Volume 26, Issue 3, Pages e5 (September 2019)

Slides:



Advertisements
Similar presentations
Figure 1. AsCpf1 and LbCpf1-mediated gene editing in human cells
Advertisements

CRISPR + CAS = Defensive or Immune System
Alternative Computational Analysis Shows No Evidence for Nucleosome Enrichment at Repetitive Sequences in Mammalian Spermatozoa  Hélène Royo, Michael Beda.
CRISPR-Cas Systems: Prokaryotes Upgrade to Adaptive Immunity
Volume 56, Issue 4, Pages (November 2014)
Volume 62, Issue 2, Pages (April 2016)
Archaeal CRISPR-based immune systems: exchangeable functional modules
Volume 44, Issue 3, Pages (November 2011)
Discovery and Characterization of piRNAs in the Human Fetal Ovary
Volume 146, Issue 6, Pages (September 2011)
Harnessing CRISPR–Cas systems for bacterial genome editing
Daan C. Swarts, John van der Oost, Martin Jinek  Molecular Cell 
Hyeshik Chang, Jaechul Lim, Minju Ha, V. Narry Kim  Molecular Cell 
CRISPR-Cas Systems: Prokaryotes Upgrade to Adaptive Immunity
Addison V. Wright, James K. Nuñez, Jennifer A. Doudna  Cell 
Volume 62, Issue 1, Pages (April 2016)
A Massively Parallel Reporter Assay of 3′ UTR Sequences Identifies In Vivo Rules for mRNA Degradation  Michal Rabani, Lindsey Pieper, Guo-Liang Chew,
Alterations in the Gut Microbiota Associated with HIV-1 Infection
CRISPR Interference Can Prevent Natural Transformation and Virulence Acquisition during In Vivo Bacterial Infection  David Bikard, Asma Hatoum-Aslan,
Mouse Genome Engineering via CRISPR-Cas9 for Study of Immune Function
Volume 65, Issue 1, Pages (January 2017)
Figure 1 Adaptive immune system of bacteria and archaea
Target-Specific Precision of CRISPR-Mediated Genome Editing
Volume 50, Issue 4, Pages (May 2013)
Single-Stranded DNA Cleavage by Divergent CRISPR-Cas9 Enzymes
Edwards Allen, Zhixin Xie, Adam M. Gustafson, James C. Carrington  Cell 
A Subset of Polysaccharide Capsules in the Human Symbiont Bacteroides thetaiotaomicron Promote Increased Competitive Fitness in the Mouse Gut  Nathan.
Volume 22, Issue 3, Pages e5 (September 2017)
Volume 18, Issue 5, Pages (November 2015)
Joseph Bondy-Denomy, Alan R. Davidson  Trends in Microbiology 
Guide RNA Functional Modules Direct Cas9 Activity and Orthogonality
Volume 22, Issue 3, Pages e3 (September 2017)
Volume 48, Issue 5, Pages (December 2012)
Volume 164, Issue 1, Pages (January 2016)
Dipali G. Sashital, Blake Wiedenheft, Jennifer A. Doudna 
Joseph Rodriguez, Jerome S. Menet, Michael Rosbash  Molecular Cell 
Adam S. Dingens, Hugh K. Haddox, Julie Overbaugh, Jesse D. Bloom 
The Evolution of Antiviral Defense Systems
Volume 70, Issue 2, Pages e5 (April 2018)
Volume 44, Issue 3, Pages (November 2011)
Volume 5, Issue 3, Pages e7 (September 2017)
Gang Wang, Na Zhao, Ben Berkhout, Atze T Das  Molecular Therapy 
Advances in Industrial Biotechnology Using CRISPR-Cas Systems
Volume 69, Issue 1, Pages e3 (January 2018)
Advances in Understanding Bacterial Pathogenesis Gained from Whole-Genome Sequencing and Phylogenetics  Elizabeth Klemm, Gordon Dougan  Cell Host & Microbe 
Baekgyu Kim, Kyowon Jeong, V. Narry Kim  Molecular Cell 
Anti-CRISPR Phages Cooperate to Overcome CRISPR-Cas Immunity
Volume 22, Issue 3, Pages e4 (September 2017)
A CRISPR Approach to Gene Targeting
Nathan Pirakitikulr, Andrew Kohlway, Brett D. Lindenbach, Anna M. Pyle 
Volume 122, Issue 6, Pages (September 2005)
Volume 18, Issue 6, Pages (December 2015)
Structure of an RNA Silencing Complex of the CRISPR-Cas Immune System
RNA Targeting by Functionally Orthogonal Type VI-A CRISPR-Cas Enzymes
Hayun Lee, Yi Zhou, David W. Taylor, Dipali G. Sashital  Molecular Cell 
DNA Looping Facilitates Targeting of a Chromatin Remodeling Enzyme
Volume 67, Issue 6, Pages e9 (September 2017)
Volume 10, Issue 2, Pages (August 2011)
Volume 20, Issue 4, Pages (October 2016)
CdpR Inhibits CRISPR-Cas Adaptive Immunity to Lower Anti-viral Defense while Avoiding Self-Reactivity  Ping Lin, Qinqin Pu, Guanwang Shen, Rongpeng Li,
The Evolution of Endogenous Viral Elements
Julia K. Nussbacher, Gene W. Yeo  Molecular Cell 
Adaptation in CRISPR-Cas Systems
Position-Dependent Function for a Tandem MicroRNA miR-122-Binding Site Located in the Hepatitis C Virus RNA Genome  Catherine L. Jopling, Sylvia Schütz,
Volume 73, Issue 2, Pages e4 (January 2019)
CRISPR Immunological Memory Requires a Host Factor for Specificity
Toward Accurate and Quantitative Comparative Metagenomics
Volume 71, Issue 2, Pages e5 (July 2018)
Potent and sequence-specific removal through genome targeting with CRISPR-Cas systems. Potent and sequence-specific removal through genome targeting with.
Bacterial Argonaute Samples the Transcriptome to Identify Foreign DNA
Presentation transcript:

Volume 26, Issue 3, Pages 325-335.e5 (September 2019) CRISPR-Cas System of a Prevalent Human Gut Bacterium Reveals Hyper-targeting against Phages in a Human Virome Catalog  Paola Soto-Perez, Jordan E. Bisanz, Joel D. Berry, Kathy N. Lam, Joseph Bondy-Denomy, Peter J. Turnbaugh  Cell Host & Microbe  Volume 26, Issue 3, Pages 325-335.e5 (September 2019) DOI: 10.1016/j.chom.2019.08.008 Copyright © 2019 Elsevier Inc. Terms and Conditions

Cell Host & Microbe 2019 26, 325-335. e5DOI: (10. 1016/j. chom. 2019 Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 1 E. lenta DSM 2243 Has a Transcriptionally and Catalytically Active CRISPR-Cas System (A) Base coverage of RNA-seq reads to the CRISPR-Cas locus in DSM 2243 and indicates active transcription. (B) Expression levels of cas genes during exponential growth measured in reads per kilobase per million mapped reads (RPKM). (C) Transcription from the CRISPR array generates a pre-CRISPR RNA that is processed by the Cas enzymes to form crRNAs that direct targeting and cleavage of foreign DNA. (D) Northern blot demonstrates the presence of short RNA species (crRNA) in a CRISPR-positive strain (DSM 2243) but not in a CRISPR-negative strain (Valencia). Growth phase is indicated above the blot: M = mid-exponential (24 h) and L = late-exponential (37 h). (E and F) Growth kinetics (n = 3) (E) and cas expression levels (F) demonstrate the stability of cas3 and cas5 across growth phases (n = 3, ∗∗∗p < 0.001 two-way ANOVA). Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 2 Heterologous Expression in P. aeruginosa Demonstrates the Ability to Target Phage and Chromosomal DNA (A) P. aeruginosa strain (PA01 tn7::lentaIC) constructed to inducibly express the minimal cas genes required for interference and a plasmid containing a minimal CRISPR array. (B) Expression of gJBD30 (phage-targeting) causes a 120-fold reduction in the number of plaque-forming units (PFUs) when compared to a NT control (n = 4, ∗p = 0.0286, Mann-Whitney U test). (C) Expression of gPhzM (chromosome-targeting) decreases the colony-forming units (CFUs) by 13,450-fold (n = 5, ∗∗p = 0.0079, Mann-Whitney U test). (D) Distribution of spacer lengths found in the E. lenta isolate genomes, metagenomes, and merged datasets. (E) Variable length crRNAs decrease the number of PFUs with the exception of a 40-nt crRNA (n = 4). Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 3 Strain-Level Variation in the E. lenta Type I-C CRISPR-Cas System (A) 15 sequenced isolate genomes contain a type I-C system that clusters into two distinct clades based on an alignment of the cas genes. The global nucleotide identity to the DSM 2243 ortholog is shown. Diamonds indicate the number of direct repeats, and the exact number of spacers in each array is displayed. (B) CRISPR spacer conservation between strains. Spacers are indicated as colored diamonds with identical spacers linked by a gray line. Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 4 Direct Repeat and Spacer Conservation across Reference Genomes and Metagenomic Datasets (A) The 33-nt E. lenta direct repeat was found to be highly conserved in all 15 CRISPR-positive isolate genomes. (B) Analysis of shared spacer content between strains provides evidence of a clade-specific pattern of conservation. Spacers were numbered 1–493 ordered by frequency of occurrence. (C) Venn diagram of shared spacer content between isolate genome clades and metagenomic data. (D) Evaluation of self- (targeting of the genome by a spacer encoded within genome) and inter-strain targeting. Alignment of spacers to the E. lenta “super genome”: a 7-Mbp non-redundant sequence representing the aggregate genomes of this bacterial species. Red triangles indicate spacer matches within putative prophage and mobile elements. Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 5 Discovering E. lenta Predators Based on Protospacer Enrichment (A) Comparison of protospacer matches within HuVirDB (249/493) versus other publicly accessible databases, including isolated and sequenced plasmids and phages from RefSeq. (B) To facilitate phage discovery, public virome sequencing data were collected and assembled for our HuVirDB. (C) The 5′ flanking sequence was enriched for the canonical type I-C protospacer adjacent motif (PAM) TTC. (D) Detailed annotation of a representative phage (ELM P1), identified based on a high frequency of matching E. lenta spacers. Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 6 E. lenta Metagenomic (ELM) Phage Genomes (A) Genomes are presented with annotated genes colored by high-level function, and protospacer locations are indicated by dashes. Protospacers were allowed to have up to 4 mismatches to the spacer sequence. The number of unique (meta)genomes targeting the seed phage and the number of unique spacers are shown. (B) Clustering for taxonomic annotation of ELM phages with prokaryotic viral genomes from Viral RefSeq v.85 based on gene sharing (Bin Jang et al., 2019) demonstrates a unique clade formed by 7 ELM phages. Only non-singleton clusters are depicted. (C) A phylogenetic tree of the ELM phages based on genome-wide BLAST distances (Meier-Kolthoff and Göker, 2017). Numbers on the tree represent pseudo-bootstrap support values from 100 replications. Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions

Figure 7 Most Mismatches between the Spacer and Protospacer Sequences Still Provide Immunity against Phages (A) The occurrence of mismatches throughout the length of the spacer was calculated using HuVirDB. (B) Plaque assays revealed two phenotypes: opaque plaques (efficient targeting) and clearer plaques (poor targeting). Controls include: a perfect match positive control and a NT negative control. A representative mismatch (A31G) is shown. (C) Plaquing efficiency (log estimation in tested gRNA divided by log estimation in NT control) reveals that mutations in the seed sequence of the crRNA allow the phage to escape CRISPR-Cas immunity (n = 4). The dashed line at 10−5 denotes the average value for the perfect match control gRNA. ∗p < 0.05 Kruskal-Wallis with uncorrected Dunnett’s (compared to targeting control). Cell Host & Microbe 2019 26, 325-335.e5DOI: (10.1016/j.chom.2019.08.008) Copyright © 2019 Elsevier Inc. Terms and Conditions