale, cross-dataset organ atlases within the Human Cell Atlas., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
133. The balance between protective and pathogenic immune responses to pneumonia in the neonatal lung is enforced by gut microbiota.
- Author
-
Stevens J, Steinmeyer S, Bonfield M, Peterson L, Wang T, Gray J, Lewkowich I, Xu Y, Du Y, Guo M, Wynn JL, Zacharias W, Salomonis N, Miller L, Chougnet C, O'Connor DH, and Deshmukh H
- Subjects
- Animals, Anti-Bacterial Agents, Dysbiosis, Female, Humans, Immunity, Lung, Macaca mulatta, Pregnancy, Proteomics, Gastrointestinal Microbiome, Pneumonia
- Abstract
Although modern clinical practices such as cesarean sections and perinatal antibiotics have improved infant survival, treatment with broad-spectrum antibiotics alters intestinal microbiota and causes dysbiosis. Infants exposed to perinatal antibiotics have an increased likelihood of life-threatening infections, including pneumonia. Here, we investigated how the gut microbiota sculpt pulmonary immune responses, promoting recovery and resolution of infection in newborn rhesus macaques. Early-life antibiotic exposure interrupted the maturation of intestinal commensal bacteria and disrupted the developmental trajectory of the pulmonary immune system, as assessed by single-cell proteomic and transcriptomic analyses. Early-life antibiotic exposure rendered newborn macaques more susceptible to bacterial pneumonia, concurrent with increases in neutrophil senescence and hyperinflammation, broad inflammatory cytokine signaling, and macrophage dysfunction. This pathogenic reprogramming of pulmonary immunity was further reflected by a hyperinflammatory signature in all pulmonary immune cell subsets coupled with a global loss of tissue-protective, homeostatic pathways in the lungs of dysbiotic newborns. Fecal microbiota transfer was associated with partial correction of the broad immune maladaptations and protection against severe pneumonia. These data demonstrate the importance of intestinal microbiota in programming pulmonary immunity and support the idea that gut microbiota promote the balance between pathways driving tissue repair and inflammatory responses associated with clinical recovery from infection in infants. Our results highlight a potential role for microbial transfer for immune support in these at-risk infants.
- Published
- 2022
- Full Text
- View/download PDF
134. Inflammatory blockade prevents injury to the developing pulmonary gas exchange surface in preterm primates.
- Author
-
Toth A, Steinmeyer S, Kannan P, Gray J, Jackson CM, Mukherjee S, Demmert M, Sheak JR, Benson D, Kitzmiller J, Wayman JA, Presicce P, Cates C, Rubin R, Chetal K, Du Y, Miao Y, Gu M, Guo M, Kalinichenko VV, Kallapur SG, Miraldi ER, Xu Y, Swarr D, Lewkowich I, Salomonis N, Miller L, Sucre JS, Whitsett JA, Chougnet CA, Jobe AH, Deshmukh H, and Zacharias WJ
- Subjects
- Animals, Female, Lung pathology, Macaca mulatta, Pregnancy, Pulmonary Gas Exchange, Chorioamnionitis chemically induced, Chorioamnionitis pathology, Premature Birth prevention & control
- Abstract
Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque ( Macaca mulatta ) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1β and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.
- Published
- 2022
- Full Text
- View/download PDF
135. CRISPRi-mediated functional analysis of NKX2-1-binding sites in the lung.
- Author
-
Stuart WD, Fink-Baldauf IM, Tomoshige K, Guo M, and Maeda Y
- Subjects
- Binding Sites genetics, CRISPR-Cas Systems genetics, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Chromatin Immunoprecipitation Sequencing methods, Gene Expression Regulation, Neoplastic genetics, Genetic Engineering methods, Humans, Lung Neoplasms genetics, Nuclear Proteins genetics, Promoter Regions, Genetic genetics, Protein Binding genetics, Transcription Factors genetics, Lung pathology, Thyroid Nuclear Factor 1 genetics, Thyroid Nuclear Factor 1 physiology
- Abstract
The transcription factor NKX2-1/TTF-1 is involved in lung pathophysiology, including breathing, innate defense and tumorigenesis. To understand the mechanism by which NKX2-1 regulates genes involved in such pathophysiology, we have previously performed ChIP-seq and identified genome-wide NKX2-1-binding sites, which revealed that NKX2-1 binds to not only proximal promoter regions but also multiple intra- and inter-genic regions of the genes regulated by NKX2-1. However, the roles of such regions, especially non-proximal ones, bound by NKX2-1 have not yet been determined. Here, using CRISPRi (CRISPR/dCas9-KRAB), we scrutinize the functional roles of 19 regions/sites bound by NKX2-1, which are located in genes involved in breathing and innate defense (SFTPB, LAMP3, SFTPA1, SFTPA2) and lung tumorigenesis (MYBPH, LMO3, CD274/PD-L1). Notably, the CRISPRi approach reveals that a portion of NKX2-1-binding sites are functionally indispensable while the rest are dispensable for the expression of the genes, indicating that functional roles of NKX2-1-binding sites are unequally yoked.
- Published
- 2021
- Full Text
- View/download PDF
136. Clinical application of a lung cancer organoid (tumoroid) culture system.
- Author
-
Yokota E, Iwai M, Yukawa T, Yoshida M, Naomoto Y, Haisa M, Monobe Y, Takigawa N, Guo M, Maeda Y, Fukazawa T, and Yamatsuji T
- Abstract
Despite high expectations for lung tumoroids, they have not been applied in the clinic due to the difficulty of their long-term culture. Here, however, using AO (airway organoid) media developed by the Clevers laboratory, we succeeded in generating 3 lung tumoroid lines for long-term culture (>13 months) from 41 lung cancer cases (primary or metastatic). Use of nutlin-3a was key to selecting lung tumoroids that harbor mutant p53 in order to eliminate normal lung epithelial organoids. Next-generation sequencing (NGS) analysis indicated that each lung tumoroid carried BRAF
G469A , TPM3-ROS1 or EGFRL858R /RB1E737* , respectively. Targeted therapies using small molecule drugs (trametinib/erlotinib for BRAFG469A , crizotinib/entrectinib for TPM3-ROS1 and ABT-263/YM-155 for EGFRL858R /RB1E737* ) significantly suppressed the growth of each lung tumoroid line. AO media was superior to 3 different media developed by other laboratories. Our experience indicates that long-term lung tumoroid culture is feasible, allowing us to identify NGS-based therapeutic targets and determine the responsiveness to corresponding small molecule drugs.- Published
- 2021
- Full Text
- View/download PDF
137. Single-cell meta-analysis of SARS-CoV-2 entry genes across tissues and demographics.
- Author
-
Muus C, Luecken MD, Eraslan G, Sikkema L, Waghray A, Heimberg G, Kobayashi Y, Vaishnav ED, Subramanian A, Smillie C, Jagadeesh KA, Duong ET, Fiskin E, Torlai Triglia E, Ansari M, Cai P, Lin B, Buchanan J, Chen S, Shu J, Haber AL, Chung H, Montoro DT, Adams T, Aliee H, Allon SJ, Andrusivova Z, Angelidis I, Ashenberg O, Bassler K, Bécavin C, Benhar I, Bergenstråhle J, Bergenstråhle L, Bolt L, Braun E, Bui LT, Callori S, Chaffin M, Chichelnitskiy E, Chiou J, Conlon TM, Cuoco MS, Cuomo ASE, Deprez M, Duclos G, Fine D, Fischer DS, Ghazanfar S, Gillich A, Giotti B, Gould J, Guo M, Gutierrez AJ, Habermann AC, Harvey T, He P, Hou X, Hu L, Hu Y, Jaiswal A, Ji L, Jiang P, Kapellos TS, Kuo CS, Larsson L, Leney-Greene MA, Lim K, Litviňuková M, Ludwig LS, Lukassen S, Luo W, Maatz H, Madissoon E, Mamanova L, Manakongtreecheep K, Leroy S, Mayr CH, Mbano IM, McAdams AM, Nabhan AN, Nyquist SK, Penland L, Poirion OB, Poli S, Qi C, Queen R, Reichart D, Rosas I, Schupp JC, Shea CV, Shi X, Sinha R, Sit RV, Slowikowski K, Slyper M, Smith NP, Sountoulidis A, Strunz M, Sullivan TB, Sun D, Talavera-López C, Tan P, Tantivit J, Travaglini KJ, Tucker NR, Vernon KA, Wadsworth MH, Waldman J, Wang X, Xu K, Yan W, Zhao W, and Ziegler CGK
- Subjects
- Adult, Aged, Aged, 80 and over, Alveolar Epithelial Cells metabolism, Alveolar Epithelial Cells virology, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 pathology, COVID-19 virology, Cathepsin L genetics, Cathepsin L metabolism, Datasets as Topic statistics & numerical data, Demography, Female, Gene Expression Profiling statistics & numerical data, Humans, Lung metabolism, Lung virology, Male, Middle Aged, Organ Specificity genetics, Respiratory System metabolism, Respiratory System virology, Sequence Analysis, RNA methods, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Single-Cell Analysis methods, COVID-19 epidemiology, COVID-19 genetics, Host-Pathogen Interactions genetics, SARS-CoV-2 physiology, Sequence Analysis, RNA statistics & numerical data, Single-Cell Analysis statistics & numerical data, Virus Internalization
- Abstract
Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2
+ TMPRSS2+ cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.- Published
- 2021
- Full Text
- View/download PDF
138. Single-cell multiomic profiling of human lungs reveals cell-type-specific and age-dynamic control of SARS-CoV2 host genes.
- Author
-
Wang A, Chiou J, Poirion OB, Buchanan J, Valdez MJ, Verheyden JM, Hou X, Kudtarkar P, Narendra S, Newsome JM, Guo M, Faddah DA, Zhang K, Young RE, Barr J, Sajti E, Misra R, Huyck H, Rogers L, Poole C, Whitsett JA, Pryhuber G, Xu Y, Gaulton KJ, Preissl S, and Sun X
- Subjects
- Adult, Age Factors, Alveolar Epithelial Cells classification, Alveolar Epithelial Cells metabolism, Alveolar Epithelial Cells virology, Angiotensin-Converting Enzyme 2 genetics, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 metabolism, Child, Preschool, Chromosome Mapping, Gene Expression Profiling, Genetic Variation, Host Microbial Interactions physiology, Humans, Infant, Newborn, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Pandemics, Receptors, Virus genetics, Receptors, Virus metabolism, SARS-CoV-2 genetics, SARS-CoV-2 pathogenicity, Single-Cell Analysis, Virus Internalization, COVID-19 genetics, COVID-19 virology, Host Microbial Interactions genetics, Lung metabolism, Lung virology
- Abstract
Respiratory failure associated with COVID-19 has placed focus on the lungs. Here, we present single-nucleus accessible chromatin profiles of 90,980 nuclei and matched single-nucleus transcriptomes of 46,500 nuclei in non-diseased lungs from donors of ~30 weeks gestation,~3 years and ~30 years. We mapped candidate cis -regulatory elements (cCREs) and linked them to putative target genes. We identified distal cCREs with age-increased activity linked to SARS-CoV-2 host entry gene TMPRSS2 in alveolar type 2 cells, which had immune regulatory signatures and harbored variants associated with respiratory traits. At the 3p21.31 COVID-19 risk locus, a candidate variant overlapped a distal cCRE linked to SLC6A20 , a gene expressed in alveolar cells and with known functional association with the SARS-CoV-2 receptor ACE2. Our findings provide insight into regulatory logic underlying genes implicated in COVID-19 in individual lung cell types across age. More broadly, these datasets will facilitate interpretation of risk loci for lung diseases., Competing Interests: AW, JC, OP, JB, MV, JV, XH, PK, SN, JN, MG, KZ, RY, JB, ES, RM, HH, LR, CP, JW, GP, YX, SP, XS No competing interests declared, DF employee of and holds stock in Vertex Pharmaceuticals, KG does consulting for Genentech, (© 2020, Wang et al.)
- Published
- 2020
- Full Text
- View/download PDF
139. Glucocorticoid regulates mesenchymal cell differentiation required for perinatal lung morphogenesis and function.
- Author
-
Bridges JP, Sudha P, Lipps D, Wagner A, Guo M, Du Y, Brown K, Filuta A, Kitzmiller J, Stockman C, Chen X, Weirauch MT, Jobe AH, Whitsett JA, and Xu Y
- Subjects
- Alveolar Epithelial Cells metabolism, Animals, Cell Proliferation physiology, Fibroblasts metabolism, Mice, Mice, Inbred C57BL, Receptors, Glucocorticoid metabolism, Signal Transduction physiology, Cell Differentiation physiology, Glucocorticoids metabolism, Lung metabolism, Mesenchymal Stem Cells metabolism
- Abstract
While antenatal glucocorticoids are widely used to enhance lung function in preterm infants, cellular and molecular mechanisms by which glucocorticoid receptor (GR) signaling influences lung maturation remain poorly understood. Deletion of the glucocorticoid receptor gene ( Nr3c1 ) from fetal pulmonary mesenchymal cells phenocopied defects caused by global Nr3c1 deletion, while lung epithelial- or endothelial-specific Nr3c1 deletion did not impair lung function at birth. We integrated genome-wide gene expression profiling, ATAC-seq, and single cell RNA-seq data in mice in which GR was deleted or activated to identify the cellular and molecular mechanisms by which glucocorticoids control prenatal lung maturation. GR enhanced differentiation of a newly defined proliferative mesenchymal progenitor cell (PMP) into matrix fibroblasts (MFBs), in part by directly activating extracellular matrix-associated target genes, including Fn1 , Col16a4 , and Eln and by modulating VEGF, JAK-STAT, and WNT signaling. Loss of mesenchymal GR signaling blocked fibroblast progenitor differentiation into mature MFBs, which in turn increased proliferation of SOX9+ alveolar epithelial progenitor cells and inhibited differentiation of mature alveolar type II (AT
2 ) and AT1 cells. GR signaling controls genes required for differentiation of a subset of proliferative mesenchymal progenitors into matrix fibroblasts, in turn, regulating signals controlling AT2 /AT1 progenitor cell proliferation and differentiation and identifying cells and processes by which glucocorticoid signaling regulates fetal lung maturation.- Published
- 2020
- Full Text
- View/download PDF
140. The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future.
- Author
-
Basil MC, Katzen J, Engler AE, Guo M, Herriges MJ, Kathiriya JJ, Windmueller R, Ysasi AB, Zacharias WJ, Chapman HA, Kotton DN, Rock JR, Snoeck HW, Vunjak-Novakovic G, Whitsett JA, and Morrisey EE
- Subjects
- Cell Communication, Pulmonary Alveoli, Trachea, Lung, Stem Cells
- Abstract
The respiratory system, which includes the trachea, airways, and distal alveoli, is a complex multi-cellular organ that intimately links with the cardiovascular system to accomplish gas exchange. In this review and as members of the NIH/NHLBI-supported Progenitor Cell Translational Consortium, we discuss key aspects of lung repair and regeneration. We focus on the cellular compositions within functional niches, cell-cell signaling in homeostatic health, the responses to injury, and new methods to study lung repair and regeneration. We also provide future directions for an improved understanding of the cell biology of the respiratory system, as well as new therapeutic avenues., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
141. Postnatal Alveologenesis Depends on FOXF1 Signaling in c-KIT + Endothelial Progenitor Cells.
- Author
-
Ren X, Ustiyan V, Guo M, Wang G, Bolte C, Zhang Y, Xu Y, Whitsett JA, Kalin TV, and Kalinichenko VV
- Subjects
- Animals, Humans, Infant, Newborn, Mice, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Tissue Culture Techniques, Endothelial Progenitor Cells physiology, Forkhead Transcription Factors physiology, Lung growth & development, Proto-Oncogene Proteins c-kit metabolism, Signal Transduction physiology
- Abstract
Rationale: Disruption of alveologenesis is associated with severe pediatric lung disorders, including bronchopulmonary dysplasia (BPD). Although c-KIT
+ endothelial cell (EC) progenitors are abundant in embryonic and neonatal lungs, their role in alveolar septation and the therapeutic potential of these cells remain unknown. Objectives: To determine whether c-KIT+ EC progenitors stimulate alveologenesis in the neonatal lung. Methods: We used single-cell RNA sequencing of neonatal human and mouse lung tissues, immunostaining, and FACS analysis to identify transcriptional and signaling networks shared by human and mouse pulmonary c-KIT+ EC progenitors. A mouse model of perinatal hyperoxia-induced lung injury was used to identify molecular mechanisms that are critical for the survival, proliferation, and engraftment of c-KIT+ EC progenitors in the neonatal lung. Measurements and Main Results: Pulmonary c-KIT+ EC progenitors expressing PECAM-1, CD34, VE-Cadherin, FLK1, and TIE2 lacked mature arterial, venal, and lymphatic cell-surface markers. The transcriptomic signature of c-KIT+ ECs was conserved in mouse and human lungs and enriched in FOXF1-regulated transcriptional targets. Expression of FOXF1 and c-KIT was decreased in the lungs of infants with BPD. In the mouse, neonatal hyperoxia decreased the number of c-KIT+ EC progenitors. Haploinsufficiency or endothelial-specific deletion of Foxf1 in mice increased apoptosis and decreased proliferation of c-KIT+ ECs. Inactivation of either Foxf1 or c-Kit caused alveolar simplification. Adoptive transfer of c-KIT+ ECs into the neonatal circulation increased lung angiogenesis and prevented alveolar simplification in neonatal mice exposed to hyperoxia. Conclusions: Cell therapy involving c-KIT+ EC progenitors can be beneficial for the treatment of BPD.- Published
- 2019
- Full Text
- View/download PDF
142. Single-Cell Transcriptome Analysis Using SINCERA Pipeline.
- Author
-
Guo M and Xu Y
- Subjects
- Humans, Computational Biology methods, Gene Expression Profiling, High-Throughput Nucleotide Sequencing methods, Sequence Analysis, RNA methods, Single-Cell Analysis methods, Software
- Abstract
Genome-scale single-cell biology has recently emerged as a powerful technology with important implications for both basic and medical research. There are urgent needs for the development of computational methods or analytic pipelines to facilitate large amounts of single-cell RNA-Seq data analysis. Here, we present a detailed protocol for SINCERA (SINgle CEll RNA-Seq profiling Analysis), a generally applicable analytic pipeline for processing single-cell data from a whole organ or sorted cells. The pipeline supports the analysis for the identification of major cell types, cell type-specific gene signatures, and driving forces of given cell types. In this chapter, we provide step-by-step instructions for the functions and features of SINCERA together with application examples to provide a practical guide for the research community. SINCERA is implemented in R, licensed under the GNU General Public License v3, and freely available from CCHMC PBGE website, https://research.cchmc.org/pbge/sincera.html .
- Published
- 2018
- Full Text
- View/download PDF
143. Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis.
- Author
-
Xu Y, Mizuno T, Sridharan A, Du Y, Guo M, Tang J, Wikenheiser-Brokamp KA, Perl AT, Funari VA, Gokey JJ, Stripp BR, and Whitsett JA
- Subjects
- Gene Expression Profiling, Genetic Markers, Humans, Lung cytology, Single-Cell Analysis, Transcriptome, Epithelial Cells cytology, Idiopathic Pulmonary Fibrosis pathology, Sequence Analysis, RNA
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal interstitial lung disease characterized by airway remodeling, inflammation, alveolar destruction, and fibrosis. We utilized single-cell RNA sequencing (scRNA-seq) to identify epithelial cell types and associated biological processes involved in the pathogenesis of IPF. Transcriptomic analysis of normal human lung epithelial cells defined gene expression patterns associated with highly differentiated alveolar type 2 (AT2) cells, indicated by enrichment of RNAs critical for surfactant homeostasis. In contrast, scRNA-seq of IPF cells identified 3 distinct subsets of epithelial cell types with characteristics of conducting airway basal and goblet cells and an additional atypical transitional cell that contributes to pathological processes in IPF. Individual IPF cells frequently coexpressed alveolar type 1 (AT1), AT2, and conducting airway selective markers, demonstrating "indeterminate" states of differentiation not seen in normal lung development. Pathway analysis predicted aberrant activation of canonical signaling via TGF-β, HIPPO/YAP, P53, WNT, and AKT/PI3K. Immunofluorescence confocal microscopy identified the disruption of alveolar structure and loss of the normal proximal-peripheral differentiation of pulmonary epithelial cells. scRNA-seq analyses identified loss of normal epithelial cell identities and unique contributions of epithelial cells to the pathogenesis of IPF. The present study provides a rich data source to further explore lung health and disease., Competing Interests: The authors have declared that no conflict of interest exists.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.