44 results on '"Thomas Eisenhaure"'
Search Results
2. Safety of procuring research tissue during a clinically indicated kidney biopsy from patients with lupus: data from the Accelerating Medicines Partnership RA/SLE Network
- Author
-
Andrew Filer, Michael H Weisman, Judith A James, Kenneth Kalunian, Michelle A Petri, Chaim Putterman, H Michael Belmont, Ilfita Sahbudin, Karim Raza, Maria Dall'Era, Jill P Buyon, Diane L Kamen, Karen Salomon-Escoto, Kazuyoshi Ishigaki, Patrick Dunn, David Wofsy, Michele Bombardieri, Vivian Bykerk, Myles Lewis, Ming Wu, Soumya Raychaudhuri, Hemant Suryawanshi, Thomas Tuschl, Christopher Ritchlin, Maureen McMahon, Jennifer Grossman, Philip M Carlucci, Alessandra Nerviani, Peter M Izmirly, Fan Zhang, Felice Rivellese, Joan Bathon, Zhu Zhu, Qian Xiao, Jessica Li, Holden Maecker, Nir Hacohen, Rong Mao, Jennifer Anolik, Javier Rangel-Moreno, Nida Meednu, Susan Goodman, Lindsy Forbess, Mariko Ishimori, Kevin Deane, David Hildeman, Yuhong Li, Laura Hughes, Robert Clancy, ANNE DAVIDSON, Matthias Kretzler, Larry Moreland, Harris Perlman, Peter Gregersen, Celine C Berthier, Andrea Fava, David Boyle, Derek M Fine, Ami Ben-Artzi, P J Utz, Melanie Smith, Beatrice Goilav, Carla Cuda, Andrew McDavid, Deepak A Rao, Joshua Keegan, Ilya Korsunsky, Joel Guthridge, Kevin Wei, Arnon Arazi, Thomas Eisenhaure, Michael Brenner, Susan Macwana, Pavel Morozov, Manjunath Kustagi, Gerald Watts, Kristina K Deonaraine, Jose Monroy-Trujillo, Mohamed G Atta, Kristin Haag, William Apruzzese, Sean Connery, Fernanda Payan-Schober, Kerry Cho, Jennifer Goff, Aparna Nathan, Joseph Mears, Nghia Millard, Kathryn Weinand, Saori Sakaue, Bill Robinson, Wade DeJager, Louis Bridges, Laura Donlin, Edward DiCarlo, Amit Lakhanpal, Heather Sherman, Anvita Singaraju, Lorien Shakib, Brendan Boyce, Darren Tabechian, Jen Albrecht, James Lederer, A Helena Jonsson, Daimon Simmons, Gregory Keras, Adam Chicoine, Zhihan Jian Li, Mandy McGeachy, Gary Firestein, Arnold Ceponis, Diane Horowitz, Salina Dominguez, Arthur Mandelin, Anjali Thakrar, Mike Holers, Jennifer Seifert, Constanino Pitzalis, Ellen Gravallese, Jennifer Barnas, Raymond Hsu, Steven Woodle, Paul Hoover, Michael Peters, Tony Jones, David Lieb, Jeffrey Hodgin, and Raji Menon
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
Objectives In lupus nephritis the pathological diagnosis from tissue retrieved during kidney biopsy drives treatment and management. Despite recent approval of new drugs, complete remission rates remain well under aspirational levels, necessitating identification of new therapeutic targets by greater dissection of the pathways to tissue inflammation and injury. This study assessed the safety of kidney biopsies in patients with SLE enrolled in the Accelerating Medicines Partnership, a consortium formed to molecularly deconstruct nephritis.Methods 475 patients with SLE across 15 clinical sites in the USA consented to obtain tissue for research purposes during a clinically indicated kidney biopsy. Adverse events (AEs) were documented for 30 days following the procedure and were determined to be related or unrelated by all site investigators. Serious AEs were defined according to the National Institutes of Health reporting guidelines.Results 34 patients (7.2%) experienced a procedure-related AE: 30 with haematoma, 2 with jets, 1 with pain and 1 with an arteriovenous fistula. Eighteen (3.8%) experienced a serious AE requiring hospitalisation; four patients (0.8%) required a blood transfusion related to the kidney biopsy. At one site where the number of cores retrieved during the biopsy was recorded, the mean was 3.4 for those who experienced a related AE (n=9) and 3.07 for those who did not experience any AE (n=140). All related AEs resolved.Conclusions Procurement of research tissue should be considered feasible, accompanied by a complication risk likely no greater than that incurred for standard clinical purposes. In the quest for targeted treatments personalised based on molecular findings, enhanced diagnostics beyond histology will likely be required.
- Published
- 2021
- Full Text
- View/download PDF
3. SLAMF6 deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
- Author
-
Emma Hajaj, Galit Eisenberg, Shiri Klein, Shoshana Frankenburg, Sharon Merims, Inna Ben David, Thomas Eisenhaure, Sarah E Henrickson, Alexandra Chloé Villani, Nir Hacohen, Nathalie Abudi, Rinat Abramovich, Jonathan E Cohen, Tamar Peretz, Andre Veillette, and Michal Lotem
- Subjects
checkpoint ,immunotherapy ,cancer ,T cells ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
SLAMF6 is a homotypic receptor of the Ig-superfamily whose exact role in immune modulation has remained elusive. Its constitutive expression on resting and activated T cells precludes it from being a bona fide exhaustion marker. By breeding Pmel-1 mice with SLAMF6 -/- mice, we generated donors for T cells lacking SLAMF6 and expressing a transgenic TCR for gp100-melanoma antigen. Activated Pmel-1xSLAMF6 -/- CD8+ T cells displayed improved polyfunctionality and strong tumor cytolysis. T-bet was the dominant transcription factor in Pmel-1 x SLAMF6 -/- cells, and upon activation, they acquired an effector-memory phenotype. Adoptive transfer of Pmel-1 x SLAMF6 -/- T cells to melanoma-bearing mice resulted in lasting tumor regression in contrast to temporary responses achieved with Pmel-1 T cells. LAG-3 expression was elevated in the SLAMF6 -/- cells, and the addition of the LAG-3-blocking antibody to the adoptive transfer protocol improved the SLAMF6 -/- T cells and expedited the antitumor response even further. The results from this study support the notion that SLAMF6 is an inhibitory immune receptor whose absence enables powerful CD8+ T cells to eradicate tumors.
- Published
- 2020
- Full Text
- View/download PDF
4. Data from Disabled Homolog 2 Controls Prometastatic Activity of Tumor-Associated Macrophages
- Author
-
Vincenzo Bronte, Peter J. Murray, Silvio Bicciato, Emilio Bria, Luisa Carbognin, Aldo Scarpa, Rita Lawlor, Stefano Piccolo, Luca Azzolin, Nir Hacohen, David Lieb, Siranush Sarkizova, Thomas Eisenhaure, Giulia Borile, Federico Boschi, Massimo Rugge, Matteo Fassan, Manuela Iezzi, Alessia Lamolinara, Anna Simonelli, Stefania Canè, Stefano Ugel, Francesco De Sanctis, Vincenzo Ingangi, Francesca Hofer, Rosalinda Trovato, and Ilaria Marigo
- Abstract
Tumor-associated macrophages (TAM) are regulators of extracellular matrix (ECM) remodeling and metastatic progression, the main cause of cancer-associated death. We found that disabled homolog 2 mitogen-responsive phosphoprotein (DAB2) is highly expressed in tumor-infiltrating TAMs and that its genetic ablation significantly impairs lung metastasis formation. DAB2-expressing TAMs, mainly localized along the tumor-invasive front, participate in integrin recycling, ECM remodeling, and directional migration in a tridimensional matrix. DAB2+ macrophages escort the invasive dissemination of cancer cells by a mechanosensing pathway requiring the transcription factor YAP. In human lobular breast and gastric carcinomas, DAB2+ TAMs correlated with a poor clinical outcome, identifying DAB2 as potential prognostic biomarker for stratification of patients with cancer. DAB2 is therefore central for the prometastatic activity of TAMs.Significance:DAB2 expression in macrophages is essential for metastasis formation but not primary tumor growth. Mechanosensing cues, activating the complex YAP–TAZ, regulate DAB2 in macrophages, which in turn controls integrin recycling and ECM remodeling in 3-D tissue matrix. The presence of DAB2+ TAMs in patients with cancer correlates with worse prognosis.This article is highlighted in the In This Issue feature, p. 1611
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data from Disabled Homolog 2 Controls Prometastatic Activity of Tumor-Associated Macrophages
- Author
-
Vincenzo Bronte, Peter J. Murray, Silvio Bicciato, Emilio Bria, Luisa Carbognin, Aldo Scarpa, Rita Lawlor, Stefano Piccolo, Luca Azzolin, Nir Hacohen, David Lieb, Siranush Sarkizova, Thomas Eisenhaure, Giulia Borile, Federico Boschi, Massimo Rugge, Matteo Fassan, Manuela Iezzi, Alessia Lamolinara, Anna Simonelli, Stefania Canè, Stefano Ugel, Francesco De Sanctis, Vincenzo Ingangi, Francesca Hofer, Rosalinda Trovato, and Ilaria Marigo
- Abstract
Supplementary Figures, Tables, Methods
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data S1 from Disabled Homolog 2 Controls Prometastatic Activity of Tumor-Associated Macrophages
- Author
-
Vincenzo Bronte, Peter J. Murray, Silvio Bicciato, Emilio Bria, Luisa Carbognin, Aldo Scarpa, Rita Lawlor, Stefano Piccolo, Luca Azzolin, Nir Hacohen, David Lieb, Siranush Sarkizova, Thomas Eisenhaure, Giulia Borile, Federico Boschi, Massimo Rugge, Matteo Fassan, Manuela Iezzi, Alessia Lamolinara, Anna Simonelli, Stefania Canè, Stefano Ugel, Francesco De Sanctis, Vincenzo Ingangi, Francesca Hofer, Rosalinda Trovato, and Ilaria Marigo
- Abstract
Marker genes for each cell type identified in tumor-infiltrating myeloid cells.Related to Fig.1A
- Published
- 2023
- Full Text
- View/download PDF
7. 1242 Altered interactions between circulating and tissue-resident CD8 T cells with the colonic mucosa define checkpoint inhibitor colitis
- Author
-
Kamil Slowikowski, Molly Thomas, Kasidet Manakongtreecheep, Pritha Sen, Jessica Tantivit, Mazen Nasrallah, Leyre Zubiri, Neal Smith, Alice Tirard, Swetha Ramesh, Benjamin Arnold, Linda Nieman, Jonathan Chen, Thomas Eisenhaure, Karin Pelka, Katherine Xu, Vjola Jorgji, Christopher Pinto, Tatyana Sharova, Rachel Glasser, PuiYee Chan, Ryan Sullivan, Hamed Khalili, Dejan Juric, Genevieve Boland, Michael Dougan, Nir Hacohen, Bo Li, Kerry Reynolds, and Alexandra-Chloé Villani
- Published
- 2022
- Full Text
- View/download PDF
8. Differentiation of exhausted CD8+ T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory
- Author
-
Joelle Brown, Hannah K. Drescher, Jenna L. Gustafson, Marcos Damasio, Sonu Subudhi, W. Nicholas Haining, David Wolski, Georg M. Lauer, Lucile Massenet-Regad, Maxwell Robidoux, Pierre Tonnerre, Daniel Kvistad, Jihad Aljabban, Arthur Y. Kim, Todd M. Allen, Jasneet Aneja, Lia Laura Lewis-Ximenez, James A. Cheney, Raymond T. Chung, David J. Bean, Damien C. Tully, Nir Hacohen, Nadia Alatrakchi, Thomas Eisenhaure, David J. Lieb, Lea M. Bartsch, Almudena Torres-Cornejo, Ruben C. Hoogeveen, Ang Cui, and Debattama R. Sen
- Subjects
T cell ,Immunology ,Lymphocyte differentiation ,Stimulation ,Hepatitis C ,Biology ,medicine.disease ,complex mixtures ,Phenotype ,medicine.anatomical_structure ,Antigen ,Antigen stimulation ,medicine ,Malignant cells ,Immunology and Allergy ,Cytotoxic T cell ,CD8 - Abstract
T cell exhaustion is associated with failure to clear chronic infections and malignant cells. Defining the molecular mechanisms of T cell exhaustion and reinvigoration is essential to improving immunotherapeutic modalities. Here we confirmed pervasive phenotypic, functional and transcriptional differences between memory and exhausted antigen-specific CD8+ T cells in human hepatitis C virus (HCV) infection before and after treatment. After viral cure, phenotypic changes in clonally stable exhausted T cell populations suggested differentiation toward a memory-like profile. However, functionally, the cells showed little improvement, and critical transcriptional regulators remained in the exhaustion state. Notably, T cells from chronic HCV infection that were exposed to antigen for less time because of viral escape mutations were functionally and transcriptionally more similar to memory T cells from spontaneously resolved HCV infection. Thus, the duration of T cell stimulation impacts exhaustion recovery, with antigen removal after long-term exhaustion being insufficient for the development of functional T cell memory. Lauer and colleagues examine CD8+ T cells following cure of human hepatitis C virus (HCV) infection. CD8+ T cells exposed to chronic HCV-specific activation show durable functional, phenotypic and transcriptional exhaustion that is maintained even after antigen stimulus is removed.
- Published
- 2021
- Full Text
- View/download PDF
9. Altered interactions between circulating and tissue-resident CD8 T cells with the colonic mucosa define colitis associated with immune checkpoint inhibitors
- Author
-
Kamil Slowikowski, Benjamin Y. Arnold, Leyre Zubiri, Katherine Xu, Bo Li, Dejan Juric, Christopher J. Pinto, Swetha Ramesh, Michael Dougan, Jonathan H. Chen, Tatyana Sharova, Genevieve M. Boland, Karin Pelka, Elaina PuiYee Chan, Linda T. Nieman, Molly Thomas, Jessica Tantivit, Pritha Sen, Hamed Khalili, Alice Tirard, Mazen Nasrallah, Rachel Glasser, Alexandra-Chloé Villani, Neal P. Smith, Vjola Jorgji, Ryan J. Sullivan, Kasidet Manakongtreecheep, Kerry L. Reynolds, Nir Hacohen, and Thomas Eisenhaure
- Subjects
Chemokine ,Immune system ,medicine.anatomical_structure ,biology ,T cell ,medicine ,biology.protein ,Cancer research ,Cytotoxic T cell ,Stem cell ,Stem cell marker ,CD8 ,Immune tolerance - Abstract
Therapeutic blockade of co-inhibitory immune receptors PD-1 and CTLA-4 has revolutionized oncology, but treatments are limited by immune-related adverse events (IRAEs). IRAE Colitis (irColitis) is the most common, severe IRAE affecting up to 25% of patients on dual PD-1 and CTLA-4 inhibition. Here, we present a systems biology approach to define the cell populations and transcriptional programs driving irColitis. We collected paired colon mucosal biopsy and blood specimens from 13 patients with irColitis, 8 healthy individuals, and 8 controls on immune checkpoint inhibitors (ICIs), and analyzed them with single-cell/nuclei RNA sequencing with paired TCR and BCR sequencing, multispectral fluorescence microscopy, and secreted factor analysis (Luminex). We profiled 299,407 cells from tissue and blood and identified 105 cell subsets that revealed significant tissue remodeling in active disease. Colon mucosal immune populations were dominated by tissue-resident memory (TRM) ITGAE-expressing CD8 T cells representing a phenotypic spectrum defined by gene programs associated with T cell activation, cytotoxicity, cycling, and exhaustion. CD8 TRM and effector CD4 T cells upregulated type 17 immune programs (IL17A, IL26) and Tfh-like programs (CXCL13, PDCD1). We also identified for the first time an increased abundance of two KLRG1 and ITGB2-expressing CD8 T cell populations with circulatory cell markers, including a GZMK TRM-like population and a CX3CR1 population that is predicted to be intravascular. These two populations were more abundant in irColitis patients treated with dual PD-1/CTLA-4 inhibition than those receiving anti-PD-1 monotherapy. They also had significant TCR sharing with PBMCs, suggesting a circulatory origin. In irColitis we observed significant epithelial turnover marked by fewer LGR5-expressing stem cells, more transit amplifying cells, and upregulation of apoptotic and DNA-sensing programs such as the cGAS-STING pathway. Mature epithelial cells with top crypt genes upregulated interferon-stimulated pathways, CD274 (PD-L1), anti-microbial genes, and MHC-class II genes, and downregulated aquaporin and solute-carrier gene families, likely contributing to epithelial cell damage and absorptive dysfunction. Mesenchymal remodeling was defined by increased endothelial cells, both in irColitis patients and specifically in patients on dual PD-1/CTLA-4 blockade. Cell-cell communication analysis identified putative receptor-ligand pairs that recruit CD8 T cells from blood to inflamed endothelium and positive feedback loops such as the CXCR3 chemokine system that retain cells in tissue. This study highlights the cellular and molecular drivers underlying irColitis and provides new insights into the role of CTLA-4 and PD-1 signaling in maintaining CD8 TRM homeostasis, regulating CD8 T recruitment from blood, and promoting epithelial-immune crosstalk critical to gastrointestinal immune tolerance and intestinal barrier function.
- Published
- 2021
- Full Text
- View/download PDF
10. Genome-wide enhancer maps link risk variants to disease genes
- Author
-
Tejal A. Patwardhan, Fritz Lekschas, Jesse M. Engreitz, Alkes L. Price, Hanspeter Pfister, Mark J. Daly, Glen Munson, Michael Kane, Jacob C. Ulirsch, Helen Y. Kang, Nir Hacohen, Ramnik J. Xavier, Anshul Kundaje, Heini M. Natri, Elle M. Weeks, Tung T. Nguyen, Drew T. Bergman, Benjamin R. Doughty, Thouis R. Jones, Eric S. Lander, Joseph Nasser, Kristy Mualim, Thomas Eisenhaure, Ryan L. Collins, Philine Guckelberger, Kushal K. Dey, John P. Ray, Ang Cui, Hilary K. Finucane, Charles P. Fulco, Hailiang Huang, Charles B. Epstein, and Institute for Molecular Medicine Finland
- Subjects
Male ,Genomics ,Genome-wide association study ,Computational biology ,Biology ,Genome ,Article ,Cell Line ,03 medical and health sciences ,Cyclophilins ,0302 clinical medicine ,CRISPR ,Humans ,Genetic Predisposition to Disease ,Enhancer ,Gene ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Translational bioinformatics ,Chromosomes, Human, Pair 10 ,Genome, Human ,Macrophages ,1184 Genetics, developmental biology, physiology ,Genetic Variation ,Dendritic Cells ,Inflammatory Bowel Diseases ,Human genetics ,Mitochondria ,Enhancer Elements, Genetic ,Phenotype ,Organ Specificity ,Female ,3111 Biomedicine ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer–gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions. Mapping enhancer regulation across human cell types and tissues illuminates genome function and provides a resource to connect risk variants for common diseases to their molecular and cellular functions.
- Published
- 2021
11. An efficient lentiviral CRISPRi approach to silence genes in primary human monocytes
- Author
-
Thomas Eisenhaure, Bingxu Liu, Matteo Gentili, Darrell J. Irvine, David J. Lieb, and Nir Hacohen
- Subjects
Immune system ,biology ,Interferon ,Lentivirus ,medicine ,Guide RNA ,biology.organism_classification ,Genome ,Gene ,Tissue homeostasis ,Cell biology ,Proinflammatory cytokine ,medicine.drug - Abstract
Human primary monocytes are critical in controlling immune responses and tissue homeostasis. However, to identify and study molecular components that underlie the function of these cells, there remains a need to develop methods to perturb genes in these cells. Here we report lentiviral-based delivery of dCas9-KRAB and guide RNAs to efficiently silent target genes, such as CD45 and CD209. We further show that sgRNAs against TICAM1 dampen proinflammatory cytokine and interferon expression in response to lipopolysaccharide. When delivered by lentivirus, sgRNAs are incorporated into the genome, thus enabling pooled screening to perturb and identify coding and non-coding elements that contribute to the functions of primary human monocytes.
- Published
- 2020
- Full Text
- View/download PDF
12. Loss of the Nuclear Protein RTF2 Enhances Influenza Virus Replication
- Author
-
Nir Hacohen, Ang Cui, Raktima Raychowdhury, David J. Lieb, Bo Li, Bing Shao Chia, and Thomas Eisenhaure
- Subjects
viruses ,Cellular Response to Infection ,Cell Cycle Proteins ,medicine.disease_cause ,Virus Replication ,influenza virus ,Gene Knockout Techniques ,0302 clinical medicine ,Interferon ,Transcription (biology) ,Chlorocebus aethiops ,Influenza A virus ,CRISPR ,Nuclear protein ,Spotlight ,innate immunity ,0303 health sciences ,Nuclear Proteins ,interferon ,antiviral ,DNA-Binding Proteins ,Host-Pathogen Interactions ,Interferon Type I ,transcription ,medicine.drug ,Immunology ,restriction factor ,Biology ,Microbiology ,Antiviral Agents ,Virus ,RTF2 ,Cell Line ,03 medical and health sciences ,Viral Proteins ,Viral life cycle ,interferon-stimulated genes ,Virology ,Influenza, Human ,medicine ,Animals ,Humans ,Vero Cells ,030304 developmental biology ,Interferon-beta ,Immunity, Innate ,HEK293 Cells ,Viral replication ,A549 Cells ,Insect Science ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Viral infection triggers the secretion of type I interferons, which in turn induce the expression of hundreds of antiviral genes. However, the roles of these induced genes in controlling viral infections remain largely unknown, limiting our ability to develop host-based antiviral therapeutics against pathogenic viruses, such as influenza virus. Here, we performed a loss-of-function genetic CRISPR screen in cells prestimulated with type I interferon to identify antiviral genes that restrict influenza A virus replication. Besides finding key components of the interferon signaling pathway, we discovered a new restriction factor, RTF2, which acts in the nucleus, restricts influenza virus transcription, and contributes to the interferon-induced upregulation of known restriction factors. Our work contributes to the field of antiviral immunology by discovering and characterizing a novel restriction factor of influenza virus and may ultimately be useful for understanding how to control a virus that causes significant morbidity and mortality worldwide., While hundreds of genes are induced by type I interferons, their roles in restricting the influenza virus life cycle remain mostly unknown. Using a loss-of-function CRISPR screen in cells prestimulated with interferon beta (IFN-β), we identified a small number of factors required for restricting influenza A virus replication. In addition to known components of the interferon signaling pathway, we found that replication termination factor 2 (RTF2) restricts influenza virus at the nuclear stage (and perhaps other stages) of the viral life cycle, based on several lines of evidence. First, a deficiency in RTF2 leads to higher levels of viral primary transcription, even in the presence of cycloheximide to block genome replication and secondary transcription. Second, cells that lack RTF2 have enhanced activity of a viral reporter that depends solely on four viral proteins that carry out replication and transcription in the nucleus. Third, when the RTF2 protein is mislocalized outside the nucleus, it is not able to restrict replication. Finally, the absence of RTF2 leads not only to enhanced viral transcription but also to reduced expression of antiviral factors in response to interferon. RTF2 thus inhibits primary influenza virus transcription, likely acts in the nucleus, and contributes to the upregulation of antiviral effectors in response to type I interferons. IMPORTANCE Viral infection triggers the secretion of type I interferons, which in turn induce the expression of hundreds of antiviral genes. However, the roles of these induced genes in controlling viral infections remain largely unknown, limiting our ability to develop host-based antiviral therapeutics against pathogenic viruses, such as influenza virus. Here, we performed a loss-of-function genetic CRISPR screen in cells prestimulated with type I interferon to identify antiviral genes that restrict influenza A virus replication. Besides finding key components of the interferon signaling pathway, we discovered a new restriction factor, RTF2, which acts in the nucleus, restricts influenza virus transcription, and contributes to the interferon-induced upregulation of known restriction factors. Our work contributes to the field of antiviral immunology by discovering and characterizing a novel restriction factor of influenza virus and may ultimately be useful for understanding how to control a virus that causes significant morbidity and mortality worldwide.
- Published
- 2020
13. Differentiation of exhausted CD8
- Author
-
Pierre, Tonnerre, David, Wolski, Sonu, Subudhi, Jihad, Aljabban, Ruben C, Hoogeveen, Marcos, Damasio, Hannah K, Drescher, Lea M, Bartsch, Damien C, Tully, Debattama R, Sen, David J, Bean, Joelle, Brown, Almudena, Torres-Cornejo, Maxwell, Robidoux, Daniel, Kvistad, Nadia, Alatrakchi, Ang, Cui, David, Lieb, James A, Cheney, Jenna, Gustafson, Lia L, Lewis-Ximenez, Lucile, Massenet-Regad, Thomas, Eisenhaure, Jasneet, Aneja, W Nicholas, Haining, Raymond T, Chung, Nir, Hacohen, Todd M, Allen, Arthur Y, Kim, and Georg M, Lauer
- Subjects
T cell exhaustion ,Cell Differentiation ,Hepacivirus ,CD8-Positive T-Lymphocytes ,Hepatitis C, Chronic ,complex mixtures ,Antiviral Agents ,Article ,HCV infection ,Epitopes ,Phenotype ,immunological recovery ,antiviral therapy ,Humans ,Immunologic Memory - Abstract
T cell exhaustion is associated with failure to clear chronic infections and malignant cells. Defining the molecular mechanisms of T cell exhaustion and reinvigoration is essential to improving immunotherapeutic modalities. Here we confirmed pervasive phenotypic, functional, and transcriptional differences between memory and exhausted antigen-specific CD8+ T cells in human hepatitis C virus (HCV) infection before and after treatment. After viral cure, phenotypic changes in clonally stable exhausted T cell populations suggested differentiation towards a memory-like profile. However, functionally, the cells showed little improvement and critical transcriptional regulators remained in the exhaustion state. Notably, T cells from chronic HCV infection that were exposed to antigen for less time because of viral escape mutations were functionally and transcriptionally more similar to memory T cells from spontaneously resolved HCV infection. Thus, T cell stimulation duration impacts exhaustion recovery, with antigen removal after long-term exhaustion being insufficient for development of functional T cell memory.
- Published
- 2020
14. An immune cell signature of bacterial sepsis
- Author
-
Deborah T. Hung, Michael R. Filbin, Kianna Billman, Roby P. Bhattacharyya, Bruce D. Levy, Thomas Eisenhaure, Nir Hacohen, Marcia B. Goldberg, Paul C. Blainey, Rebecca M. Baron, and Miguel Reyes
- Subjects
0301 basic medicine ,Transcription, Genetic ,CD14 ,Bone Marrow Cells ,Biology ,medicine.disease_cause ,Peripheral blood mononuclear cell ,General Biochemistry, Genetics and Molecular Biology ,Article ,Monocytes ,Epigenesis, Genetic ,Sepsis ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,Bacteria ,Sequence Analysis, RNA ,Monocyte ,Gene Expression Profiling ,Reproducibility of Results ,General Medicine ,Gene signature ,Immune dysregulation ,medicine.disease ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,ROC Curve ,030220 oncology & carcinogenesis ,Immunology ,Biomarkers - Abstract
Dysregulation of the immune response to bacterial infection can lead to sepsis, a condition with high mortality. Multiple whole-blood gene-expression studies have defined sepsis-associated molecular signatures, but have not resolved changes in transcriptional states of specific cell types. Here, we used single-cell RNA-sequencing to profile the blood of people with sepsis (n = 29) across three clinical cohorts with corresponding controls (n = 36). We profiled total peripheral blood mononuclear cells (PBMCs, 106,545 cells) and dendritic cells (19,806 cells) across all subjects and, on the basis of clustering of their gene-expression profiles, defined 16 immune-cell states. We identified a unique CD14+ monocyte state that is expanded in people with sepsis and validated its power in distinguishing these individuals from controls using public transcriptomic data from subjects with different disease etiologies and from multiple geographic locations (18 cohorts, n = 1,467 subjects). We identified a panel of surface markers for isolation and quantification of the monocyte state and characterized its epigenomic and functional phenotypes, and propose a model for its induction from human bone marrow. This study demonstrates the utility of single-cell genomics in discovering disease-associated cytologic signatures and provides insight into the cellular basis of immune dysregulation in bacterial sepsis. Single-cell transcriptomic analysis identifies a distinct gene signature associated with peripheral monocyte populations that distinguishes people with sepsis from those with sterile inflammation and uninfected controls.
- Published
- 2020
15. Genome-wide CRISPR screen identifies host dependency factors for influenza A virus infection
- Author
-
Syed I. A. Bukhari, Sara Clohisey, John G. Doench, Paul Hoover, Paul Digard, Nicholas J. Parkinson, Daniel Lingwood, Maya Sangesland, Bo Wang, Tim Regan, Thomas Eisenhaure, Nir Hacohen, Irit Gat-Viks, Shobha Vasudevan, Bo Li, Nikki Smith, Lawrence D. Schweitzer, David H. Dockrell, David Farr, Bing Shao Chia, Michael U. Gutmann, Andy Law, Ang Cui, J Kenneth Baillie, and Aharon Nachshon
- Subjects
0301 basic medicine ,CRISPR-Cas9 genome editing ,Pyridines ,viruses ,General Physics and Astronomy ,medicine.disease_cause ,Genome ,0302 clinical medicine ,Influenza A virus ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,lcsh:Science ,Multidisciplinary ,Triazines ,3. Good health ,Host-Pathogen Interactions ,Thiepins ,Dibenzothiepins ,Vacuolar Proton-Translocating ATPases ,Pyridones ,Science ,Morpholines ,Nerve Tissue Proteins ,Computational biology ,Biology ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,Cap snatching ,Cell Line ,03 medical and health sciences ,Viral entry ,Influenza, Human ,Oxazines ,medicine ,Humans ,Genetic Predisposition to Disease ,Adaptor Proteins, Signal Transducing ,Cas9 ,Host (biology) ,Membrane Proteins ,General Chemistry ,Methyltransferases ,Virus Internalization ,030104 developmental biology ,A549 Cells ,lcsh:Q ,CRISPR-Cas Systems ,Influenza virus ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Host dependency factors that are required for influenza A virus infection may serve as therapeutic targets as the virus is less likely to bypass them under drug-mediated selection pressure. Previous attempts to identify host factors have produced largely divergent results, with few overlapping hits across different studies. Here, we perform a genome-wide CRISPR/Cas9 screen and devise a new approach, meta-analysis by information content (MAIC) to systematically combine our results with prior evidence for influenza host factors. MAIC out-performs other meta-analysis methods when using our CRISPR screen as validation data. We validate the host factors, WDR7, CCDC115 and TMEM199, demonstrating that these genes are essential for viral entry and regulation of V-type ATPase assembly. We also find that CMTR1, a human mRNA cap methyltransferase, is required for efficient viral cap snatching and regulation of a cell autonomous immune response, and provides synergistic protection with the influenza endonuclease inhibitor Xofluza., Here, Li et al. perform a genome-wide CRISPR screen to identify host dependency factors for influenza A virus infection and show that the host mRNA cap methyltransferase CMTR1 is important for viral cap snatching and that it affects expression of antiviral genes.
- Published
- 2020
- Full Text
- View/download PDF
16. A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression
- Author
-
Sachet A. Shukla, Edward P. Browne, Hung C. Pham, Thomas Eisenhaure, Vassiliki A. Boussiotis, Catherine J. Wu, Apoorvi Chaudhri, Gordon J. Freeman, Baogong Zhu, Nikolaos Patsoukis, Nir Hacohen, Ping Hua, Xia Bu, William F. Pendergraft, Arnon Arazi, Kathleen M. Mahoney, and Edward F. Fritsch
- Subjects
0301 basic medicine ,PD-L1 ,Cancer Research ,Lymphocyte ,Placenta ,RNA Splicing ,Immunology ,Cell ,B7-H1 Antigen ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Immune system ,Splice variants ,Pregnancy ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Humans ,splice ,Receptor ,biology ,Chemistry ,Gene Expression Profiling ,Myeloid-Derived Suppressor Cells ,Immune checkpoint ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Original Article ,Female ,Protein Multimerization ,Isoforms ,Immunosuppressive Agents - Abstract
Targeting immune checkpoint pathways, such as programmed death ligand-1 (PD-L1, also known as CD274 or B7-H1) or its receptor programmed cell death-1 (PD-1) has shown improved survival for patients with numerous types of cancers, not limited to lung cancer, melanoma, renal cell carcinoma, and Hodgkin lymphoma. PD-L1 is a co-inhibitory molecule whose expression on the surface of tumor cells is associated with worse prognosis in many tumors. Here we describe a splice variant (secPD-L1) that does not splice into the transmembrane domain, but instead produces a secreted form of PD-L1 that has a unique 18 amino acid tail containing a cysteine that allows it to homodimerize and more effectively inhibit lymphocyte function than monomeric soluble PD-L1. We show that recombinant secPD-L1 can dimerize and inhibit T-cell proliferation and IFN-gamma production in vitro. The secPD-L1 variant is expressed by malignant cells in vitro that also express high levels of full-length PD-L1. Transcriptomic analysis of gene expression across The Cancer Genome Atlas found the strongest association of secPD-L1 with full-length PD-L1, but also with subsets of immunologic genes, such as in myeloid-derived suppressor cells. Moreover, the splice variant is also expressed in normal tissues and within normal peripheral blood cells it is preferentially expressed in activated myeloid cells. This is the first report of a form of secreted PD-L1 that homodimerizes and is functionally active. SecPD-L1 may function as a paracrine negative immune regulator within the tumor, since secPD-L1 does not require a cell-to-cell interaction to mediate its inhibitory effect. Electronic supplementary material The online version of this article (10.1007/s00262-018-2282-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
17. An Integrative Framework Reveals Signaling-to-Transcription Events in Toll-like Receptor Signaling
- Author
-
Karl R. Clauser, Thomas Eisenhaure, Takashi Satoh, Nir Hacohen, Aviv Regev, Shizuo Akira, Steven A. Carr, Dariusz Przybylski, Tanja Maritzen, Jana Qiao, Volker Haucke, Nicolas Chevrier, Philipp Mertins, Nir Yosef, Raktima Raychowdhury, Broad Institute of MIT and Harvard, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Mertins, Philipp, Przybylski, Dariusz, Clauser, Karl R, Eisenhaure, Thomas, Carr, Steven A, Regev, Aviv, Hacohen, Nir, and Chevrier, Nicolas
- Subjects
0301 basic medicine ,protein-protein interactions ,Computational biology ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,large-scale in vitro kinase assay ,PICALM ,Protein–protein interaction ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Humans ,TLRs ,transcriptional network analysis ,Phosphorylation ,lcsh:QH301-705.5 ,Toll-like receptor ,Toll-Like Receptors ,pathogen-sensing pathways ,Phosphoproteomics ,phosphoproteomics ,Dendritic Cells ,Cell biology ,Vesicular transport protein ,030104 developmental biology ,lcsh:Biology (General) ,Technology Platforms ,Signal transduction ,signaling ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Building an integrated view of cellular responses to environmental cues remains a fundamental challenge due to the complexity of intracellular networks in mammalian cells. Here, we introduce an integrative biochemical and genetic framework to dissect signal transduction events using multiple data types and, in particular, to unify signaling and transcriptional networks. Using the Toll-like receptor (TLR) system as a model cellular response, we generate multifaceted datasets on physical, enzymatic, and functional interactions and integrate these data to reveal biochemical paths that connect TLR4 signaling to transcription. We define the roles of proximal TLR4 kinases, identify and functionally test two dozen candidate regulators, and demonstrate a role for Ap1ar (encoding the Gadkin protein) and its binding partner, Picalm, potentially linking vesicle transport with pro-inflammatory responses. Our study thus demonstrates how deciphering dynamic cellular responses by integrating datasets on various regulatory layers defines key components and higher-order logic underlying signaling-to-transcription pathways. Keywords: pathogen-sensing pathways; Toll-like receptors; TLRs; phosphoproteomics; protein-protein interactions; large-scale in vitro kinase assay; signaling; transcriptional network analysis, National Institutes of Health (U.S.) (Grant U54 AI057159), National Institutes of Health (U.S.) (Award DP2 OD002230), National Institutes of Health (U.S.) (Award P50 HG006193)
- Published
- 2017
- Full Text
- View/download PDF
18. Mass Spectrometry Profiling of HLA-Associated Peptidomes in Mono-allelic Cells Enables More Accurate Epitope Prediction
- Author
-
Guang Lan Zhang, Wandi Zhang, Siranush Sarkizova, Catherine J. Wu, Derin B. Keskin, William J. Lane, Nir Hacohen, Michael S. Rooney, Jennifer G. Abelin, Karl R. Clauser, Jonathan Stevens, John Sidney, Steven A. Carr, Thomas Eisenhaure, and Christina R. Hartigan
- Subjects
0301 basic medicine ,Subdominant ,Immunology ,Antigen presentation ,Human leukocyte antigen ,Biology ,Tandem mass spectrometry ,Article ,Epitope ,Cell Line ,Epitopes ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,Humans ,Immunology and Allergy ,Protein Interaction Domains and Motifs ,Allele ,Gene ,Alleles ,Genetics ,Antigen Presentation ,Gene Expression Profiling ,Histocompatibility Antigens Class I ,Gene expression profiling ,030104 developmental biology ,Infectious Diseases ,030220 oncology & carcinogenesis ,Neural Networks, Computer ,Peptides ,Algorithms ,Chromatography, Liquid - Abstract
Identification of human leukocyte antigen (HLA)-bound peptides by liquid chromatography-tandem mass spectrometry (LC-MS/MS) is poised to provide a deep understanding of rules underlying antigen presentation. However, a key obstacle is the ambiguity that arises from the co-expression of multiple HLA alleles. Here, we have implemented a scalable mono-allelic strategy for profiling the HLA peptidome. By using cell lines expressing a single HLA allele, optimizing immunopurifications, and developing an application-specific spectral search algorithm, we identified thousands of peptides bound to 16 different HLA class I alleles. These data enabled the discovery of subdominant binding motifs and an integrative analysis quantifying the contribution of factors critical to epitope presentation, such as protein cleavage and gene expression. We trained neural-network prediction algorithms with our large dataset (>24,000 peptides) and outperformed algorithms trained on data-sets of peptides with measured affinities. We thus demonstrate a strategy for systematically learning the rules of endogenous antigen presentation.
- Published
- 2017
- Full Text
- View/download PDF
19. SLAMF6 deficiency augments tumor killing and skews toward an effector phenotype revealing it as a novel T cell checkpoint
- Author
-
Emma Hajaj, Michal Lotem, Nir Hacohen, Inna Ben David, Rinat Abramovich, Jonathan Cohen, Nathalie Abudi, Tamar Peretz, Alexandra-Chloé Villani, Thomas Eisenhaure, Sharon Merims, Sarah E. Henrickson, Shiri Klein, Galit Eisenberg, André Veillette, and Shoshana Frankenburg
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Mouse ,medicine.medical_treatment ,Melanoma, Experimental ,Apoptosis ,Immune receptor ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Mice ,0302 clinical medicine ,Immunology and Inflammation ,checkpoint ,Cytotoxic T cell ,Biology (General) ,0303 health sciences ,Effector ,General Neuroscience ,General Medicine ,Natural killer T cell ,Cell biology ,3. Good health ,medicine.anatomical_structure ,Medicine ,immunotherapy ,medicine.symptom ,Research Article ,Human ,QH301-705.5 ,Transgene ,Science ,T cell ,T cells ,Inflammation ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,Antigen ,Signaling Lymphocytic Activation Molecule Family ,medicine ,Animals ,Humans ,cancer ,Human Biology and Medicine ,030304 developmental biology ,General Immunology and Microbiology ,business.industry ,T-cell receptor ,Immunotherapy ,medicine.disease ,Mice, Inbred C57BL ,Cytolysis ,030104 developmental biology ,Cancer cell ,Cancer research ,Skin cancer ,business ,030215 immunology - Abstract
SLAMF6 is a homotypic receptor of the Ig-superfamily whose exact role in immune modulation has remained elusive. Its constitutive expression on resting and activated T cells precludes it from being a bona fide exhaustion marker. By breeding Pmel-1 mice with SLAMF6 -/- mice, we generated donors for T cells lacking SLAMF6 and expressing a transgenic TCR for gp100-melanoma antigen. Activated Pmel-1xSLAMF6 -/- CD8+ T cells displayed improved polyfunctionality and strong tumor cytolysis. T-bet was the dominant transcription factor in Pmel-1 x SLAMF6 -/- cells, and upon activation, they acquired an effector-memory phenotype. Adoptive transfer of Pmel-1 x SLAMF6 -/- T cells to melanoma-bearing mice resulted in lasting tumor regression in contrast to temporary responses achieved with Pmel-1 T cells. LAG-3 expression was elevated in the SLAMF6 -/- cells, and the addition of the LAG-3-blocking antibody to the adoptive transfer protocol improved the SLAMF6 -/- T cells and expedited the antitumor response even further. The results from this study support the notion that SLAMF6 is an inhibitory immune receptor whose absence enables powerful CD8+ T cells to eradicate tumors., eLife digest The immune system helps to protect our bodies from illnesses and infections. Immunotherapies are medicines designed to treat diseases, such as cancer, by boosting the immune system against the condition. This is a powerful approach but so far immunotherapies have only had partial success and there is a need for further improvements. One protein called SLAMF6 is found on cells from the immune system that attack and kill cancer cells. Immunotherapies that suppress SLAMF6 on immune cells called killer T cells could increase immune system activity helping to treat cancers, particularly melanoma skin cancers. So far the potential for SLAMF6 as a target for immunotherapy has not been fully explored. Hajaj et al. created mice with killer T cells that recognized skin cancer cells and lacked SLAMF6. These modified cells were better at fighting cancer, producing more anti-cancer chemicals called cytokines and killing more cancer cells. The modified cells had a lasting effect on tumors and helped the mice to live longer. The effects could be further boosted by treating the mice in combination with other immunotherapies. SLAMF6 is a possible new target for skin cancer immunotherapy that could help more people to live longer following cancer diagnosis. The next step is to create a drug to target SLAMF6 in humans and to test it in clinical trials.
- Published
- 2019
20. The immune cell landscape in kidneys of patients with lupus nephritis
- Author
-
William F. Pendergraft, Richard A. Furie, Fan Zhang, Diane L. Kamen, Jennifer H. Anolik, Jill P. Buyon, Soumya Raychaudhuri, Yanyan Liu, Elena Massarotti, Kamil Slowikowski, Chaim Putterman, James A. Lederer, Michael B. Brenner, Arnon Arazi, Akiko Noma, Betty Diamond, Patti Tosta, Matthias Kretzler, Kenneth C. Kalunian, David A. Hildeman, Meyeon Park, Edward P. Browne, Shuqiang Li, Thomas Eisenhaure, Celine C. Berthier, David Wofsy, Anne Davidson, Paul Hoover, William Apruzzese, Chad Nusbaum, Nir Hacohen, E. Steve Woodle, Elizabeth A. McInnis, David J. Lieb, Scott Steelman, A. Helena Jonsson, Maria Dall'Era, Deepak A. Rao, Fernanda Payan-Schober, Dawn E. Smilek, Danielle Sutherby, Michelle Petri, and Adam Chicoine
- Subjects
0301 basic medicine ,Kidney Disease ,Biopsy ,Lupus nephritis ,Kidney ,Chemokine receptor ,0302 clinical medicine ,Immunophenotyping ,Interferon ,Leukocytes ,Immunology and Allergy ,Cluster Analysis ,2.1 Biological and endogenous factors ,Myeloid Cells ,Lymphocytes ,Aetiology ,Flow Cytometry ,Lupus Nephritis ,3. Good health ,Monocyte differentiation ,Single-Cell Analysis ,medicine.drug ,1.1 Normal biological development and functioning ,Accelerating Medicines Partnership in SLE network ,Immunology ,Renal and urogenital ,Lupus ,Autoimmune Disease ,03 medical and health sciences ,Immune system ,Clinical Research ,Underpinning research ,medicine ,Genetics ,Humans ,Autoimmune disease ,business.industry ,Gene Expression Profiling ,Inflammatory and immune system ,Kidney metabolism ,Computational Biology ,Epithelial Cells ,Molecular Sequence Annotation ,medicine.disease ,030104 developmental biology ,Gene Expression Regulation ,Interferons ,business ,Transcriptome ,Biomarkers ,030215 immunology - Abstract
Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.
- Published
- 2019
21. A large peptidome dataset improves HLA class I epitope prediction across most of the human population
- Author
-
Letitia Li, Thomas Eisenhaure, Derin B. Keskin, William J. Lane, Hasmik Keshishian, Nir Hacohen, Tamara Ouspenskaia, Christina R. Hartigan, Sune Justesen, Phuong M. Le, Steven A. Carr, Ioannis K. Zervantonakis, Karl R. Clauser, Pavan Bachireddy, David A. Braun, Keith L. Ligon, Jennifer M. Rosenbluth, Wandi Zhang, Catherine J. Wu, Travis Law, Guang Lan Zhang, Giacomo Oliveira, Jonathan Stevens, Susan Klaeger, and Siranush Sarkizova
- Subjects
Proteasome Endopeptidase Complex ,Proteome ,Population ,Amino Acid Motifs ,Biomedical Engineering ,Bioengineering ,Endogeny ,Peptide ,Human leukocyte antigen ,Computational biology ,Biology ,Ligands ,Applied Microbiology and Biotechnology ,Epitope ,Article ,Cell Line ,03 medical and health sciences ,Epitopes ,0302 clinical medicine ,Humans ,Allele ,education ,Databases, Protein ,Alleles ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,education.field_of_study ,Histocompatibility Antigens Class I ,Predictive value ,chemistry ,Genetic Loci ,Molecular Medicine ,Peptides ,030217 neurology & neurosurgery ,Algorithms ,Biotechnology ,Peptide Hydrolases - Abstract
Prediction of HLA epitopes is important for the development of cancer immunotherapies and vaccines. However, current prediction algorithms have limited predictive power, in part because they were not trained on high-quality epitope datasets covering a broad range of HLA alleles. To enable prediction of endogenous HLA class I-associated peptides across a large fraction of the human population, we used mass spectrometry to profile >185,000 peptides eluted from 95 HLA-A, -B, -C and -G mono-allelic cell lines. We identified canonical peptide motifs per HLA allele, unique and shared binding submotifs across alleles and distinct motifs associated with different peptide lengths. By integrating these data with transcript abundance and peptide processing, we developed HLAthena, providing allele-and-length-specific and pan-allele-pan-length prediction models for endogenous peptide presentation. These models predicted endogenous HLA class I-associated ligands with 1.5-fold improvement in positive predictive value compared with existing tools and correctly identified >75% of HLA-bound peptides that were observed experimentally in 11 patient-derived tumor cell lines.
- Published
- 2019
22. 204 The immune cell landscape in kidneys of lupus nephritis patients
- Author
-
Nir Hacohen, Betty Diamond, Arnon Arazi, Deepak A. Rao, David J. Lieb, Matthias Kretzler, Edward P. Browne, Thomas Eisenhaure, Celine C. Berthier, and Anne Davidson
- Subjects
Cell type ,Kidney ,Myeloid ,business.industry ,Lupus nephritis ,medicine.disease ,medicine.anatomical_structure ,Immune system ,Interferon ,Monocyte differentiation ,Immunology ,Medicine ,business ,B cell ,medicine.drug - Abstract
Background Lupus nephritis is a potentially fatal autoimmune disease, whose current treatment is ineffective and often toxic. In 2014, the National Institute of Health (NIH), industry and non-profit organizations joined their efforts with the AMP project, whose goal is to identify new diagnostic and therapeutic targets through a better understanding of the mechanisms by which individual cell types contribute to autoimmune tissue damage. Methods To gain insights into disease mechanisms, we analyzed kidney samples from lupus nephritis patients and healthy controls using single-cell RNA-seq. Renal biopsies from 24 LN patients and 10 pre-transplant living donors (LD) were acquired across a distributed research network using a single, uniform pipeline developed by the AMP network. In brief, biopsies were cryopreserved and shipped to a centralized processing site for tissue dissociation. A total of 3541 leukocytes and 1621 epithelial cells were sorted from LN kidney samples. 438 leukocytes and 572 epithelial cells were sorted from LD biopsies. The transcriptome of those LN single tissue-infiltrating cells were assessed using single cell RNA-seq. Results Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid, T, NK and B cells, demonstrating both pro-inflammatory and resolving responses. We found evidence of local activation of B cells correlated with an age-associated B cell signature, and of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, C×CR4 and C×3CR1, were broadly expressed, pointing to potential therapeutic targets. Gene expression of immune cells in urine and kidney was highly correlated, suggesting urine may be a surrogate for kidney biopsies. Conclusions Our results provide a first comprehensive view of the complex network of leukocytes active in lupus nephritis kidneys. Results from this Phase 1 study identified LN active cells and pathways that can be used to guide the development of novel therapies. Analyses at a bigger scale (n=200 LN) in Phase 2 will allow to correlate patterns and signatures of infiltrating cells with those of intrinsic renal cells, particularly the epithelial cells that make up 90% of renal cells and that are prone to hypoxic damage and cellular stress. It will accelerate the discovery of new therapeutic targets and identification of biomarkers to guide therapeutic decisions in LN and integrate the treatment effect. Funding Source(s): Funding was provided through grants from the National Institutes of Health (UH2-AR067676, UH2-AR067677, UH2-AR067679, UH2-AR067681, UH2-AR067685, UH2-AR067688, UH2-AR067689, UH2-AR067690, UH2-AR067691, UH2-AR067694, and UM2-AR067678).
- Published
- 2019
- Full Text
- View/download PDF
23. Multiplexed enrichment and genomic profiling of peripheral blood cells reveal subset-specific immune signatures
- Author
-
Dwayne Vickers, Miguel Reyes, Paul Hoover, Thomas Eisenhaure, Deepak A. Rao, Nir Hacohen, Paul C. Blainey, Kianna Billman, and Edward P. Browne
- Subjects
Immunology ,Microfluidics ,Lipopolysaccharide Receptors ,Computational biology ,Disease ,Biology ,Monocytes ,Transcriptome ,03 medical and health sciences ,Engineering ,0302 clinical medicine ,Immune system ,Gene expression ,medicine ,Humans ,Lupus Erythematosus, Systemic ,Lymphocytes ,RNA-Seq ,Research Articles ,030304 developmental biology ,Whole blood ,Autoimmune disease ,0303 health sciences ,Multidisciplinary ,Lupus erythematosus ,SciAdv r-articles ,Flow Cytometry ,medicine.disease ,3. Good health ,Infectious disease (medical specialty) ,030220 oncology & carcinogenesis ,Single-Cell Analysis ,Research Article - Abstract
A single microscale system integrates multiplexed subset purification and gene expression profiling., Specialized immune cell subsets are involved in autoimmune disease, cancer immunity, and infectious disease through a diverse range of functions mediated by overlapping pathways and signals. However, subset-specific responses may not be detectable in analyses of whole blood samples, and no efficient approach for profiling cell subsets at high throughput from small samples is available. We present a low-input microfluidic system for sorting immune cells into subsets and profiling their gene expression. We validate the system’s technical performance against standard subset isolation and library construction protocols and demonstrate the importance of subset-specific profiling through in vitro stimulation experiments. We show the ability of this integrated platform to identify subset-specific disease signatures by profiling four immune cell subsets in blood from patients with systemic lupus erythematosus (SLE) and matched control subjects. The platform has the potential to make multiplexed subset-specific analysis routine in many research laboratories and clinical settings.
- Published
- 2019
- Full Text
- View/download PDF
24. Methods for high-dimensional analysis of cells dissociated from cryopreserved synovial tissue
- Author
-
Joshua Hillman, Mandy J. McGeachy, Nir Hacohen, Brendan F. Boyce, David L. Boyle, V. Michael Holers, Paul J. Utz, Maria Gutierrez-Arcelus, Peter K. Gregersen, Joshua Keegan, Susan M. Goodman, Thomas Eisenhaure, Cristina Rozo, Stephen Kelly, Nida Meednu, Accelerating Medicines Partnership Ra, William H. Robinson, Deepak A. Rao, Edward P. Browne, Shuqiang Li, Kaylin Muskat, Andrew Filer, Vivian P. Bykerk, Danielle Sutherby, Laura T. Donlin, Gary S. Firestein, Chad Nusbaum, Kevin Wei, Fumitaka Mizoguchi, Kamil Slowikowski, Akiko Noma, Edd Ricker, Larry W. Moreland, Lionel B. Ivashkiv, Michael B. Brenner, Alessandra B. Pernis, Jennifer H. Anolik, David J. Lieb, Jason D. Turner, Soumya Raychaudhuri, James A. Lederer, Ellen M. Gravallese, Costantino Pitzalis, and Adam Chicoine
- Subjects
0301 basic medicine ,lcsh:Diseases of the musculoskeletal system ,Cell ,Accelerating Medicines Partnership ,Arthritis, Rheumatoid ,Rheumatoid ,2.1 Biological and endogenous factors ,Aetiology ,Accelerating Medicines Partnership RA/SLE Network ,education.field_of_study ,medicine.diagnostic_test ,Synovial Membrane ,RNA sequencing ,Cell sorting ,Flow Cytometry ,medicine.anatomical_structure ,Public Health and Health Services ,Mass cytometry ,CyTOF ,Synovial tissue ,Biotechnology ,Research Article ,Stromal cell ,Clinical Sciences ,Immunology ,Population ,Bioengineering ,Biology ,Autoimmune Disease ,Arthroplasty ,03 medical and health sciences ,Clinical Research ,Biopsy ,Genetics ,medicine ,Humans ,Rheumatoid arthritis ,education ,Fibroblast ,Cryopreservation ,Arthritis ,Inflammatory and immune system ,Lineage markers ,Human Genome ,Molecular biology ,Arthritis & Rheumatology ,High-Throughput Screening Assays ,Synovial biopsy ,030104 developmental biology ,lcsh:RC925-935 - Abstract
Background Detailed molecular analyses of cells from rheumatoid arthritis (RA) synovium hold promise in identifying cellular phenotypes that drive tissue pathology and joint damage. The Accelerating Medicines Partnership RA/SLE Network aims to deconstruct autoimmune pathology by examining cells within target tissues through multiple high-dimensional assays. Robust standardized protocols need to be developed before cellular phenotypes at a single cell level can be effectively compared across patient samples. Methods Multiple clinical sites collected cryopreserved synovial tissue fragments from arthroplasty and synovial biopsy in a 10% DMSO solution. Mechanical and enzymatic dissociation parameters were optimized for viable cell extraction and surface protein preservation for cell sorting and mass cytometry, as well as for reproducibility in RNA sequencing (RNA-seq). Cryopreserved synovial samples were collectively analyzed at a central processing site by a custom-designed and validated 35-marker mass cytometry panel. In parallel, each sample was flow sorted into fibroblast, T-cell, B-cell, and macrophage suspensions for bulk population RNA-seq and plate-based single-cell CEL-Seq2 RNA-seq. Results Upon dissociation, cryopreserved synovial tissue fragments yielded a high frequency of viable cells, comparable to samples undergoing immediate processing. Optimization of synovial tissue dissociation across six clinical collection sites with ~ 30 arthroplasty and ~ 20 biopsy samples yielded a consensus digestion protocol using 100 μg/ml of Liberase™ TL enzyme preparation. This protocol yielded immune and stromal cell lineages with preserved surface markers and minimized variability across replicate RNA-seq transcriptomes. Mass cytometry analysis of cells from cryopreserved synovium distinguished diverse fibroblast phenotypes, distinct populations of memory B cells and antibody-secreting cells, and multiple CD4+ and CD8+ T-cell activation states. Bulk RNA-seq of sorted cell populations demonstrated robust separation of synovial lymphocytes, fibroblasts, and macrophages. Single-cell RNA-seq produced transcriptomes of over 1000 genes/cell, including transcripts encoding characteristic lineage markers identified. Conclusions We have established a robust protocol to acquire viable cells from cryopreserved synovial tissue with intact transcriptomes and cell surface phenotypes. A centralized pipeline to generate multiple high-dimensional analyses of synovial tissue samples collected across a collaborative network was developed. Integrated analysis of such datasets from large patient cohorts may help define molecular heterogeneity within RA pathology and identify new therapeutic targets and biomarkers. Electronic supplementary material The online version of this article (10.1186/s13075-018-1631-y) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
25. The immune cell landscape in kidneys of lupus nephritis patients
- Author
-
David A. Hildeman, Meyeon Park, Yanyan Liu, Deepak A. Rao, Edward P. Browne, Elena Massarotti, Jill P. Buyon, Shuqiang Li, A. Helena Jonsson, Nir Hacohen, David Wofsy, Michael B. Brenner, William Apruzzese, Patti Tosta, Chad Nusbaum, Fernanda Payan-Schober, Anne Davidson, Jennifer H. Anolik, Paul Hoover, David J. Lieb, Chaim Putterman, Betty Diamond, Akiko Noma, Maria Dall'Era, Richard A. Furie, Scott Steelman, James A. Lederer, Diane L. Kamen, E. Steve Woodle, Kenneth C. Kalunian, Michelle Petri, Thomas Eisenhaure, Celine C. Berthier, Dawn E. Smilek, Danielle Sutherby, Arnon Arazi, Adam Chicoine, and Matthias Kretzler
- Subjects
Autoimmune disease ,0303 health sciences ,Kidney ,Myeloid ,business.industry ,Lupus nephritis ,medicine.disease ,3. Good health ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Monocyte differentiation ,Immunology ,Medicine ,business ,B cell ,030304 developmental biology ,030215 immunology - Abstract
Lupus nephritis is a potentially fatal autoimmune disease, whose current treatment is ineffective and often toxic. To gain insights into disease mechanisms, we analyzed kidney samples from lupus nephritis patients and healthy controls using single-cell RNA-seq. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid, T, NK and B cells, demonstrating both pro-inflammatory and resolving responses. We found evidence of local activation of B cells correlated with an age-associated B cell signature, and of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, pointing to potential therapeutic targets. Gene expression of immune cells in urine and kidney was highly correlated, suggesting urine may be a surrogate for kidney biopsies. Our results provide a first comprehensive view of the complex network of leukocytes active in lupus nephritis kidneys.
- Published
- 2018
- Full Text
- View/download PDF
26. A protocol for single-cell transcriptomics from cryopreserved renal tissue and urine for the Accelerating Medicine Partnership (AMP) RA/SLE network
- Author
-
James A. Lederer, Edward P. Browne, Deepak A. Rao, David Wofsy, Jennifer H. Anolik, Matthias Kretzler, Patti Tosta, E. Steve Woodle, Nir Hacohen, Anne Davidson, Thomas Eisenhaure, Michael B. Brenner, Yanyan Liu, Dawn E. Smilek, David A. Hildeman, Celine C. Berthier, David J. Lieb, Betty Diamond, Arnon Arazi, and Adam Chicoine
- Subjects
030203 arthritis & rheumatology ,0303 health sciences ,Kidney ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Cell ,Lupus nephritis ,medicine.disease ,Cryopreservation ,Epithelium ,3. Good health ,Flow cytometry ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Cryopreserved Tissue ,business ,030304 developmental biology - Abstract
OBJECTIVEThere is a critical need to define the cells that mediate tissue damage in lupus nephritis. Here we aimed to establish a protocol to preserve lupus nephritis kidney biopsies and urine cell samples obtained at multiple clinical sites for subsequent isolation and transcriptomic analysis of single cells.METHODSFresh and cryopreserved kidney tissue from tumor nephrectomies and lupus nephritis kidney biopsies were disaggregated by enzymatic digestion. Cell yields and cell composition were assessed by flow cytometry. Transcriptomes of leukocytes and epithelial cells were evaluated by low-input and single cell RNA-seq.RESULTSCryopreserved kidney tissue from tumor nephrectomies and lupus nephritis biopsies can be thawed and dissociated to yield intact, viable leukocytes and epithelial cells. Cryopreservation of intact kidney tissue provides higher epithelial cell yields compared to cryopreservation of single cell suspensions from dissociated kidneys. Cell yields and flow cytometric cell phenotypes are comparable between cryopreserved kidney samples and paired kidney samples shipped overnight on wet ice. High-quality single cell and low-input transcriptomic data were generated from leukocytes from both cryopreserved lupus nephritis kidney biopsies and urine, as well as from a subset of kidney epithelial cells.CONCLUSIONThe AMP RA/SLE cryopreserved tissue analysis pipeline provides a method for centralized processing of lupus nephritis kidney biopsies and urine samples to generate robust transcriptomic analyses in multi-center studies.
- Published
- 2018
- Full Text
- View/download PDF
27. Multiplexed enrichment and genomic profiling of peripheral immune cell subsets on a microfluidic chip
- Author
-
Miguel Reyes, Dwayne Vickers, Kianna Billman, Edward P. Browne, Nir Hacohen, Paul Hoover, Thomas Eisenhaure, Deepak A. Rao, and Paul C. Blainey
- Subjects
Autoimmune disease ,0303 health sciences ,Systemic lupus erythematosus ,Disease ,Computational biology ,Biology ,medicine.disease ,3. Good health ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Infectious disease (medical specialty) ,Interferon ,030220 oncology & carcinogenesis ,medicine ,Ex vivo ,030304 developmental biology ,medicine.drug - Abstract
The human immune system consists of many specialized cell subsets that simultaneously carry out a diverse range of functions using overlapping pathways and signals. Subset-specific immune profiling can resolve immune activity in autoimmune disease, cancer immunity, and infectious disease that may not be discoverable or detectable in analyses of crude blood samples. The activity of specific subsets may help predict the course of disease and response to therapy in certain patient populations. Here, we present a low-input microfluidic system for sorting immune cells into subsets and profiling their cellular states by gene expression analysis using full-length RNA-seq. Our system is robust and has the potential to make multiplexed subset-specific analysis routine in many research laboratories and clinical settings. We validate the device’s technical performance by benchmarking its subset enrichment and genomic profiling performance against standard protocols. We make the added value of subset-resolved profiling over crude samples clear throughex vivoexperiments that show subset-specific stimulated responses. Finally, we demonstrate the scalability of our device by profiling four immune cell subsets in blood from systemic lupus erythematosus (SLE) patients and matched controls enrolled in a clinical study. The results from our initial cohort confirm the role of type I interferons in lupus pathogenesis and further show that the canonical interferon signature for SLE is prominent in B cells, demonstrating the ability of our integrated analytical platform to identify cell-specific disease signatures.
- Published
- 2018
- Full Text
- View/download PDF
28. 264 Pearl: pathway exploration and analysis in renal disease in the accelerating medicine partnership (amp) lupus network
- Author
-
Edward P. Browne, Nir Hacohen, Thomas Eisenhaure, Arnon Arazi, Celine C. Berthier, David J. Lieb, Betty Diamond, Da Rao, and Matthias Kretzler
- Subjects
Kidney ,Pathology ,medicine.medical_specialty ,Systemic lupus erythematosus ,business.industry ,medicine.medical_treatment ,Lupus nephritis ,medicine.disease ,Nephrectomy ,End stage renal disease ,Transcriptome ,medicine.anatomical_structure ,Interferon ,medicine ,Cytotoxic T cell ,business ,medicine.drug - Abstract
Background and aims Despite treatments, a substantial proportion of lupus nephritis (LN) patients progress to end stage renal disease and death. Detailed transcriptomic analyses of LN kidneys may identify new therapeutic targets. Our goal is to demonstrate the feasibility of single cell and low-input transcriptomic analyses of LN kidney and urine cells. Methods Cells from urine and renal biopsies performed for clinical diagnosis from inform-consented patients (1 class III, 3 class IV+V, 1 class V) and 1 control (healthy part of tumour nephrectomy) were isolated, frozen, sorted and analysed by RNAseq. Results Bulk flow sorted cell populations (CD45, epithelial) from kidney samples can separate LN from controls based on gene expression. IFN stimulated genes were differentially expressed in renal CD45 LN cells. Analysis of single cells sorted from 4 LN kidney biopsies revealed major differences in infiltrates composition, with 2 samples demonstrating a high percentage of B cells (average of 18% compared to no B cells in the other 2 samples) and CD4 T cells (18% vs 8%), and low percentage of CD8 T cells (9% vs 23%). A high transcriptomic lupus interferon signature was detected in urine CD45 cells. Distinct infiltrates and distinct expression profiles were detected across patients. Conclusions The PEARL-Phase 0 project shows the feasibility of single cell isolation and transcriptomic analysis from LN kidney and urine. Analyses at a bigger scale in the two next phases of the project will accelerate discovery of new therapeutic targets and identification of biomarkers to guide therapeutic decisions in lupus nephritis and integrate the treatment effect.
- Published
- 2017
- Full Text
- View/download PDF
29. 1830. Single-cell Transcriptional Profiling Reveals an Immune Cell State Signature of Bacterial Sepsis
- Author
-
Paul C. Blainey, Bruce D. Levy, Michael R. Filbin, Kianna Billman, Marcia B. Goldberg, Roby P. Bhattacharyya, Miguel Reyes, Nir Hacohen, Rebecca M. Baron, Thomas Eisenhaure, and Deborah T. Hung
- Subjects
medicine.diagnostic_test ,business.industry ,Cell ,Genomics ,medicine.disease ,Peripheral blood mononuclear cell ,Cell biology ,Flow cytometry ,Bacterial sepsis ,Sepsis ,Abstracts ,Infectious Diseases ,medicine.anatomical_structure ,Immune system ,Oral Abstracts ,Oncology ,Gene expression ,Medicine ,business - Abstract
Background Despite intense efforts to understand the immunopathology of sepsis, no clinically reliable diagnostic biomarkers exist. Multiple whole-blood gene expression studies have sought sepsis-associated molecular signatures, but these have not yet resolved immune phenomena at the cellular level. Using single-cell RNA sequencing (scRNA-Seq) to profile peripheral blood mononuclear cells (PBMCs), we identified a novel cellular state enriched in patients with sepsis. Methods We performed scRNA-Seq on PBMCs from 26 patients with sepsis and 47 controls at two hospitals (mean age 57.5 years, SD 16.6; 54% male; 82% white), analyzing >200,000 single cells in total on a 10× Genomics platform. We identified immune cell states by stepwise clustering, first to identify the major immune cell types, then clustering each cell type into substates. Substate abundances were compared between cases and controls using the Wilcoxon rank-sum test. Results We identified 18 immune cell substates (Figure 1a), including a novel CD14+ monocyte substate (MS1) that is enriched in patients with sepsis (Figure 1b). The fractional abundance of the MS1 substate alone (ROC AUC 0.88) outperformed published bulk transcriptional signatures in identifying sepsis (AUC 0.68–0.82) across our clinical cohorts. Deconvolution of publicly available bulk transcriptional data to infer the abundance of the MS1 substate externally validated its accuracy in predicting sepsis of various etiologies across diverse geographic locations (Figure 1c), matching the best previously identified bulk signatures. Flow cytometry using cell surface markers unique to MS1 confirmed its marked expansion in sepsis, facilitating quantitation and isolation of this substate for further study. Conclusion This study demonstrates the utility of scRNA-Seq in discovering disease-associated cytologic signatures in blood and identifies a cell state signature for sepsis in patients with bacterial infections. This novel monocyte substate matched the performance of the best bulk transcriptional signatures in classifying patients as septic, and pointed to a specific cell state for further molecular and functional characterization of sepsis immunopathogenesis. Disclosures All Authors: No reported Disclosures.
- Published
- 2019
- Full Text
- View/download PDF
30. Publisher Correction: The immune cell landscape in kidneys of patients with lupus nephritis
- Author
-
David J. Lieb, William F. Pendergraft, Deepak A. Rao, A. Helena Jonsson, Thomas Eisenhaure, Arnon Arazi, Celine C. Berthier, Michelle Petri, E. Steve Woodle, Kamil Slowikowski, Jennifer H. Anolik, Nir Hacohen, Fan Zhang, Chaim Putterman, Elizabeth A. McInnis, Fernanda Payan-Schober, Diane L. Kamen, Soumya Raychaudhuri, Akiko Noma, Adam Chicoine, Michael B. Brenner, David A. Hildeman, Elena Massarotti, Jill P. Buyon, Dawn E. Smilek, Betty Diamond, Maria Dall'Era, Anne Davidson, William Apruzzese, Kenneth C. Kalunian, Scott Steelman, Patti Tosta, Danielle Sutherby, Paul Hoover, Yanyan Liu, James A. Lederer, Matthias Kretzler, Meyeon Park, Edward P. Browne, Chad Nusbaum, Shuqiang Li, Richard Furie, and David Wofsy
- Subjects
Immune system ,medicine.anatomical_structure ,business.industry ,Immunology ,Cell ,Lupus nephritis ,Immunology and Allergy ,Medicine ,business ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
31. Extranuclear DNA accumulates in aged cells and contributes to senescence and inflammation
- Author
-
Nir Hacohen, James M. Heather, Raktima Raychowdhury, Christopher Garris, Thomas Eisenhaure, David J. Lieb, and Yuk Yuen Lan
- Subjects
0301 basic medicine ,Premature aging ,Senescence ,Aging ,Inflammation ,Biology ,Systemic inflammation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Extrachromosomal DNA ,extranuclear DNA ,medicine ,Animals ,Humans ,cellular senescence ,Cells, Cultured ,Cell Nucleus ,Mice, Knockout ,Original Paper ,Endodeoxyribonucleases ,Innate immune system ,premature aging ,DNA ,Cell Biology ,Original Papers ,STING pathway ,Cell biology ,Nuclear DNA ,030104 developmental biology ,Dnase2a ,medicine.symptom ,Cell aging ,030217 neurology & neurosurgery - Abstract
Systemic inflammation is central to aging‐related conditions. However, the intrinsic factors that induce inflammation are not well understood. We previously identified a cell‐autonomous pathway through which damaged nuclear DNA is trafficked to the cytosol where it activates innate cytosolic DNA sensors that trigger inflammation. These results led us to hypothesize that DNA released after cumulative damage contributes to persistent inflammation in aging cells through a similar mechanism. Consistent with this notion, we found that older cells harbored higher levels of extranuclear DNA compared to younger cells. Extranuclear DNA was exported by a leptomycin B‐sensitive process, degraded through the autophagosome–lysosomal pathway and triggered innate immune responses through the DNA‐sensing cGAS‐STING pathway. Patient cells from the aging diseases ataxia and progeria also displayed extranuclear DNA accumulation, increased pIRF3 and pTBK1, and STING‐dependent p16 expression. Removing extranuclear DNA in old cells using DNASE2A reduced innate immune responses and senescence‐associated (SA) β‐gal enzyme activity. Cells and tissues of Dnase2a− / − mice with defective DNA degradation exhibited slower growth, higher activity of β‐gal, or increased expression of HP‐1β and p16 proteins, while Dnase2a− / −;Sting− / − cells and tissues were rescued from these phenotypes, supporting a role for extranuclear DNA in senescence. We hypothesize a direct role for excess DNA in aging‐related inflammation and in replicative senescence, and propose DNA degradation as a therapeutic approach to remove intrinsic DNA and revert inflammation associated with aging.
- Published
- 2019
- Full Text
- View/download PDF
32. Erratum: Corrigendum: An immunogenic personal neoantigen vaccine for patients with melanoma
- Author
-
Lauren Peter, Evisa Gjini, Jerome Ritz, Indu Javeri, Wandi Zhang, William J. Lane, Zhuting Hu, Kai W. Wucherpfennig, David J. Bozym, Jon C. Aster, Anita Giobbie-Hurder, Shuqiang Li, Donna Neuberg, Nir Hacohen, Charles H. Yoon, Todd A. Carter, Kaliappanadar Nellaiappan, Heather Daley, Jonathan Stevens, Michael S. Seaman, Gad Getz, Jing Sun, David J. Lieb, Thomas Eisenhaure, Elizabeth I. Buchbinder, Derin B. Keskin, Edward F. Fritsch, Stacey Gabriel, Niall Lennon, Andres M. Salazar, Dan H. Barouch, Christina Chen, Adrienne M. Luoma, Oriol Olive, Eric S. Lander, Patrick A. Ott, Maegan Harden, Sachet A. Shukla, Scott J. Rodig, and Catherine J. Wu
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,Multidisciplinary ,Computer science ,Published Erratum ,Section (typography) ,MEDLINE ,Library science ,Data availability - Abstract
Nature 547, 217–221 (2017); doi:10.1038/nature22991 In this Letter, the ‘Data availability’ section in the Methods should state ‘WES and RNA-seq data are deposited in dbGaP (https://www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs001451.v1.p1). All other data are available from the corresponding author upon reasonable request.
- Published
- 2018
- Full Text
- View/download PDF
33. A Genome-wide CRISPR Screen in Primary Immune Cells to Dissect Regulatory Networks
- Author
-
Philipp Mertins, Randall Jeffrey Platt, Oren Parnas, Neville E. Sanjana, Nir Hacohen, Ophir Shalem, Steven A. Carr, Raktima Raychowdhury, Atray Dixit, Marko Jovanovic, Dariusz Przybylski, Rahul Satija, Chun Jimmie Ye, Thomas Eisenhaure, Feng Zhang, Rebecca H. Herbst, Aviv Regev, Itay Tirosh, Institute for Medical Engineering and Science, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Dixit, Atray C., Platt, Randall Jeffrey, Sanjana, Neville E, Shalem, Ophir, and Zhang, Feng
- Subjects
Candidate gene ,Cell Survival ,Cellular differentiation ,1.1 Normal biological development and functioning ,Gene regulatory network ,Mice, Transgenic ,Bone Marrow Cells ,Computational biology ,Biology ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transgenic ,Vaccine Related ,Gene Knockout Techniques ,Mice ,Immune system ,Underpinning research ,Biodefense ,Genetics ,CRISPR ,2.1 Biological and endogenous factors ,Animals ,Innate ,Gene Regulatory Networks ,Aetiology ,Gene ,Innate immune system ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Necrosis Factor-alpha ,Prevention ,Inflammatory and immune system ,Oligosaccharyltransferase ,Human Genome ,Immunity ,Membrane Proteins ,Cell Differentiation ,Dendritic Cells ,Biological Sciences ,Immunity, Innate ,3. Good health ,Toll-Like Receptor 4 ,Infectious Diseases ,Genetic Techniques ,Hexosyltransferases ,CRISPR-Cas Systems ,Biotechnology ,Developmental Biology - Abstract
Finding the components of cellular circuits and determining their functions systematically remains a major challenge in mammalian cells. Here, we introduced genome-wide pooled CRISPR-Cas9 libraries into dendritic cells (DCs) to identify genes that control the induction of tumor necrosis factor (Tnf) by bacterial lipopolysaccharide (LPS), a key process in the host response to pathogens, mediated by the Tlr4 pathway. We found many of the known regulators of Tlr4 signaling, as well as dozens of previously unknown candidates that we validated. By measuring protein markers and mRNA profiles in DCs that are deficient in known or candidate genes, we classified the genes into three functional modules with distinct effects on the canonical responses to LPS and highlighted functions for the PAF complex and oligosaccharyltransferase (OST) complex. Our findings uncover new facets of innate immune circuits in primary cells and provide a genetic approach for dissection of mammalian cell circuits., Broad Institute, National Institutes of Health (U.S.) (NIMH: 5DP1-MH100706), National Institutes of Health (U.S.) (NIDDK: 5R01-DK097768), National Science Foundation (U.S.) (Waterman Award), W. M. Keck Foundation, New York Stem Cell Foundation, Damon Runyon Cancer Research Foundation, Searle Scholars Program, Vallee Foundation, Robert Metcalfe, Massachusetts Institute of Technology. Simons Center for the Social Brain, National Human Genome Research Institute (U.S.) (K99- HG008171), National Science Foundation (U.S.). Graduate Research Fellowship Program (grant number 1122374), Human Frontier Science Program (Strasbourg, France)
- Published
- 2015
34. Dynamic profiling of the protein life cycle in response to pathogens
- Author
-
Raktima Raychowdhury, Rahul Satija, Toni Delorey, Nicolas Chevrier, David Gennert, Michael S. Rooney, Thomas Eisenhaure, Schraga Schwartz, Edwin H. Rodriguez, Aviv Regev, Alexander P. Fields, Maxwell R. Mumbach, Diana Lu, Jonathan S. Weissman, Nir Hacohen, Philipp Mertins, Marko Jovanovic, Dariusz Przybylski, Michal Rabani, and Steve Carr
- Subjects
Transcription, Genetic ,Proteolysis ,Quantitative Trait Loci ,Cell ,Bone Marrow Cells ,Molecular Dynamics Simulation ,Biology ,Article ,Retinoblastoma-like protein 1 ,Gene expression ,Protein biosynthesis ,medicine ,Animals ,Humans ,RNA, Messenger ,Messenger RNA ,Multidisciplinary ,medicine.diagnostic_test ,Genetic Variation ,RNA ,Dendritic Cells ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Protein Biosynthesis ,Host-Pathogen Interactions ,Proteome ,sense organs - Abstract
How the immune system readies for battle Although gene expression is tightly controlled at both the RNA and protein levels, the quantitative contribution of each step, especially during dynamic responses, remains largely unknown. Indeed, there has been much debate whether changes in RNA level contribute substantially to protein-level regulation. Jovanovic et al. built a genome-scale model of the temporal dynamics of differential protein expression during the stimulation of immunological dendritic cells (see the Perspective by Li and Biggin). Newly stimulated functions involved the up-regulation of specific RNAs and concomitant increases in the levels of the proteins they encode, whereas housekeeping functions were regulated posttranscriptionally at the protein level. Science , this issue 10.1126/science.1259038 ; see also p. 1066
- Published
- 2015
- Full Text
- View/download PDF
35. Abstract B13: Integrative genomic and transcriptomic analysis of 775 human cancer cell lines reveals tumor-derived immune programs and their regulators
- Author
-
Alexandra-Chloé Villani, Thomas Eisenhaure, Aviv Regev, Nir Hacohen, Weibo Li, Raktima Raychowdhury, and Ye Chun
- Subjects
Genetics ,Cancer Research ,Somatic cell ,medicine.medical_treatment ,Immunology ,Cancer ,Computational biology ,Immunotherapy ,Tumor-Derived ,Biology ,medicine.disease ,Transcriptome ,Immune system ,Gene expression ,medicine ,Gene - Abstract
The role of tumor cell-derived immune factors in the development of cancer and tumor immunity remains poorly understood. We hypothesized that tumors express recurrent and stable immunoregulatory programs that impact cancer growth and immunity. To identify these programs, we performed an unbiased integrative analysis of chromosomal copy number aberrations and gene expression analysis on 775 primary solid tumor cell lines, derived from 23 different tissues of origin. Using modified versions of GISTIC 2.0 and CONEXIC, a Bayesian module network method, we identified cancer drivers that correlated with specific transcriptional regulatory programs across multiple cell lines (after considering confounding variables, including histology, sub-histology, ethnicity, gender, center of collection, experimental batches and unknown variables using Surrogate Variable Analysis). We identified a total of 60 amplified and 222 deleted somatic regions, of which 52% contained a candidate driver gene whose expression was associated with the expression of a target module. The driver genes consisted of putative novel and known cancer drivers, including deletions in CDKN2A-CDKN2B (q< 4.2E-176), WWOX (q< 5.3E-100), and amplifications in MYC (q< 2.1E-81) and CCND1 (q< 1.4E-44) genes. The target modules were enriched for known cancer-dysregulated processes, including cell cycle (q< 2.1E-91) and DNA damage/repair (q< 9.3E-50), validating the analysis. We identified and focused on immune programs common to several tissues of origin, including a type I interferon-related program (q Note: This abstract was not presented at the conference. Citation Format: Alexandra-Chloe Villani, Ye Chun, Raktima Raychowdhury, Weibo Li, Thomas Eisenhaure, Aviv Regev, Nir Hacohen. Integrative genomic and transcriptomic analysis of 775 human cancer cell lines reveals tumor-derived immune programs and their regulators. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2016 Oct 20-23; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2017;5(3 Suppl):Abstract nr B13.
- Published
- 2017
- Full Text
- View/download PDF
36. Translation Rate of Human Tyrosinase Determines ItsN-Linked Glycosylation Level
- Author
-
Ruth Halaban, Daniel N. Hebert, Hadas A. Parag, Yoel Smicun, Thomas Eisenhaure, Rebecca Aron, Andrea Újvári, and Elaine Cheng
- Subjects
Glycosylation ,Tyrosinase ,Melanoma, Experimental ,Biology ,Cycloheximide ,Endoplasmic Reticulum ,Biochemistry ,Mice ,chemistry.chemical_compound ,N-linked glycosylation ,Consensus Sequence ,Protein biosynthesis ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Cells, Cultured ,Membrane Glycoproteins ,Cell-Free System ,Monophenol Monooxygenase ,Endoplasmic reticulum ,Translation (biology) ,Cell Biology ,Transport protein ,Protein Transport ,chemistry ,Protein Biosynthesis ,Melanocytes ,Protein Processing, Post-Translational - Abstract
Tyrosinase is a type I membrane glycoprotein essential for melanin synthesis. Mutations in tyrosinase lead to albinism due, at least in part, to aberrant retention of the protein in the endoplasmic reticulum and subsequent degradation by the cytosolic ubiquitin-proteasomal pathway. A similar premature degradative fate for wild type tyrosinase also occurs in amelanotic melanoma cells. To understand critical cotranslational events, the glycosylation and rate of translation of tyrosinase was studied in normal melanocytes, melanoma cells, an in vitro cell-free system, and semi-permeabilized cells. Site-directed mutagenesis revealed that all seven N-linked consensus sites are utilized in human tyrosinase. However, glycosylation at Asn-290 (Asn-Gly-Thr-Pro) was suppressed, particularly when translation proceeded rapidly, producing a protein doublet with six or seven N-linked core glycans. The inefficient glycosylation of Asn-290, due to the presence of a proximal Pro, was enhanced in melanoma cells possessing 2-3-fold faster (7.7-10.0 amino acids/s) protein translation rates compared with normal melanocytes (3.5 amino acids/s). Slowing the translation rate with the protein synthesis inhibitor cycloheximide increased the glycosylation efficiency in live cells and in the cell-free system. Therefore, the rate of protein translation can regulate the level of tyrosinase N-linked glycosylation, as well as other potential cotranslational maturation events.
- Published
- 2001
- Full Text
- View/download PDF
37. Common Genetic Variants Modulate Pathogen-Sensing Responses in Human Dendritic Cells
- Author
-
F. Ann Ran, Michelle Lee, Portia Chipendo, Soumya Raychaudhuri, Towfique Raj, Weibo Li, Barbara E. Stranger, Aviv Regev, Christophe Benoist, Kamil Slowikowski, Irene Y. Frohlich, David A. Hafler, Thomas Eisenhaure, Manolis Kellis, Nir Hacohen, Chun Ye, Alexandra-Chloé Villani, Mark Lee, Selina Imboywa, Khadir Raddassi, Cristin McCabe, Feng Zhang, Philip L. De Jager, Lucas D. Ward, Massachusetts Institute of Technology. Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Massachusetts Institute of Technology. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology. Department of Electrical Engineering and Computer Science, McGovern Institute for Brain Research at MIT, Zhang, Feng, Ran, F. Ann, Regev, Aviv, Hafler, David A., Ward, Lucas D., and Kellis, Manolis
- Subjects
Lipopolysaccharides ,Male ,Interferon Regulatory Factor-7 ,Genome-wide association study ,medicine.disease_cause ,Influenza A virus ,2.1 Biological and endogenous factors ,Aetiology ,Genetics ,Multidisciplinary ,Single Nucleotide ,Middle Aged ,STAT Transcription Factors ,Infectious Diseases ,Host-Pathogen Interactions ,Pneumonia & Influenza ,Female ,Infection ,Adult ,General Science & Technology ,Quantitative Trait Loci ,Context (language use) ,Biology ,Communicable Diseases ,Autoimmune Diseases ,Vaccine Related ,Young Adult ,Biodefense ,Genetic variation ,medicine ,Escherichia coli ,Humans ,Allele ,Polymorphism ,Gene ,Transcription factor ,Prevention ,Inflammatory and immune system ,Dendritic Cells ,Interferon-beta ,Influenza ,HEK293 Cells ,Emerging Infectious Diseases ,Genetic Loci ,IRF7 ,Gene-Environment Interaction ,Transcriptome ,Genome-Wide Association Study - Abstract
Little is known about how human genetic variation affects the responses to environmental stimuli in the context of complex diseases. Experimental and computational approaches were applied to determine the effects of genetic variation on the induction of pathogen-responsive genes in human dendritic cells. We identified 121 common genetic variants associated in cis with variation in expression responses to Escherichia coli lipopolysaccharide, influenza, or interferon-β (IFN-β). We localized and validated causal variants to binding sites of pathogen-activated STAT (signal transducer and activator of transcription) and IRF (IFN-regulatory factor) transcription factors. We also identified a common variant in IRF7 that is associated in trans with type I IFN induction in response to influenza infection. Our results reveal common alleles that explain interindividual variation in pathogen sensing and provide functional annotation for genetic variants that alter susceptibility to inflammatory diseases., National Human Genome Research Institute (U.S.) (Grant P50 HG006193), National Institutes of Health (U.S.). Pioneer Award (DP1 CA174427), Howard Hughes Medical Institute, National Institutes of Health (U.S.) (Grant HG004037), National Institutes of Health (U.S.). Pioneer Award (DP1 MH100706), National Institutes of Health (U.S.) (Transformative R01 Grant R01 DK097768), W. M. Keck Foundation, McKnight Foundation, Merkin, Richard N., Damon Runyon Cancer Research Foundation, Searle Scholars Program, Simons Foundation
- Published
- 2013
38. Systematic Discovery of TLR Signaling Components Delineates Viral-Sensing Circuits
- Author
-
Nir Yosef, Mark F. Ciaccio, Thomas Eisenhaure, Marciela M. DeGrace, Philipp Mertins, Richard Bradley Jones, Matteo Iannacone, Elena Tonti, Irit Gat-Viks, Hongkun Park, Ido Amit, Steven A. Carr, Jacob T. Robinson, Manuel Garber, Mette S. Andersen, Alex K. Shalek, Nicolas Chevrier, David E. Root, Maxim N. Artyomov, Amy Sutton, Ulrich H. von Andrian, Karl R. Clauser, Nir Hacohen, Aviv Regev, Chevrier, N, Mertins, P, Artyomov, Mn, Shalek, Ak, Iannacone, M, Ciaccio, Mf, Gat-Viks, I, Tonti, E, Degrace, Mm, Clauser, Kr, Garber, M, Eisenhaure, Tm, Yosef, N, Robinson, J, Sutton, A, Andersen, M, Root, De, von Andrian, U, Jones, Rb, Park, H, Carr, Sa, Regev, A, Amit, I, and Hacohen, N
- Subjects
Disease ,Computational biology ,Protein Serine-Threonine Kinases ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,In vivo ,Animals ,Humans ,Receptor ,Gene ,030304 developmental biology ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Kinase ,Toll-Like Receptors ,ComputingMilieux_PERSONALCOMPUTING ,Phosphoproteomics ,Dendritic Cells ,Cell biology ,Mice, Inbred C57BL ,030220 oncology & carcinogenesis ,Viruses ,Female ,Interferon Regulatory Factor-3 ,Interferons ,Signal transduction ,Signal Transduction - Abstract
SUMMARY Deciphering the signaling networks that underlie normal and disease processes remains a major challenge. Here, we report the discovery of signaling components involved in the Toll-like receptor (TLR) response of immune dendritic cells (DCs), including a previously unkown pathway shared across mammalian antiviral responses. By combining transcriptional profiling, genetic and small-molecule perturbations, and phosphoproteomics, we uncover 35 signaling regulators, including 16 known regulators, involved in TLR signaling. In particular, we find that Polo-like kinases (Plk) 2 and 4 are essential components of antiviral pathways in vitro and in vivo and activate a signaling branch involving a dozen proteins, among which is Tnfaip2, a gene associated with autoimmune diseases but whose role was unknown. Our study illustrates the power of combining systematic measurements and perturbations to elucidate complex signaling circuits and discover potential therapeutic targets.
- Published
- 2011
39. RNA Interference Screen in Primary Human T Cells Reveals FLT3 as a Modulator of IL-10 Levels
- Author
-
Stephen M. Anderton, David A. Hafler, Thomas Eisenhaure, Anne Astier, Nir Hacohen, Gaëlle Beriou, Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Recherche en Transplantation et Immunologie (U1064 Inserm - CRTI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Yale University School of Medicine, and Massachusetts General Hospital [Boston]
- Subjects
T-Lymphocytes ,T cell ,Immunology ,Biology ,Ligands ,Lymphocyte Activation ,Article ,Membrane Cofactor Protein ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,medicine ,Humans ,Immunologic Factors ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,RNA, Small Interfering ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,Gene Library ,030304 developmental biology ,Interleukin 3 ,Feedback, Physiological ,0303 health sciences ,ZAP70 ,Lentivirus ,CD28 ,Molecular biology ,Interleukin-10 ,medicine.anatomical_structure ,Gene Expression Regulation ,fms-Like Tyrosine Kinase 3 ,Interleukin 12 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,RNA Interference ,030215 immunology - Abstract
Functional studies of human primary immune cells have been hampered by the lack of tools to silence gene functions. In this study, we report the application of a lentiviral RNA interference library in primary human T cells. Using a subgenomic short hair RNA library targeting ∼1000 signaling genes, we identified novel genes that control the levels of IL-10 produced. IL-10 is a potent anti-inflammatory cytokine secreted by several cell types, including T regulatory type 1 cells, a subset of T regulatory cells that exert their suppressive activity through IL-10 secretion. FLT3, a known hematopoeitic growth factor, was found to be a negative regulator of IL-10 levels in activated T cells. This was based on several observations. First, FLT3 and its ligand (FL) were both induced by T cell activation. Second, silencing of FLT3 led to increased IL-10 levels, whereas addition of FL suppressed IL-10 secretion and increased FLT3 surface levels. Third, engagement of CD46, a known inducer of T regulatory type 1 cells, upregulated surface FLT3, and secreted FL, which then inhibited IL-10 production by T cells. Hence, FL and FLT3 form a novel regulatory feedback loop that limits IL-10 production in T cells. Our results identified FLT3 as a new regulator of T cell function and offer a strategy to genetically dissect specific pathways in T cells.
- Published
- 2010
- Full Text
- View/download PDF
40. Unbiased Reconstruction of a Mammalian Transcriptional Network Mediating Pathogen Responses
- Author
-
Alexander Meissner, Ana Paula Leite, Lisa A. Schubert, Yoni Donner, Alon Goren, Moran N. Cabili, Rebecca C. McDonald, Brian Birditt, Jennifer K. Grenier, Aviv Regev, Thomas Eisenhaure, Nicolas Chevrier, Mitchell Guttman, Weibo Li, Manuel Garber, Zachary D. Smith, Tal Shay, Nir Hacohen, John L. Rinn, David E. Root, Or Zuk, Bradley E. Bernstein, Xiaolan Zhang, Ido Amit, Raquel P. Deering, Massachusetts Institute of Technology. Computational and Systems Biology Program, Massachusetts Institute of Technology. Department of Biology, Amit, Ido, Guttman, Mitchell, Leite, Ana Paula, and Regev, Aviv
- Subjects
Lipopolysaccharides ,Transcription, Genetic ,Gene regulatory network ,DNA, Single-Stranded ,RNA-binding protein ,Computational biology ,Biology ,Article ,03 medical and health sciences ,Lipopeptides ,Mice ,0302 clinical medicine ,Transcription (biology) ,Gene expression ,Animals ,Gene Regulatory Networks ,Transcription factor ,030304 developmental biology ,Regulation of gene expression ,Genetics ,Feedback, Physiological ,Inflammation ,0303 health sciences ,Multidisciplinary ,Bacteria ,Gene Expression Profiling ,Toll-Like Receptors ,RNA-Binding Proteins ,Dendritic Cells ,Chromatin Assembly and Disassembly ,Chromatin ,Gene expression profiling ,Mice, Inbred C57BL ,Poly I-C ,Gene Expression Regulation ,Viruses ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Models of mammalian regulatory networks controlling gene expression have been inferred from genomic data but have largely not been validated. We present an unbiased strategy to systematically perturb candidate regulators and monitor cellular transcriptional responses. We applied this approach to derive regulatory networks that control the transcriptional response of mouse primary dendritic cells to pathogens. Our approach revealed the regulatory functions of 125 transcription factors, chromatin modifiers, and RNA binding proteins, which enabled the construction of a network model consisting of 24 core regulators and 76 fine-tuners that help to explain how pathogen-sensing pathways achieve specificity. This study establishes a broadly applicable, comprehensive, and unbiased approach to reveal the wiring and functions of a regulatory network controlling a major transcriptional response in primary mammalian cells., Burroughs Wellcome Fund (Career Award at the Scientific Interface), National Institutes of Health (U.S.) (Pioneer Award), Alfred P. Sloan Foundation
- Published
- 2009
41. The Rho guanine nucleotide exchange factor Lsc homo-oligomerizes and is negatively regulated through domains in its carboxyl terminus that are absent in novel splenic isoforms
- Author
-
Shaun R. Coughlin, Daniel J. Lerner, L David Willison, Thomas Eisenhaure, and Sanjeev A. Francis
- Subjects
Gene isoform ,rho GTP-Binding Proteins ,Molecular Sequence Data ,Biology ,Biochemistry ,Mice ,Isomerism ,Transcription (biology) ,In vivo ,Proto-Oncogene Proteins ,Two-Hybrid System Techniques ,Animals ,Guanine Nucleotide Exchange Factors ,Amino Acid Sequence ,Molecular Biology ,Mammals ,C-terminus ,Cell Biology ,3T3 Cells ,Yeast ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Regulatory sequence ,Mutagenesis ,COS Cells ,sense organs ,Guanine nucleotide exchange factor ,Signal transduction ,Rho Guanine Nucleotide Exchange Factors ,Spleen - Abstract
Rho GTPases control fundamental cellular processes, including cytoskeletal reorganization and transcription. Rho guanine nucleotide exchange factors (GEFs) compose a large (>65) and diverse family of related proteins that activate Rho GTPases. Lsc/p115-RhoGEF is a Rho-specific GEF required for normal B and T lymphocyte function. Despite its essential role in lymphocytes, Lsc/p115-RhoGEF signaling in vivo is not well understood. To define Lsc/p115-RhoGEF signaling pathways in vivo, we set out to identify proteins that interact with regulatory regions of Lsc. The 146-amino acid C terminus of Lsc contains a predicted coiled-coil domain, and we demonstrated that deletion of this C terminus confers a gain of function in vivo. Surprisingly, a yeast two-hybrid screen for proteins that interact with this regulatory C terminus isolated a larger C-terminal fragment of Lsc itself. Co-immunoprecipitation experiments in mammalian cells demonstrated that Lsc specifically homo-oligomerizes and that the coiled-coil domain in the C terminus is required for homo-oligomerization. Mutagenesis experiments revealed that homo-oligomerization and negative regulation are distinct functions of the C terminus. Two novel isoforms of Lsc found in the spleen lack portions of this C terminus, including the coiled-coil domain. Importantly, the C termini of both isoforms confer a gain of function and eliminate homo-oligomerization. These results define two important features of Lsc signaling. First, Lsc homo-oligomerizes and is negatively regulated through domains in its C terminus; and second, functionally distinct isoforms of Lsc lacking these domains are present in the spleen.
- Published
- 2003
42. Flt3, a New Regulator of IL-10 Production by Human T Cells, Identified Using an RNAi Library
- Author
-
David A. Hafler, Anne Astier, Thomas Eisenhaure, Gaelle Beriou, and Nir Hacohen
- Subjects
Interleukin 10 ,RNA interference ,Immunology ,Regulator ,Immunology and Allergy ,Biology ,Cell biology - Published
- 2008
- Full Text
- View/download PDF
43. OR.80. Identifying Regulators of Cytokine Production in Primary Human T-Cells Using a Lentiviral RNAi Library
- Author
-
Gaelle Beriou, David A. Hafler, Nir Hacohen, Anne Astier, and Thomas Eisenhaure
- Subjects
Cytokine ,Primary (chemistry) ,RNA interference ,medicine.medical_treatment ,Immunology ,medicine ,Immunology and Allergy ,Biology ,Virology ,Cell biology - Published
- 2006
- Full Text
- View/download PDF
44. A Lentiviral RNAi Library for Human and Mouse Genes Applied to an Arrayed Viral High-Content Screen
- Author
-
Xiaoping Yang, Angela M. Kloepfer, Sheila A. Stewart, Jason Moffat, Dorre A. Grueneberg, Anne E. Carpenter, Gregory Hinkle, Thomas Eisenhaure, Bruno Piqani, David M. Sabatini, So Young Kim, David E. Root, Nir Hacohen, Jennifer K. Grenier, Brent R. Stockwell, Eric S. Lander, Biao Luo, William C. Hahn, and Shi Yin Foo
- Subjects
Genetic Vectors ,Libraries ,Cell Cycle Proteins ,Computational biology ,Biology ,Genome ,General Biochemistry, Genetics and Molecular Biology ,Small hairpin RNA ,Mice ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,Animals ,Humans ,Genomic library ,RNA, Small Interfering ,Gene ,Cells, Cultured ,Gene Library ,030304 developmental biology ,Genetics ,0303 health sciences ,Microarray analysis techniques ,Biochemistry, Genetics and Molecular Biology(all) ,Lentivirus ,RNA ,Microarray Analysis ,030220 oncology & carcinogenesis ,Human genome ,Genetic Engineering - Abstract
To enable arrayed or pooled loss-of-function screens in a wide range of mammalian cell types, including primary and nondividing cells, we are developing lentiviral short hairpin RNA (shRNA) libraries targeting the human and murine genomes. The libraries currently contain 104,000 vectors, targeting each of 22,000 human and mouse genes with multiple sequence-verified constructs. To test the utility of the library for arrayed screens, we developed a screen based on high-content imaging to identify genes required for mitotic progression in human cancer cells and applied it to an arrayed set of 5,000 unique shRNA-expressing lentiviruses that target 1,028 human genes. The screen identified several known and approximately 100 candidate regulators of mitotic progression and proliferation; the availability of multiple shRNAs targeting the same gene facilitated functional validation of putative hits. This work provides a widely applicable resource for loss-of-function screens, as well as a roadmap for its application to biological discovery.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.