257 results on '"Chow, Eric D."'
Search Results
2. Therapeutic algebra of immunomodulatory drug responses at single-cell resolution
- Author
-
Jiang, Jialong, Chen, Sisi, Tsou, Tiffany, McGinnis, Christopher S., Khazaei, Tahmineh, Zhu, Qin, Park, Jong H., Rivaud, Paul, Strazhnik, Inna-Marie, Chow, Eric D., Sivak, David A., Gartner, Zev J., and Thomson, Matt
- Subjects
Quantitative Biology - Genomics - Abstract
Therapeutic modulation of immune states is central to the treatment of human disease. However, how drugs and drug combinations impact the diverse cell types in the human immune system remains poorly understood at the transcriptome scale. Here, we apply single-cell mRNA-seq to profile the response of human immune cells to 502 immunomodulatory drugs alone and in combination. We develop a unified mathematical model that quantitatively describes the transcriptome scale response of myeloid and lymphoid cell types to individual drugs and drug combinations through a single inferred regulatory network. The mathematical model reveals how drug combinations generate novel, macrophage and T-cell states by recruiting combinations of gene expression programs through both additive and non-additive drug interactions. A simplified drug response algebra allows us to predict the continuous modulation of immune cell populations between activated, resting and hyper-inhibited states through combinatorial drug dose titrations. Our results suggest that transcriptome-scale mathematical models could enable the design of therapeutic strategies for programming the human immune system using combinations of therapeutics., Comment: 19 pages, 5 figures
- Published
- 2022
3. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution.
- Author
-
Yang, Dian, Jones, Matthew G, Naranjo, Santiago, Rideout, William M, 3rd, Min, Kyung Hoi Joseph, Ho, Raymond, Wu, Wei, Replogle, Joseph M, Page, Jennifer L, Quinn, Jeffrey J, Horns, Felix, Qiu, Xiaojie, Chen, Michael Z, Freed-Pastor, William A, McGinnis, Christopher S, Patterson, David M, Gartner, Zev J, Chow, Eric D, Bivona, Trever G, Chan, Michelle M, Yosef, Nir, Jacks, Tyler, and Weissman, Jonathan S
- Subjects
Animals ,Genes ,ras ,Mice ,Neoplasms: genetics ,Phylogeny ,Whole Exome Sequencing - Abstract
Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth and expansion to neighboring and distal tissues. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma and tracked tumor evolution from single-transformed cells to metastatic tumors at unprecedented resolution. We found that the loss of the initial, stable alveolar-type2-like state was accompanied by a transient increase in plasticity. This was followed by the adoption of distinct transcriptional programs that enable rapid expansion and, ultimately, clonal sweep of stable subclones capable of metastasizing. Finally, tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates progression by creating novel trajectories. Our study elucidates the hierarchical nature of tumor evolution and, more broadly, enables in-depth studies of tumor progression.
- Published
- 2022
4. Estimation of Secondary Household Attack Rates for Emergent Spike L452R Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants Detected by Genomic Surveillance at a Community-Based Testing Site in San Francisco
- Author
-
Peng, James, Liu, Jamin, Mann, Sabrina A, Mitchell, Anthea M, Laurie, Matthew T, Sunshine, Sara, Pilarowski, Genay, Ayscue, Patrick, Kistler, Amy, Vanaerschot, Manu, Li, Lucy M, McGeever, Aaron, Chow, Eric D, Marquez, Carina, Nakamura, Robert, Rubio, Luis, Chamie, Gabriel, Jones, Diane, Jacobo, Jon, Rojas, Susana, Rojas, Susy, Tulier-Laiwa, Valerie, Black, Douglas, Martinez, Jackie, Naso, Jamie, Schwab, Joshua, Petersen, Maya, Havlir, Diane, DeRisi, Joseph, and Team, IDseq
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Infectious Diseases ,Infection ,Good Health and Well Being ,COVID-19 ,Genomics ,Humans ,Incidence ,SARS-CoV-2 ,San Francisco ,variant ,spike mutation ,secondary attack rates ,household transmission ,IDseq Team ,Biological Sciences ,Medical and Health Sciences ,Microbiology ,Clinical sciences - Abstract
BackgroundSequencing of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral genome from patient samples is an important epidemiological tool for monitoring and responding to the pandemic, including the emergence of new mutations in specific communities.MethodsSARS-CoV-2 genomic sequences were generated from positive samples collected, along with epidemiological metadata, at a walk-up, rapid testing site in the Mission District of San Francisco, California during 22 November to 1 December, 2020, and 10-29 January 2021. Secondary household attack rates and mean sample viral load were estimated and compared across observed variants.ResultsA total of 12 124 tests were performed yielding 1099 positives. From these, 928 high-quality genomes were generated. Certain viral lineages bearing spike mutations, defined in part by L452R, S13I, and W152C, comprised 54.4% of the total sequences from January, compared to 15.7% in November. Household contacts exposed to the "California" or "West Coast" variants (B.1.427 and B.1.429) were at higher risk of infection compared to household contacts exposed to lineages lacking these variants (0.36 vs 0.29, risk ratio [RR] = 1.28; 95% confidence interval [CI]: 1.00-1.64). The reproductive number was estimated to be modestly higher than other lineages spreading in California during the second half of 2020. Viral loads were similar among persons infected with West Coast versus non-West Coast strains, as was the proportion of individuals with symptoms (60.9% vs 64.3%).ConclusionsThe increase in prevalence, relative household attack rates, and reproductive number are consistent with a modest transmissibility increase of the West Coast variants. Summary: We observed a growing prevalence and modestly elevated attack rate for "West Coast" severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in a community testing setting in San Francisco during January 2021, suggesting its modestly higher transmissibility.
- Published
- 2022
5. SCITO-seq: single-cell combinatorial indexed cytometry sequencing
- Author
-
Hwang, Byungjin, Lee, David S, Tamaki, Whitney, Sun, Yang, Ogorodnikov, Anton, Hartoularos, George C, Winters, Aidan, Yeung, Bertrand Z, Nazor, Kristopher L, Song, Yun S, Chow, Eric D, Spitzer, Matthew H, and Ye, Chun Jimmie
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Human Genome ,Cancer Genomics ,Biotechnology ,Genetics ,Cancer ,Generic health relevance ,Case-Control Studies ,Flow Cytometry ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,Humans ,Microfluidics ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Transcriptome ,Technology ,Medical and Health Sciences ,Developmental Biology ,Biological sciences - Abstract
The development of DNA-barcoded antibodies to tag cell surface molecules has enabled the use of droplet-based single-cell sequencing (dsc-seq) to profile protein abundances from thousands of cells simultaneously. As compared to flow and mass cytometry, the high per cell cost of current dsc-seq-based workflows precludes their use in clinical applications and large-scale pooled screens. Here, we introduce SCITO-seq, a workflow that uses splint oligonucleotides (oligos) to enable combinatorially indexed dsc-seq of DNA-barcoded antibodies from over 105 cells per reaction using commercial microfluidics. By encoding sample barcodes into splint oligos, we demonstrate that multiplexed SCITO-seq produces reproducible estimates of cellular composition and surface protein expression comparable to those from mass cytometry. We further demonstrate two modified splint oligo designs that extend SCITO-seq to achieve compatibility with commercial DNA-barcoded antibodies and simultaneous expression profiling of the transcriptome and surface proteins from the same cell. These results demonstrate SCITO-seq as a flexible and ultra-high-throughput platform for sequencing-based single-cell protein and multimodal profiling.
- Published
- 2021
6. XYZeq: Spatially resolved single-cell RNA sequencing reveals expression heterogeneity in the tumor microenvironment
- Author
-
Lee, Youjin, Bogdanoff, Derek, Wang, Yutong, Hartoularos, George C, Woo, Jonathan M, Mowery, Cody T, Nisonoff, Hunter M, Lee, David S, Sun, Yang, Lee, James, Mehdizadeh, Sadaf, Cantlon, Joshua, Shifrut, Eric, Ngyuen, David N, Roth, Theodore L, Song, Yun S, Marson, Alexander, Chow, Eric D, and Ye, Chun Jimmie
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Regenerative Medicine ,Human Genome ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Stem Cell Research ,Cancer ,Cancer Genomics ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Animals ,Gene Expression Profiling ,Mice ,Neoplasms ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Transcriptome ,Tumor Microenvironment ,Exome Sequencing - Abstract
Single-cell RNA sequencing (scRNA-seq) of tissues has revealed remarkable heterogeneity of cell types and states but does not provide information on the spatial organization of cells. To better understand how individual cells function within an anatomical space, we developed XYZeq, a workflow that encodes spatial metadata into scRNA-seq libraries. We used XYZeq to profile mouse tumor models to capture spatially barcoded transcriptomes from tens of thousands of cells. Analyses of these data revealed the spatial distribution of distinct cell types and a cell migration-associated transcriptomic program in tumor-associated mesenchymal stem cells (MSCs). Furthermore, we identify localized expression of tumor suppressor genes by MSCs that vary with proximity to the tumor core. We demonstrate that XYZeq can be used to map the transcriptome and spatial localization of individual cells in situ to reveal how cell composition and cell states can be affected by location within complex pathological tissue.
- Published
- 2021
7. Estimation of secondary household attack rates for emergent spike L452R SARS-CoV-2 variants detected by genomic surveillance at a community-based testing site in San Francisco.
- Author
-
Peng, James, Liu, Jamin, Mann, Sabrina A, Mitchell, Anthea M, Laurie, Matthew T, Sunshine, Sara, Pilarowski, Genay, Ayscue, Patrick, Kistler, Amy, Vanaerschot, Manu, Li, Lucy M, McGeever, Aaron, Chow, Eric D, Marquez, Carina, Nakamura, Robert, Rubio, Luis, Chamie, Gabriel, Jones, Diane, Jacobo, Jon, Rojas, Susana, Rojas, Susy, Tulier-Laiwa, Valerie, Black, Douglas, Martinez, Jackie, Naso, Jamie, Schwab, Joshua, Petersen, Maya, Havlir, Diane, DeRisi, Joseph, and IDseq Team
- Subjects
IDseq Team ,SARS-CoV-2 ,household transmission ,secondary attack rates ,spike mutation ,variant ,Vaccine Related ,Prevention ,Infectious Diseases ,Biodefense ,Infection ,Microbiology ,Biological Sciences ,Medical and Health Sciences - Abstract
BackgroundSequencing of the SARS-CoV-2 viral genome from patient samples is an important epidemiological tool for monitoring and responding to the pandemic, including the emergence of new mutations in specific communities.MethodsSARS-CoV-2 genomic sequences were generated from positive samples collected, along with epidemiological metadata, at a walk-up, rapid testing site in the Mission District of San Francisco, California during November 22-December 1, 2020 and January 10-29, 2021. Secondary household attack rates and mean sample viral load were estimated and compared across observed variants.ResultsA total of 12,124 tests were performed yielding 1,099 positives. From these, 928 high quality genomes were generated. Certain viral lineages bearing spike mutations, defined in part by L452R, S13I, and W152C, comprised 54.4% of the total sequences from January, compared to 15.7% in November. Household contacts exposed to the "California" or "West Coast" variants (B.1.427 and B.1.429) were at higher risk of infection compared to household contacts exposed to lineages lacking these variants (0.36 vs 0.29, RR=1.28; 95% CI:1.00-1.64). The reproductive number was estimated to be modestly higher than other lineages spreading in California during the second half of 2020. Viral loads were similar among persons infected with West Coast versus non-West Coast strains, as was the proportion of individuals with symptoms (60.9% vs 64.3%).ConclusionsThe increase in prevalence, relative household attack rates, and reproductive number are consistent with a modest transmissibility increase of the West Coast variants.
- Published
- 2021
8. Rapid pathogen detection by metagenomic next-generation sequencing of infected body fluids
- Author
-
Gu, Wei, Deng, Xianding, Lee, Marco, Sucu, Yasemin D, Arevalo, Shaun, Stryke, Doug, Federman, Scot, Gopez, Allan, Reyes, Kevin, Zorn, Kelsey, Sample, Hannah, Yu, Guixia, Ishpuniani, Gurpreet, Briggs, Benjamin, Chow, Eric D, Berger, Amy, Wilson, Michael R, Wang, Candace, Hsu, Elaine, Miller, Steve, DeRisi, Joseph L, and Chiu, Charles Y
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Biotechnology ,Bioengineering ,Nanotechnology ,Clinical Research ,Infectious Diseases ,Genetics ,4.1 Discovery and preclinical testing of markers and technologies ,Infection ,Good Health and Well Being ,Adult ,Aged ,Bacteria ,Body Fluids ,Cell-Free Nucleic Acids ,Female ,Fungi ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Metagenomics ,Middle Aged ,Medical and Health Sciences ,Immunology ,Biomedical and clinical sciences ,Health sciences - Abstract
We developed a metagenomic next-generation sequencing (mNGS) test using cell-free DNA from body fluids to identify pathogens. The performance of mNGS testing of 182 body fluids from 160 patients with acute illness was evaluated using two sequencing platforms in comparison to microbiological testing using culture, 16S bacterial PCR and/or 28S-internal transcribed ribosomal gene spacer (28S-ITS) fungal PCR. Test sensitivity and specificity of detection were 79 and 91% for bacteria and 91 and 89% for fungi, respectively, by Illumina sequencing; and 75 and 81% for bacteria and 91 and 100% for fungi, respectively, by nanopore sequencing. In a case series of 12 patients with culture/PCR-negative body fluids but for whom an infectious diagnosis was ultimately established, seven (58%) were mNGS positive. Real-time computational analysis enabled pathogen identification by nanopore sequencing in a median 50-min sequencing and 6-h sample-to-answer time. Rapid mNGS testing is a promising tool for diagnosis of unknown infections from body fluids.
- Published
- 2021
9. Macrolide and Nonmacrolide Resistance with Mass Azithromycin Distribution
- Author
-
Doan, Thuy, Worden, Lee, Hinterwirth, Armin, Arzika, Ahmed M, Maliki, Ramatou, Abdou, Amza, Zhong, Lina, Chen, Cindi, Cook, Catherine, Lebas, Elodie, O’Brien, Kieran S, Oldenburg, Catherine E, Chow, Eric D, Porco, Travis C, Lipsitch, Marc, Keenan, Jeremy D, and Lietman, Thomas M
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Clinical Trials and Supportive Activities ,HIV/AIDS ,Clinical Research ,Pediatric ,Antimicrobial Resistance ,Infectious Diseases ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Infection ,Good Health and Well Being ,Anti-Bacterial Agents ,Azithromycin ,Child Mortality ,Child ,Preschool ,Drug Resistance ,Bacterial ,Female ,Gastrointestinal Microbiome ,Humans ,Infant ,Macrolides ,Male ,Mass Drug Administration ,Metagenome ,Niger ,Sequence Analysis ,DNA ,Medical and Health Sciences ,General & Internal Medicine ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundMass distribution of azithromycin to preschool children twice yearly for 2 years has been shown to reduce childhood mortality in sub-Saharan Africa but at the cost of amplifying macrolide resistance. The effects on the gut resistome, a reservoir of antimicrobial resistance genes in the body, of twice-yearly administration of azithromycin for a longer period are unclear.MethodsWe investigated the gut resistome of children after they received twice-yearly distributions of azithromycin for 4 years. In the Niger site of the MORDOR trial, we enrolled 30 villages in a concurrent trial in which they were randomly assigned to receive mass distribution of either azithromycin or placebo, offered to all children 1 to 59 months of age every 6 months for 4 years. Rectal swabs were collected at baseline, 36 months, and 48 months for analysis of the participants' gut resistome. The primary outcome was the ratio of macrolide-resistance determinants in the azithromycin group to those in the placebo group at 48 months.ResultsOver the entire 48-month period, the mean (±SD) coverage was 86.6±12% in the villages that received placebo and 83.2±16.4% in the villages that received azithromycin. A total of 3232 samples were collected during the entire trial period; of the samples obtained at the 48-month monitoring visit, 546 samples from 15 villages that received placebo and 504 from 14 villages that received azithromycin were analyzed. Determinants of macrolide resistance were higher in the azithromycin group than in the placebo group: 7.4 times as high (95% confidence interval [CI], 4.0 to 16.7) at 36 months and 7.5 times as high (95% CI, 3.8 to 23.1) at 48 months. Continued mass azithromycin distributions also selected for determinants of nonmacrolide resistance, including resistance to beta-lactam antibiotics, an antibiotic class prescribed frequently in this region of Africa.ConclusionsAmong villages assigned to receive mass distributions of azithromycin or placebo twice yearly for 4 years, antibiotic resistance was more common in the villages that received azithromycin than in those that received placebo. This trial showed that mass azithromycin distributions may propagate antibiotic resistance. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT02047981.).
- Published
- 2020
10. Rapid deployment of SARS-CoV-2 testing: The CLIAHUB.
- Author
-
Crawford, Emily D, Acosta, Irene, Ahyong, Vida, Anderson, Erika C, Arevalo, Shaun, Asarnow, Daniel, Axelrod, Shannon, Ayscue, Patrick, Azimi, Camillia S, Azumaya, Caleigh M, Bachl, Stefanie, Bachmutsky, Iris, Bhaduri, Aparna, Brown, Jeremy Bancroft, Batson, Joshua, Behnert, Astrid, Boileau, Ryan M, Bollam, Saumya R, Bonny, Alain R, Booth, David, Borja, Michael Jerico B, Brown, David, Buie, Bryan, Burnett, Cassandra E, Byrnes, Lauren E, Cabral, Katelyn A, Cabrera, Joana P, Caldera, Saharai, Canales, Gabriela, Castañeda, Gloria R, Chan, Agnes Protacio, Chang, Christopher R, Charles-Orszag, Arthur, Cheung, Carly, Chio, Unseng, Chow, Eric D, Citron, Y Rose, Cohen, Allison, Cohn, Lillian B, Chiu, Charles, Cole, Mitchel A, Conrad, Daniel N, Constantino, Angela, Cote, Andrew, Crayton-Hall, Tre'Jon, Darmanis, Spyros, Detweiler, Angela M, Dial, Rebekah L, Dong, Shen, Duarte, Elias M, Dynerman, David, Egger, Rebecca, Fanton, Alison, Frumm, Stacey M, Fu, Becky Xu Hua, Garcia, Valentina E, Garcia, Julie, Gladkova, Christina, Goldman, Miriam, Gomez-Sjoberg, Rafael, Gordon, M Grace, Grove, James CR, Gupta, Shweta, Haddjeri-Hopkins, Alexis, Hadley, Pierce, Haliburton, John, Hao, Samantha L, Hartoularos, George, Herrera, Nadia, Hilberg, Melissa, Ho, Kit Ying E, Hoppe, Nicholas, Hosseinzadeh, Shayan, Howard, Conor J, Hussmann, Jeffrey A, Hwang, Elizabeth, Ingebrigtsen, Danielle, Jackson, Julia R, Jowhar, Ziad M, Kain, Danielle, Kim, James YS, Kistler, Amy, Kreutzfeld, Oriana, Kulsuptrakul, Jessie, Kung, Andrew F, Langelier, Charles, Laurie, Matthew T, Lee, Lena, Leng, Kun, Leon, Kristoffer E, Leonetti, Manuel D, Levan, Sophia R, Li, Sam, Li, Aileen W, Liu, Jamin, Lubin, Heidi S, Lyden, Amy, Mann, Jennifer, Mann, Sabrina, and Margulis, Gorica
- Subjects
Humans ,Pneumonia ,Viral ,Coronavirus Infections ,Clinical Laboratory Techniques ,California ,Workflow ,Pandemics ,Clinical Laboratory Services ,Betacoronavirus ,COVID-19 ,SARS-CoV-2 ,COVID-19 Testing ,Pneumonia ,Viral ,Microbiology ,Immunology ,Medical Microbiology ,Virology - Published
- 2020
11. Robust Sequence Determinants of α‑Synuclein Toxicity in Yeast Implicate Membrane Binding
- Author
-
Newberry, Robert W, Arhar, Taylor, Costello, Jean, Hartoularos, George C, Maxwell, Alison M, Naing, Zun Zar Chi, Pittman, Maureen, Reddy, Nishith R, Schwarz, Daniel MC, Wassarman, Douglas R, Wu, Taia S, Barrero, Daniel, Caggiano, Christa, Catching, Adam, Cavazos, Taylor B, Estes, Laurel S, Faust, Bryan, Fink, Elissa A, Goldman, Miriam A, Gomez, Yessica K, Gordon, M Grace, Gunsalus, Laura M, Hoppe, Nick, Jaime-Garza, Maru, Johnson, Matthew C, Jones, Matthew G, Kung, Andrew F, Lopez, Kyle E, Lumpe, Jared, Martyn, Calla, McCarthy, Elizabeth E, Miller-Vedam, Lakshmi E, Navarro, Erik J, Palar, Aji, Pellegrino, Jenna, Saylor, Wren, Stephens, Christina A, Strickland, Jack, Torosyan, Hayarpi, Wankowicz, Stephanie A, Wong, Daniel R, Wong, Garrett, Redding, Sy, Chow, Eric D, DeGrado, William F, and Kampmann, Martin
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Parkinson's Disease ,Brain Disorders ,Neurosciences ,Genetics ,Neurodegenerative ,2.1 Biological and endogenous factors ,Generic health relevance ,Amino Acid Sequence ,Humans ,Mutation ,Parkinson Disease ,Protein Conformation ,Saccharomyces cerevisiae ,alpha-Synuclein ,Chemical Sciences ,Organic Chemistry ,Biological sciences ,Chemical sciences - Abstract
Protein conformations are shaped by cellular environments, but how environmental changes alter the conformational landscapes of specific proteins in vivo remains largely uncharacterized, in part due to the challenge of probing protein structures in living cells. Here, we use deep mutational scanning to investigate how a toxic conformation of α-synuclein, a dynamic protein linked to Parkinson's disease, responds to perturbations of cellular proteostasis. In the context of a course for graduate students in the UCSF Integrative Program in Quantitative Biology, we screened a comprehensive library of α-synuclein missense mutants in yeast cells treated with a variety of small molecules that perturb cellular processes linked to α-synuclein biology and pathobiology. We found that the conformation of α-synuclein previously shown to drive yeast toxicity-an extended, membrane-bound helix-is largely unaffected by these chemical perturbations, underscoring the importance of this conformational state as a driver of cellular toxicity. On the other hand, the chemical perturbations have a significant effect on the ability of mutations to suppress α-synuclein toxicity. Moreover, we find that sequence determinants of α-synuclein toxicity are well described by a simple structural model of the membrane-bound helix. This model predicts that α-synuclein penetrates the membrane to constant depth across its length but that membrane affinity decreases toward the C terminus, which is consistent with orthogonal biophysical measurements. Finally, we discuss how parallelized chemical genetics experiments can provide a robust framework for inquiry-based graduate coursework.
- Published
- 2020
12. ZipSeq: barcoding for real-time mapping of single cell transcriptomes
- Author
-
Hu, Kenneth H, Eichorst, John P, McGinnis, Chris S, Patterson, David M, Chow, Eric D, Kersten, Kelly, Jameson, Stephen C, Gartner, Zev J, Rao, Arjun A, and Krummel, Matthew F
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Human Genome ,Biotechnology ,Genetics ,Animals ,Computational Biology ,DNA Barcoding ,Taxonomic ,Gene Expression Regulation ,Lymph Nodes ,Mice ,NIH 3T3 Cells ,Single-Cell Analysis ,T-Lymphocytes ,Transcriptome ,Tumor Microenvironment ,Technology ,Medical and Health Sciences ,Developmental Biology ,Biological sciences - Abstract
Spatial transcriptomics seeks to integrate single cell transcriptomic data within the three-dimensional space of multicellular biology. Current methods to correlate a cell's position with its transcriptome in living tissues have various limitations. We developed an approach, called 'ZipSeq', that uses patterned illumination and photocaged oligonucleotides to serially print barcodes ('zipcodes') onto live cells in intact tissues, in real time and with an on-the-fly selection of patterns. Using ZipSeq, we mapped gene expression in three settings: in vitro wound healing, live lymph node sections and a live tumor microenvironment. In all cases, we discovered new gene expression patterns associated with histological structures. In the tumor microenvironment, this demonstrated a trajectory of myeloid and T cell differentiation from the periphery inward. A combinatorial variation of ZipSeq efficiently scales in the number of regions defined, providing a pathway for complete mapping of live tissues, subsequent to real-time imaging or perturbation.
- Published
- 2020
13. The DNA methylation landscape of advanced prostate cancer
- Author
-
Zhao, Shuang G, Chen, William S, Li, Haolong, Foye, Adam, Zhang, Meng, Sjöström, Martin, Aggarwal, Rahul, Playdle, Denise, Liao, Arnold, Alumkal, Joshi J, Das, Rajdeep, Chou, Jonathan, Hua, Junjie T, Barnard, Travis J, Bailey, Adina M, Chow, Eric D, Perry, Marc D, Dang, Ha X, Yang, Rendong, Moussavi-Baygi, Ruhollah, Zhang, Li, Alshalalfa, Mohammed, Laura Chang, S, Houlahan, Kathleen E, Shiah, Yu-Jia, Beer, Tomasz M, Thomas, George, Chi, Kim N, Gleave, Martin, Zoubeidi, Amina, Reiter, Robert E, Rettig, Matthew B, Witte, Owen, Yvonne Kim, M, Fong, Lawrence, Spratt, Daniel E, Morgan, Todd M, Bose, Rohit, Huang, Franklin W, Li, Hui, Chesner, Lisa, Shenoy, Tanushree, Goodarzi, Hani, Asangani, Irfan A, Sandhu, Shahneen, Lang, Joshua M, Mahajan, Nupam P, Lara, Primo N, Evans, Christopher P, Febbo, Phillip, Batzoglou, Serafim, Knudsen, Karen E, He, Housheng H, Huang, Jiaoti, Zwart, Wilbert, Costello, Joseph F, Luo, Jianhua, Tomlins, Scott A, Wyatt, Alexander W, Dehm, Scott M, Ashworth, Alan, Gilbert, Luke A, Boutros, Paul C, Farh, Kyle, Chinnaiyan, Arul M, Maher, Christopher A, Small, Eric J, Quigley, David A, and Feng, Felix Y
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer Genomics ,Biotechnology ,Prostate Cancer ,Cancer ,Urologic Diseases ,Human Genome ,2.1 Biological and endogenous factors ,Aged ,Aged ,80 and over ,Carcinogenesis ,DNA Methylation ,Epigenomics ,Gene Expression Regulation ,Neoplastic ,Genome ,Humans ,Male ,Middle Aged ,Mutation ,Prospective Studies ,Prostatic Neoplasms ,Sequence Analysis ,DNA ,Exome Sequencing ,Whole Genome Sequencing ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Although DNA methylation is a key regulator of gene expression, the comprehensive methylation landscape of metastatic cancer has never been defined. Through whole-genome bisulfite sequencing paired with deep whole-genome and transcriptome sequencing of 100 castration-resistant prostate metastases, we discovered alterations affecting driver genes that were detectable only with integrated whole-genome approaches. Notably, we observed that 22% of tumors exhibited a novel epigenomic subtype associated with hypermethylation and somatic mutations in TET2, DNMT3B, IDH1 and BRAF. We also identified intergenic regions where methylation is associated with RNA expression of the oncogenic driver genes AR, MYC and ERG. Finally, we showed that differential methylation during progression preferentially occurs at somatic mutational hotspots and putative regulatory regions. This study is a large integrated study of whole-genome, whole-methylome and whole-transcriptome sequencing in metastatic cancer that provides a comprehensive overview of the important regulatory role of methylation in metastatic castration-resistant prostate cancer.
- Published
- 2020
14. Deep mutational scanning reveals the structural basis for α-synuclein activity.
- Author
-
Newberry, Robert W, Leong, Jaime T, Chow, Eric D, Kampmann, Martin, and DeGrado, William F
- Subjects
Yeasts ,Amyloid ,Amino Acid Sequence ,Protein Conformation ,Protein Binding ,Structure-Activity Relationship ,Phenotype ,Mutation ,Genomic Library ,Models ,Molecular ,Mutant Proteins ,alpha-Synuclein ,Medicinal and Biomolecular Chemistry ,Biochemistry and Cell Biology ,Biochemistry & Molecular Biology - Abstract
Defining the biologically active structures of proteins in their cellular environments remains challenging for proteins with multiple conformations and functions, where only a minor conformer might be associated with a given function. Here, we use deep mutational scanning to probe the structure and dynamics of α-synuclein, a protein known to adopt disordered, helical and amyloid conformations. We examined the effects of 2,600 single-residue substitutions on the ability of intracellularly expressed α-synuclein to slow the growth of yeast. Computational analysis of the data showed that the conformation responsible for this phenotype is a long, uninterrupted, amphiphilic helix with increasing dynamics toward the C terminus. Deep mutational scanning can therefore determine biologically active conformations in cellular environments, even for a highly dynamic multi-conformational protein.
- Published
- 2020
15. FLASH: a next-generation CRISPR diagnostic for multiplexed detection of antimicrobial resistance sequences
- Author
-
Quan, Jenai, Langelier, Charles, Kuchta, Alison, Batson, Joshua, Teyssier, Noam, Lyden, Amy, Caldera, Saharai, McGeever, Aaron, Dimitrov, Boris, King, Ryan, Wilheim, Jordan, Murphy, Maxwell, Ares, Lara Pesce, Travisano, Katherine A, Sit, Rene, Amato, Roberto, Mumbengegwi, Davis R, Smith, Jennifer L, Bennett, Adam, Gosling, Roly, Mourani, Peter M, Calfee, Carolyn S, Neff, Norma F, Chow, Eric D, Kim, Peter S, Greenhouse, Bryan, DeRisi, Joseph L, and Crawford, Emily D
- Subjects
Microbiology ,Biological Sciences ,Bioinformatics and Computational Biology ,Biotechnology ,Emerging Infectious Diseases ,Genetics ,Antimicrobial Resistance ,Infectious Diseases ,4.1 Discovery and preclinical testing of markers and technologies ,4.2 Evaluation of markers and technologies ,Infection ,Good Health and Well Being ,Anti-Bacterial Agents ,Bacteria ,Bacterial Infections ,CRISPR-Cas Systems ,Computational Biology ,Drug Resistance ,Bacterial ,High-Throughput Nucleotide Sequencing ,Humans ,Metagenomics ,Reproducibility of Results ,Sensitivity and Specificity ,Environmental Sciences ,Information and Computing Sciences ,Developmental Biology ,Biological sciences ,Chemical sciences ,Environmental sciences - Abstract
The growing prevalence of deadly microbes with resistance to previously life-saving drug therapies is a dire threat to human health. Detection of low abundance pathogen sequences remains a challenge for metagenomic Next Generation Sequencing (NGS). We introduce FLASH (Finding Low Abundance Sequences by Hybridization), a next-generation CRISPR/Cas9 diagnostic method that takes advantage of the efficiency, specificity and flexibility of Cas9 to enrich for a programmed set of sequences. FLASH-NGS achieves up to 5 orders of magnitude of enrichment and sub-attomolar gene detection with minimal background. We provide an open-source software tool (FLASHit) for guide RNA design. Here we applied it to detection of antimicrobial resistance genes in respiratory fluid and dried blood spots, but FLASH-NGS is applicable to all areas that rely on multiplex PCR.
- Published
- 2019
16. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices
- Author
-
McGinnis, Christopher S, Patterson, David M, Winkler, Juliane, Conrad, Daniel N, Hein, Marco Y, Srivastava, Vasudha, Hu, Jennifer L, Murrow, Lyndsay M, Weissman, Jonathan S, Werb, Zena, Chow, Eric D, and Gartner, Zev J
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer Genomics ,Biotechnology ,Women's Health ,Cancer ,Breast Cancer ,Human Genome ,Animals ,Base Sequence ,HEK293 Cells ,High-Throughput Nucleotide Sequencing ,Humans ,Lipids ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Technology ,Medical and Health Sciences ,Developmental Biology ,Biological sciences - Abstract
Sample multiplexing facilitates scRNA-seq by reducing costs and identifying artifacts such as cell doublets. However, universal and scalable sample barcoding strategies have not been described. We therefore developed MULTI-seq: multiplexing using lipid-tagged indices for single-cell and single-nucleus RNA sequencing. MULTI-seq reagents can barcode any cell type or nucleus from any species with an accessible plasma membrane. The method involves minimal sample processing, thereby preserving cell viability and endogenous gene expression patterns. When cells are classified into sample groups using MULTI-seq barcode abundances, data quality is improved through doublet identification and recovery of cells with low RNA content that would otherwise be discarded by standard quality-control workflows. We use MULTI-seq to track the dynamics of T-cell activation, perform a 96-plex perturbation experiment with primary human mammary epithelial cells and multiplex cryopreserved tumors and metastatic sites isolated from a patient-derived xenograft mouse model of triple-negative breast cancer.
- Published
- 2019
17. Pulmonary Metagenomic Sequencing Suggests Missed Infections in Immunocompromised Children.
- Author
-
Zinter, Matt S, Dvorak, Christopher C, Mayday, Madeline Y, Iwanaga, Kensho, Ly, Ngoc P, McGarry, Meghan E, Church, Gwynne D, Faricy, Lauren E, Rowan, Courtney M, Hume, Janet R, Steiner, Marie E, Crawford, Emily D, Langelier, Charles, Kalantar, Katrina, Chow, Eric D, Miller, Steve, Shimano, Kristen, Melton, Alexis, Yanik, Gregory A, Sapru, Anil, and DeRisi, Joseph L
- Subjects
Lung ,Humans ,Bacteria ,Viruses ,Fungi ,Lung Diseases ,Retrospective Studies ,Pilot Projects ,Immunocompromised Host ,Adolescent ,Child ,Child ,Preschool ,Female ,Male ,Metagenome ,Metagenomics ,High-Throughput Nucleotide Sequencing ,Microbiota ,Missed Diagnosis ,immunocompromised host ,intensive care units ,metagenomics ,microbiota ,pediatric ,respiratory tract infections ,Preschool ,Biological Sciences ,Medical and Health Sciences ,Microbiology - Abstract
BackgroundDespite improved diagnostics, pulmonary pathogens in immunocompromised children frequently evade detection, leading to significant mortality. Therefore, we aimed to develop a highly sensitive metagenomic next-generation sequencing (mNGS) assay capable of evaluating the pulmonary microbiome and identifying diverse pathogens in the lungs of immunocompromised children.MethodsWe collected 41 lower respiratory specimens from 34 immunocompromised children undergoing evaluation for pulmonary disease at 3 children's hospitals from 2014-2016. Samples underwent mechanical homogenization, parallel RNA/DNA extraction, and metagenomic sequencing. Sequencing reads were aligned to the National Center for Biotechnology Information nucleotide reference database to determine taxonomic identities. Statistical outliers were determined based on abundance within each sample and relative to other samples in the cohort.ResultsWe identified a rich cross-domain pulmonary microbiome that contained bacteria, fungi, RNA viruses, and DNA viruses in each patient. Potentially pathogenic bacteria were ubiquitous among samples but could be distinguished as possible causes of disease by parsing for outlier organisms. Samples with bacterial outliers had significantly depressed alpha-diversity (median, 0.61; interquartile range [IQR], 0.33-0.72 vs median, 0.96; IQR, 0.94-0.96; P < .001). Potential pathogens were detected in half of samples previously negative by clinical diagnostics, demonstrating increased sensitivity for missed pulmonary pathogens (P < .001).ConclusionsAn optimized mNGS assay for pulmonary microbes demonstrates significant inoculation of the lower airways of immunocompromised children with diverse bacteria, fungi, and viruses. Potential pathogens can be identified based on absolute and relative abundance. Ongoing investigation is needed to determine the pathogenic significance of outlier microbes in the lungs of immunocompromised children with pulmonary disease.
- Published
- 2019
18. Miniaturization and optimization of 384-well compatible RNA sequencing library preparation.
- Author
-
Mayday, Madeline Y, Khan, Lillian M, Chow, Eric D, Zinter, Matt S, and DeRisi, Joseph L
- Subjects
Microchemistry ,Feasibility Studies ,Sequence Analysis ,RNA ,Cost Savings ,Metagenomics ,Automation ,Laboratory ,High-Throughput Nucleotide Sequencing ,Automation ,Laboratory ,Sequence Analysis ,RNA ,General Science & Technology - Abstract
Preparation of high-quality sequencing libraries is a costly and time-consuming component of metagenomic next generation sequencing (mNGS). While the overall cost of sequencing has dropped significantly over recent years, the reagents needed to prepare sequencing samples are likely to become the dominant expense in the process. Furthermore, libraries prepared by hand are subject to human variability and needless waste due to limitations of manual pipetting volumes. Reduction of reaction volumes, combined with sub-microliter automated dispensing of reagents without consumable pipette tips, has the potential to provide significant advantages. Here, we describe the integration of several instruments, including the Labcyte Echo 525 acoustic liquid handler and the iSeq and NovaSeq Illumina sequencing platforms, to miniaturize and automate mNGS library preparation, significantly reducing the cost and the time required to prepare samples. Through the use of External RNA Controls Consortium (ERCC) spike-in RNAs, we demonstrated the fidelity of the miniaturized preparation to be equivalent to full volume reactions. Furthermore, detection of viral and microbial species from cell culture and patient samples was also maintained in the miniaturized libraries. For 384-well mNGS library preparations, we achieved cost savings of over 80% in materials and reagents alone, and reduced preparation time by 90% compared to manual approaches, without compromising quality or representation within the library.
- Published
- 2019
19. Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults
- Author
-
Langelier, Charles, Kalantar, Katrina L, Moazed, Farzad, Wilson, Michael R, Crawford, Emily D, Deiss, Thomas, Belzer, Annika, Bolourchi, Samaneh, Caldera, Saharai, Fung, Monica, Jauregui, Alejandra, Malcolm, Katherine, Lyden, Amy, Khan, Lillian, Vessel, Kathryn, Quan, Jenai, Zinter, Matt, Chiu, Charles Y, Chow, Eric D, Wilson, Jenny, Miller, Steve, Matthay, Michael A, Pollard, Katherine S, Christenson, Stephanie, Calfee, Carolyn S, and DeRisi, Joseph L
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Microbiology ,Clinical Sciences ,Medical Microbiology ,Infectious Diseases ,Emerging Infectious Diseases ,Rare Diseases ,Genetics ,Microbiome ,Acute Respiratory Distress Syndrome ,Human Genome ,Biotechnology ,Lung ,2.1 Biological and endogenous factors ,4.1 Discovery and preclinical testing of markers and technologies ,Infection ,Good Health and Well Being ,Adult ,Aged ,Aged ,80 and over ,Area Under Curve ,Case-Control Studies ,Cohort Studies ,Critical Illness ,Female ,Humans ,Male ,Microbiota ,Middle Aged ,Predictive Value of Tests ,Respiratory Tract Infections ,Sequence Analysis ,DNA ,Transcriptome ,Whole Genome Sequencing ,lower respiratory tract infection ,pneumonia ,next-generation sequencing ,transcriptome ,mechanical ventilation - Abstract
Lower respiratory tract infections (LRTIs) lead to more deaths each year than any other infectious disease category. Despite this, etiologic LRTI pathogens are infrequently identified due to limitations of existing microbiologic tests. In critically ill patients, noninfectious inflammatory syndromes resembling LRTIs further complicate diagnosis. To address the need for improved LRTI diagnostics, we performed metagenomic next-generation sequencing (mNGS) on tracheal aspirates from 92 adults with acute respiratory failure and simultaneously assessed pathogens, the airway microbiome, and the host transcriptome. To differentiate pathogens from respiratory commensals, we developed a rules-based model (RBM) and logistic regression model (LRM) in a derivation cohort of 20 patients with LRTIs or noninfectious acute respiratory illnesses. When tested in an independent validation cohort of 24 patients, both models achieved accuracies of 95.5%. We next developed pathogen, microbiome diversity, and host gene expression metrics to identify LRTI-positive patients and differentiate them from critically ill controls with noninfectious acute respiratory illnesses. When tested in the validation cohort, the pathogen metric performed with an area under the receiver-operating curve (AUC) of 0.96 (95% CI, 0.86-1.00), the diversity metric with an AUC of 0.80 (95% CI, 0.63-0.98), and the host transcriptional classifier with an AUC of 0.88 (95% CI, 0.75-1.00). Combining these achieved a negative predictive value of 100%. This study suggests that a single streamlined protocol offering an integrated genomic portrait of pathogen, microbiome, and host transcriptome may hold promise as a tool for LRTI diagnosis.
- Published
- 2018
20. Mapping the Genetic Landscape of Human Cells
- Author
-
Horlbeck, Max A, Xu, Albert, Wang, Min, Bennett, Neal K, Park, Chong Y, Bogdanoff, Derek, Adamson, Britt, Chow, Eric D, Kampmann, Martin, Peterson, Tim R, Nakamura, Ken, Fischbach, Michael A, Weissman, Jonathan S, and Gilbert, Luke A
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Cancer ,2.1 Biological and endogenous factors ,Generic health relevance ,Biomarkers ,Cholesterol ,Clustered Regularly Interspaced Short Palindromic Repeats ,Epistasis ,Genetic ,Gene Regulatory Networks ,High-Throughput Nucleotide Sequencing ,Humans ,Jurkat Cells ,K562 Cells ,Protein Interaction Mapping ,CRISPR ,CRISPRi ,epistasis ,functional genomics ,genetic interactions ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Seminal yeast studies have established the value of comprehensively mapping genetic interactions (GIs) for inferring gene function. Efforts in human cells using focused gene sets underscore the utility of this approach, but the feasibility of generating large-scale, diverse human GI maps remains unresolved. We developed a CRISPR interference platform for large-scale quantitative mapping of human GIs. We systematically perturbed 222,784 gene pairs in two cancer cell lines. The resultant maps cluster functionally related genes, assigning function to poorly characterized genes, including TMEM261, a new electron transport chain component. Individual GIs pinpoint unexpected relationships between pathways, exemplified by a specific cholesterol biosynthesis intermediate whose accumulation induces deoxynucleotide depletion, causing replicative DNA damage and a synthetic-lethal interaction with the ATR/9-1-1 DNA repair pathway. Our map provides a broad resource, establishes GI maps as a high-resolution tool for dissecting gene function, and serves as a blueprint for mapping the genetic landscape of human cells.
- Published
- 2018
21. Mass Azithromycin Distribution and Community Microbiome: A Cluster-Randomized Trial.
- Author
-
Doan, Thuy, Hinterwirth, Armin, Arzika, Ahmed M, Cotter, Sun Y, Ray, Kathryn J, O'Brien, Kieran S, Zhong, Lina, Chow, Eric D, Zhou, Zhaoxia, Cummings, Susie L, Fry, Dionna, Oldenburg, Catherine E, Worden, Lee, Porco, Travis C, Keenan, Jeremy D, and Lietman, Thomas M
- Subjects
antibiotics ,azithromycin ,children ,gut microbiome ,randomized controlled trial ,Pediatric ,Clinical Research ,Clinical Trials and Supportive Activities ,Nutrition ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Good Health and Well Being - Abstract
BackgroundMass distributions of oral azithromycin have long been used to eliminate trachoma, and they are now being proposed to reduce childhood mortality. The observed benefit appears to be augmented with each additional treatment, suggesting a possible community-level effect. Here, we assess whether 2 biannual mass treatments of preschool children affect the community's gut microbiome at 6 months after the last distribution.MethodsIn this cluster-randomized controlled trial, children aged 1-60 months in the Dossa region of Niger were randomized at the village level to receive a single dose of azithromycin or placebo every 6 months. Fecal samples were collected 6 months after the second treatment for metagenomic deep sequencing. The prespecified primary outcome was the Euclidean PERMANOVA of the gut microbiome, or effectively the distance between the genus-level centroid at the community level, with the secondary outcome being the Simpson's α diversity.ResultsIn the azithromycin arm, the gut microbial structures were significantly different than in the placebo arm (Euclidean PERMANOVA, P < .001). Further, the diversity of the gut microbiome in the azithromycin arm was significantly lower than in the placebo arm (inverse Simpson's index, P = .005).ConclusionsTwo mass azithromycin administrations, 6 months apart, in preschool children led to long-term alterations of the gut microbiome structure and community diversity. Here, long-term microbial alterations in the community did not imply disease but were associated with an improvement in childhood mortality.Clinical trials registrationNCT02048007.
- Published
- 2018
22. Chronic Meningitis Investigated via Metagenomic Next-Generation Sequencing
- Author
-
Wilson, Michael R, O’Donovan, Brian D, Gelfand, Jeffrey M, Sample, Hannah A, Chow, Felicia C, Betjemann, John P, Shah, Maulik P, Richie, Megan B, Gorman, Mark P, Hajj-Ali, Rula A, Calabrese, Leonard H, Zorn, Kelsey C, Chow, Eric D, Greenlee, John E, Blum, Jonathan H, Green, Gary, Khan, Lillian M, Banerji, Debarko, Langelier, Charles, Bryson-Cahn, Chloe, Harrington, Whitney, Lingappa, Jairam R, Shanbhag, Niraj M, Green, Ari J, Brew, Bruce J, Soldatos, Ariane, Strnad, Luke, Doernberg, Sarah B, Jay, Cheryl A, Douglas, Vanja, Josephson, S Andrew, and DeRisi, Joseph L
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Clinical Sciences ,Infectious Diseases ,Clinical Research ,2.2 Factors relating to the physical environment ,Infection ,Good Health and Well Being ,Adolescent ,Adult ,Animals ,Aspergillus oryzae ,Candida ,Candidiasis ,Child ,Chronic Disease ,Cryptococcus neoformans ,Female ,HIV Infections ,HIV-1 ,High-Throughput Nucleotide Sequencing ,Histoplasma ,Histoplasmosis ,Humans ,Male ,Meningitis ,Meningitis ,Cryptococcal ,Metagenome ,Metagenomics ,Middle Aged ,Neuroaspergillosis ,Neurocysticercosis ,Sequence Analysis ,RNA ,Taenia solium ,Young Adult - Abstract
ImportanceIdentifying infectious causes of subacute or chronic meningitis can be challenging. Enhanced, unbiased diagnostic approaches are needed.ObjectiveTo present a case series of patients with diagnostically challenging subacute or chronic meningitis using metagenomic next-generation sequencing (mNGS) of cerebrospinal fluid (CSF) supported by a statistical framework generated from mNGS of control samples from the environment and from patients who were noninfectious.Design, setting, and participantsIn this case series, mNGS data obtained from the CSF of 94 patients with noninfectious neuroinflammatory disorders and from 24 water and reagent control samples were used to develop and implement a weighted scoring metric based on z scores at the species and genus levels for both nucleotide and protein alignments to prioritize and rank the mNGS results. Total RNA was extracted for mNGS from the CSF of 7 participants with subacute or chronic meningitis who were recruited between September 2013 and March 2017 as part of a multicenter study of mNGS pathogen discovery among patients with suspected neuroinflammatory conditions. The neurologic infections identified by mNGS in these 7 participants represented a diverse array of pathogens. The patients were referred from the University of California, San Francisco Medical Center (n = 2), Zuckerberg San Francisco General Hospital and Trauma Center (n = 2), Cleveland Clinic (n = 1), University of Washington (n = 1), and Kaiser Permanente (n = 1). A weighted z score was used to filter out environmental contaminants and facilitate efficient data triage and analysis.Main outcomes and measuresPathogens identified by mNGS and the ability of a statistical model to prioritize, rank, and simplify mNGS results.ResultsThe 7 participants ranged in age from 10 to 55 years, and 3 (43%) were female. A parasitic worm (Taenia solium, in 2 participants), a virus (HIV-1), and 4 fungi (Cryptococcus neoformans, Aspergillus oryzae, Histoplasma capsulatum, and Candida dubliniensis) were identified among the 7 participants by using mNGS. Evaluating mNGS data with a weighted z score-based scoring algorithm reduced the reported microbial taxa by a mean of 87% (range, 41%-99%) when taxa with a combined score of 0 or less were removed, effectively separating bona fide pathogen sequences from spurious environmental sequences so that, in each case, the causative pathogen was found within the top 2 scoring microbes identified using the algorithm.Conclusions and relevanceDiverse microbial pathogens were identified by mNGS in the CSF of patients with diagnostically challenging subacute or chronic meningitis, including a case of subarachnoid neurocysticercosis that defied diagnosis for 1 year, the first reported case of CNS vasculitis caused by Aspergillus oryzae, and the fourth reported case of C dubliniensis meningitis. Prioritizing metagenomic data with a scoring algorithm greatly clarified data interpretation and highlighted the problem of attributing biological significance to organisms present in control samples used for metagenomic sequencing studies.
- Published
- 2018
23. Extending chemical perturbations of the ubiquitin fitness landscape in a classroom setting reveals new constraints on sequence tolerance.
- Author
-
Mavor, David, Barlow, Kyle A, Asarnow, Daniel, Birman, Yuliya, Britain, Derek, Chen, Weilin, Green, Evan M, Kenner, Lillian R, Mensa, Bruk, Morinishi, Leanna S, Nelson, Charlotte A, Poss, Erin M, Suresh, Pooja, Tian, Ruilin, Arhar, Taylor, Ary, Beatrice E, Bauer, David P, Bergman, Ian D, Brunetti, Rachel M, Chio, Cynthia M, Dai, Shizhong A, Dickinson, Miles S, Elledge, Susanna K, Helsell, Cole V M, Hendel, Nathan L, Kang, Emily, Kern, Nadja, Khoroshkin, Matvei S, Kirkemo, Lisa L, Lewis, Greyson R, Lou, Kevin, Marin, Wesley M, Maxwell, Alison M, McTigue, Peter F, Myers-Turnbull, Douglas, Nagy, Tamas L, Natale, Andrew M, Oltion, Keely, Pourmal, Sergei, Reder, Gabriel K, Rettko, Nicholas J, Rohweder, Peter J, Schwarz, Daniel M C, Tan, Sophia K, Thomas, Paul V, Tibble, Ryan W, Town, Jason P, Tsai, Mary K, Ugur, Fatima S, Wassarman, Douglas R, Wolff, Alexander M, Wu, Taia S, Bogdanoff, Derek, Li, Jennifer, Thorn, Kurt S, O'Conchúir, Shane, Swaney, Danielle L, Chow, Eric D, Madhani, Hiten D, Redding, Sy, Bolon, Daniel N, Kortemme, Tanja, DeRisi, Joseph L, Kampmann, Martin, and Fraser, James S
- Abstract
Although the primary protein sequence of ubiquitin (Ub) is extremely stable over evolutionary time, it is highly tolerant to mutation during selection experiments performed in the laboratory. We have proposed that this discrepancy results from the difference between fitness under laboratory culture conditions and the selective pressures in changing environments over evolutionary timescales. Building on our previous work (Mavor et al., 2016), we used deep mutational scanning to determine how twelve new chemicals (3-Amino-1,2,4-triazole, 5-fluorocytosine, Amphotericin B, CaCl2, Cerulenin, Cobalt Acetate, Menadione, Nickel Chloride, p-Fluorophenylalanine, Rapamycin, Tamoxifen, and Tunicamycin) reveal novel mutational sensitivities of ubiquitin residues. Collectively, our experiments have identified eight new sensitizing conditions for Lys63 and uncovered a sensitizing condition for every position in Ub except Ser57 and Gln62. By determining the ubiquitin fitness landscape under different chemical constraints, our work helps to resolve the inconsistencies between deep mutational scanning experiments and sequence conservation over evolutionary timescales.
- Published
- 2018
24. Supplementary Table 2 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
25. Supplementary Table 3 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
26. Supplementary Figure 4 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
27. Supplementary Figure 5 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
28. Supplementary Figure 1 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
29. Supplementary Figure 3 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
30. Supplementary Table 1 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
31. Data from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
32. Supplementary Figure 2 from Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer
- Author
-
Chang, Hewitt, primary, Marquez, Jaqueline, primary, Chen, Brandon K., primary, Kim, Daniel M., primary, Cheng, Michael L., primary, Liu, Eric V., primary, Yang, Hai, primary, Zhang, Li, primary, Sinha, Meenal, primary, Cheung, Alexander, primary, Kwek, Serena S., primary, Chow, Eric D., primary, Bridge, Mark, primary, Aggarwal, Rahul R., primary, Friedlander, Terence W., primary, Small, Eric J., primary, Anderson, Mark, primary, and Fong, Lawrence, primary
- Published
- 2024
- Full Text
- View/download PDF
33. Erratum to: Illuminating uveitis: metagenomic deep sequencing identifies common and rare pathogens
- Author
-
Doan, Thuy, Wilson, Michael R, Crawford, Emily D, Chow, Eric D, Khan, Lillian M, Knopp, Kristeene A, O’Donovan, Brian D, Xia, Dongxiang, Hacker, Jill K, Stewart, Jay M, Gonzales, John A, Acharya, Nisha R, and DeRisi, Joseph L
- Subjects
Biological Sciences ,Genetics ,Clinical Sciences - Abstract
It has come to our attention that there is an error in Fig. 3a for this article [1]. The correct version of Fig. 3a can be found below. The red markers now reflect the sequence differences. The text is correct. There was also a row omitted in Additional file 1: Table S1. The revised version can be found below.
- Published
- 2016
34. Illuminating uveitis: metagenomic deep sequencing identifies common and rare pathogens
- Author
-
Doan, Thuy, Wilson, Michael R, Crawford, Emily D, Chow, Eric D, Khan, Lillian M, Knopp, Kristeene A, O’Donovan, Brian D, Xia, Dongxiang, Hacker, Jill K, Stewart, Jay M, Gonzales, John A, Acharya, Nisha R, and DeRisi, Joseph L
- Subjects
Biological Sciences ,Genetics ,Eye Disease and Disorders of Vision ,Rare Diseases ,Biodefense ,Emerging Infectious Diseases ,Infectious Diseases ,4.1 Discovery and preclinical testing of markers and technologies ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Infection ,Good Health and Well Being ,Aqueous Humor ,Cryptococcosis ,Cryptococcus neoformans ,Herpes Simplex ,Herpesvirus 1 ,Human ,High-Throughput Nucleotide Sequencing ,Humans ,Metagenomics ,Phylogeny ,Rubella ,Rubella virus ,Toxoplasma ,Toxoplasmosis ,Uvea ,Uveitis ,Virus Replication ,Metagenomic deep sequencing ,Pathogen discovery ,Clinical Sciences - Abstract
BackgroundOcular infections remain a major cause of blindness and morbidity worldwide. While prognosis is dependent on the timing and accuracy of diagnosis, the etiology remains elusive in ~50 % of presumed infectious uveitis cases. The objective of this study is to determine if unbiased metagenomic deep sequencing (MDS) can accurately detect pathogens in intraocular fluid samples of patients with uveitis.MethodsThis is a proof-of-concept study, in which intraocular fluid samples were obtained from five subjects with known diagnoses, and one subject with bilateral chronic uveitis without a known etiology. Samples were subjected to MDS, and results were compared with those from conventional diagnostic tests. Pathogens were identified using a rapid computational pipeline to analyze the non-host sequences obtained from MDS.ResultsUnbiased MDS of intraocular fluid produced results concordant with known diagnoses in subjects with (n = 4) and without (n = 1) uveitis. Samples positive for Cryptococcus neoformans, Toxoplasma gondii, and herpes simplex virus 1 as tested by a Clinical Laboratory Improvement Amendments-certified laboratory were correctly identified with MDS. Rubella virus was identified in one case of chronic bilateral idiopathic uveitis. The subject's strain was most closely related to a German rubella virus strain isolated in 1992, one year before he developed a fever and rash while living in Germany. The pattern and the number of viral identified mutations present in the patient's strain were consistent with long-term viral replication in the eye.ConclusionsMDS can identify fungi, parasites, and DNA and RNA viruses in minute volumes of intraocular fluid samples. The identification of chronic intraocular rubella virus infection highlights the eye's role as a long-term pathogen reservoir, which has implications for virus eradication and emerging global epidemics.
- Published
- 2016
35. Immune modulation with RANKL blockade through denosumab treatment in patients with cancer
- Author
-
Chang, Hewitt, primary, Marquez Garcia, Jaqueline, additional, Chen, Brandon K., additional, Kim, Daniel M., additional, Cheng, Michael L., additional, Liu, Eric V., additional, Yang, Hai, additional, Zhang, Li, additional, Sinha, Meenal, additional, Cheung, Alexander, additional, Kwek, Serena S., additional, Chow, Eric D., additional, Bridge, Mark, additional, Aggarwal, Rahul R., additional, Friedlander, Terence W., additional, Small, Eric J., additional, Anderson, Mark, additional, and Fong, Lawrence, additional
- Published
- 2024
- Full Text
- View/download PDF
36. Predicting Splicing from Primary Sequence with Deep Learning
- Author
-
Jaganathan, Kishore, Kyriazopoulou Panagiotopoulou, Sofia, McRae, Jeremy F., Darbandi, Siavash Fazel, Knowles, David, Li, Yang I., Kosmicki, Jack A., Arbelaez, Juan, Cui, Wenwu, Schwartz, Grace B., Chow, Eric D., Kanterakis, Efstathios, Gao, Hong, Kia, Amirali, Batzoglou, Serafim, Sanders, Stephan J., and Farh, Kyle Kai-How
- Published
- 2019
- Full Text
- View/download PDF
37. D-SPIN constructs gene regulatory network models from multiplexed scRNA-seq data revealing organizing principles of cellular perturbation response
- Author
-
Jiang, Jialong, Chen, Sisi, Tsou, Tiffany, McGinnis, Christopher S., Khazaei, Tahmineh, Zhu, Qin, Park, Jong H., Strazhnik, Inna-Marie, Hanna, John, Chow, Eric D., Sivak, David A., Gartner, Zev J., Thomson, Matt, Jiang, Jialong, Chen, Sisi, Tsou, Tiffany, McGinnis, Christopher S., Khazaei, Tahmineh, Zhu, Qin, Park, Jong H., Strazhnik, Inna-Marie, Hanna, John, Chow, Eric D., Sivak, David A., Gartner, Zev J., and Thomson, Matt
- Abstract
Gene regulatory networks within cells modulate the expression of the genome in response to signals and changing environmental conditions. Reconstructions of gene regulatory networks can reveal the information processing and control principles used by cells to maintain homeostasis and execute cell-state transitions. Here, we introduce a computational framework, D-SPIN, that generates quantitative models of generegulatory networks from single-cell mRNA-seq data sets collected across thousands of distinct perturbation conditions. D-SPIN models the cell as a collection of interacting gene-expression programs, and constructs a probabilistic model to infer regulatory interactions between gene-expression programs and external perturbations. Using large Perturb-seq and drug-response datasets, we demonstrate that D-SPIN models reveal the organization of cellular pathways, sub-functions of macromolecular complexes, and the logic of cellular regulation of transcription, translation, metabolism, and protein degradation in response to gene knockdown perturbations. D-SPIN can also be applied to dissect drug response mechanisms in heterogeneous cell populations, elucidating how combinations of immunomodulatory drugs can induce novel cell states through additive recruitment of gene expression programs. D-SPIN provides a computational framework for constructing interpretable models of gene-regulatory networks to reveal principles of cellular information processing and physiological control.
- Published
- 2023
38. D-SPIN constructs gene regulatory network models from multiplexed scRNA-seq data revealing organizing principles of cellular perturbation response
- Author
-
Jiang, Jialong, primary, Chen, Sisi, additional, Tsou, Tiffany, additional, McGinnis, Christopher S, additional, Khazaei, Tahmineh, additional, Zhu, Qin, additional, Park, Jong H, additional, Strazhnik, Inna-Marie, additional, Hanna, John, additional, Chow, Eric D, additional, Sivak, David A, additional, Gartner, Zev J, additional, and Thomson, Matt, additional
- Published
- 2023
- Full Text
- View/download PDF
39. Subclonal mutations in SETBP1 confer a poor prognosis in juvenile myelomonocytic leukemia
- Author
-
Stieglitz, Elliot, Troup, Camille B., Gelston, Laura C., Haliburton, John, Chow, Eric D., Yu, Kristie B., Akutagawa, Jon, Taylor-Weiner, Amaro N., Liu, Y. Lucy, Wang, Yong-Dong, Beckman, Kyle, Emanuel, Peter D., Braun, Benjamin S., Abate, Adam, Gerbing, Robert B., Alonzo, Todd A., and Loh, Mignon L.
- Published
- 2015
- Full Text
- View/download PDF
40. Optimized CRISPR guide RNA library cloning reduces skew and enables more compact genetic screens
- Author
-
Heo, Seok-Jin, primary, Enriquez, Lauren D., additional, Federman, Scot, additional, Chang, Amy Y., additional, Mace, Rachel, additional, Shevade, Kaivalya, additional, Nguyen, Phuong, additional, Litterman, Adam J., additional, Shafer, Shawn, additional, Przybyla, Laralynne, additional, and Chow, Eric D., additional
- Published
- 2022
- Full Text
- View/download PDF
41. Selection of Macrolide and Non-Macrolide Resistance with Mass Azithromycin Distribution: A Community-Randomized Trial
- Author
-
Doan, Thuy, Worden, Lee, Hinterwirth, Armin, Arzika, Ahmed M., Maliki, Ramatou, Abdou, Amza, Zhong, Lina, Chen, Cindi, Cook, Catherine, Lebas, Elodie, O’Brien, Kieran S., Oldenburg, Catherine E., Chow, Eric D., Porco, Travis C., Lipsitch, Marc, Keenan, Jeremy D., and Lietman, Thomas M.
- Subjects
Male ,Infant ,Sequence Analysis, DNA ,Azithromycin ,Article ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Child, Preschool ,Child Mortality ,Drug Resistance, Bacterial ,Humans ,Mass Drug Administration ,Metagenome ,Female ,Macrolides ,Niger - Abstract
Mass distribution of azithromycin to preschool children twice yearly for 2 years has been shown to reduce childhood mortality in sub-Saharan Africa but at the cost of amplifying macrolide resistance. The effects on the gut resistome, a reservoir of antimicrobial resistance genes in the body, of twice-yearly administration of azithromycin for a longer period are unclear.We investigated the gut resistome of children after they received twice-yearly distributions of azithromycin for 4 years. In the Niger site of the MORDOR trial, we enrolled 30 villages in a concurrent trial in which they were randomly assigned to receive mass distribution of either azithromycin or placebo, offered to all children 1 to 59 months of age every 6 months for 4 years. Rectal swabs were collected at baseline, 36 months, and 48 months for analysis of the participants' gut resistome. The primary outcome was the ratio of macrolide-resistance determinants in the azithromycin group to those in the placebo group at 48 months.Over the entire 48-month period, the mean (±SD) coverage was 86.6±12% in the villages that received placebo and 83.2±16.4% in the villages that received azithromycin. A total of 3232 samples were collected during the entire trial period; of the samples obtained at the 48-month monitoring visit, 546 samples from 15 villages that received placebo and 504 from 14 villages that received azithromycin were analyzed. Determinants of macrolide resistance were higher in the azithromycin group than in the placebo group: 7.4 times as high (95% confidence interval [CI], 4.0 to 16.7) at 36 months and 7.5 times as high (95% CI, 3.8 to 23.1) at 48 months. Continued mass azithromycin distributions also selected for determinants of nonmacrolide resistance, including resistance to beta-lactam antibiotics, an antibiotic class prescribed frequently in this region of Africa.Among villages assigned to receive mass distributions of azithromycin or placebo twice yearly for 4 years, antibiotic resistance was more common in the villages that received azithromycin than in those that received placebo. This trial showed that mass azithromycin distributions may propagate antibiotic resistance. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT02047981.).
- Published
- 2020
42. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution
- Author
-
Massachusetts Institute of Technology. Department of Biology, Yang, Dian, Jones, Matthew G, Naranjo, Santiago, Rideout, William M, Min, Kyung Hoi Joseph, Ho, Raymond, Wu, Wei, Replogle, Joseph M, Page, Jennifer L, Quinn, Jeffrey J, Horns, Felix, Qiu, Xiaojie, Chen, Michael Z, Freed-Pastor, William A, McGinnis, Christopher S, Patterson, David M, Gartner, Zev J, Chow, Eric D, Bivona, Trever G, Chan, Michelle M, Yosef, Nir, Jacks, Tyler, Weissman, Jonathan S, Massachusetts Institute of Technology. Department of Biology, Yang, Dian, Jones, Matthew G, Naranjo, Santiago, Rideout, William M, Min, Kyung Hoi Joseph, Ho, Raymond, Wu, Wei, Replogle, Joseph M, Page, Jennifer L, Quinn, Jeffrey J, Horns, Felix, Qiu, Xiaojie, Chen, Michael Z, Freed-Pastor, William A, McGinnis, Christopher S, Patterson, David M, Gartner, Zev J, Chow, Eric D, Bivona, Trever G, Chan, Michelle M, Yosef, Nir, Jacks, Tyler, and Weissman, Jonathan S
- Abstract
Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth and expansion to neighboring and distal tissues. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma and tracked tumor evolution from single-transformed cells to metastatic tumors at unprecedented resolution. We found that the loss of the initial, stable alveolar-type2-like state was accompanied by a transient increase in plasticity. This was followed by the adoption of distinct transcriptional programs that enable rapid expansion and, ultimately, clonal sweep of stable subclones capable of metastasizing. Finally, tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates progression by creating novel trajectories. Our study elucidates the hierarchical nature of tumor evolution and, more broadly, enables in-depth studies of tumor progression.
- Published
- 2022
43. The MmpL Protein Family
- Author
-
Jain, Madhulika, primary, Chow, Eric D., additional, and Cox, Jeffery S., additional
- Published
- 2014
- Full Text
- View/download PDF
44. Lineage Recording Reveals the Phylodynamics, Plasticity and Paths of Tumor Evolution
- Author
-
Yang, Dian, primary, Jones, Matthew G., additional, Naranjo, Santiago, additional, Rideout, William M., additional, Min, Kyung Hoi (Joseph), additional, Ho, Raymond, additional, Wu, Wei, additional, Replogle, Joseph M., additional, Page, Jennifer L., additional, Quinn, Jeffrey J., additional, Horns, Felix, additional, Qiu, Xiaojie, additional, Chen, Michael Z., additional, Freed-Pastor, William A., additional, McGinnis, Christopher S., additional, Patterson, David M., additional, Gartner, Zev J., additional, Chow, Eric D., additional, Bivona, Trever G., additional, Chan, Michelle M., additional, Yosef, Nir, additional, Jacks, Tyler, additional, and Weissman, Jonathan S., additional
- Published
- 2021
- Full Text
- View/download PDF
45. Systematic Genetic Analysis of Virulence in the Human Fungal Pathogen Cryptococcus neoformans
- Author
-
Liu, Oliver W., Chun, Cheryl D., Chow, Eric D., Chen, Changbin, Madhani, Hiten D., and Noble, Suzanne M.
- Published
- 2008
- Full Text
- View/download PDF
46. Exploration of whole-genome responses of the human AIDS-associated yeast pathogen Cryptococcus neoformans var grubii: nitric oxide stress and body temperature
- Author
-
Chow, Eric D., Liu, Oliver W., O’Brien, Sean, and Madhani, Hiten D.
- Published
- 2007
- Full Text
- View/download PDF
47. Lineage Recording Reveals the Phylodynamics, Plasticity and Paths of Tumor Evolution
- Author
-
Yang, Dian, Jones, Matthew G., Naranjo, Santiago, Rideout, William M., III, Min, Kyung Hoi Joseph, Ho, Raymond, Wu, Wei, Replogle, Joseph M., Page, Jennifer L., Quinn, Jeffrey J., Horns, Felix, Qiu, Xiaojie, Chen, Michael Z., Freed-Pastor, William A., McGinnis, Christopher S., Patterson, David M., Gartner, Zev J., Chow, Eric D., Bivona, Trever G., Chan, Michelle M., Yosef, Nir, Jacks, Tyler, Weissman, Jonathan S., Yang, Dian, Jones, Matthew G., Naranjo, Santiago, Rideout, William M., III, Min, Kyung Hoi Joseph, Ho, Raymond, Wu, Wei, Replogle, Joseph M., Page, Jennifer L., Quinn, Jeffrey J., Horns, Felix, Qiu, Xiaojie, Chen, Michael Z., Freed-Pastor, William A., McGinnis, Christopher S., Patterson, David M., Gartner, Zev J., Chow, Eric D., Bivona, Trever G., Chan, Michelle M., Yosef, Nir, Jacks, Tyler, and Weissman, Jonathan S.
- Abstract
Tumor evolution is driven by the progressive acquisition of genetic and epigenetic alterations that enable uncontrolled growth, expansion to neighboring and distal tissues, and therapeutic resistance. The study of phylogenetic relationships between cancer cells provides key insights into these processes. Here, we introduced an evolving lineage-tracing system with a single-cell RNA-seq readout into a mouse model of Kras;Trp53(KP)-driven lung adenocarcinoma which enabled us to track tumor evolution from single transformed cells to metastatic tumors at unprecedented resolution. We found that loss of the initial, stable alveolar-type2-like state was accompanied by transient increase in plasticity. This was followed by adoption of distinct fitness associated transcriptional programs which enable rapid expansion and ultimately clonal sweep of rare, stable subclones capable of metastasizing to distant sites. Finally, we showed that tumors develop through stereotypical evolutionary trajectories, and perturbing additional tumor suppressors accelerates tumor progression by creating novel evolutionary paths. Overall, our study elucidates the hierarchical nature of tumor evolution, and more broadly enables the in-depth study of tumor progression.
- Published
- 2021
48. Estimation of secondary household attack rates for emergent SARS-CoV-2 variants detected by genomic surveillance at a community-based testing site in San Francisco
- Author
-
Peng, James, primary, Mann, Sabrina A, additional, Mitchell, Anthea M, additional, Liu, Jamin, additional, Laurie, Matthew T., additional, Sunshine, Sara, additional, Pilarowski, Genay, additional, Ayscue, Patrick, additional, Kistler, Amy, additional, Vanaerschot, Manu, additional, Li, Lucy M., additional, McGeever, Aaron, additional, Chow, Eric D., additional, Team, IDseq, additional, Marquez, Carina, additional, Nakamura, Robert, additional, Rubio, Luis, additional, Chamie, Gabriel, additional, Jones, Diane, additional, Jacobo, Jon, additional, Rojas, Susana, additional, Rojas, Susy, additional, Tulier-Laiwa, Valerie, additional, Black, Douglas, additional, Martinez, Jackie, additional, Naso, Jamie, additional, Schwab, Joshua, additional, Petersen, Maya, additional, Havlir, Diane, additional, and DeRisi, Joseph, additional
- Published
- 2021
- Full Text
- View/download PDF
49. Genetic and Mass Spectrometric Tools for Elucidating the Physiological Function(s) of Cytochrome P450 Enzymes from Mycobacterium tuberculosis
- Author
-
Ouellet, Hugues, primary, Chow, Eric D., additional, Guan, Shenheng, additional, Cox, Jeffery S., additional, Burlingame, Alma L., additional, and de Montellano, Paul R. Ortiz, additional
- Published
- 2013
- Full Text
- View/download PDF
50. Rapid pathogen detection by metagenomic next-generation sequencing of infected body fluids
- Author
-
Gu, Wei, primary, Deng, Xianding, additional, Lee, Marco, additional, Sucu, Yasemin D., additional, Arevalo, Shaun, additional, Stryke, Doug, additional, Federman, Scot, additional, Gopez, Allan, additional, Reyes, Kevin, additional, Zorn, Kelsey, additional, Sample, Hannah, additional, Yu, Guixia, additional, Ishpuniani, Gurpreet, additional, Briggs, Benjamin, additional, Chow, Eric D., additional, Berger, Amy, additional, Wilson, Michael R., additional, Wang, Candace, additional, Hsu, Elaine, additional, Miller, Steve, additional, DeRisi, Joseph L., additional, and Chiu, Charles Y., additional
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.