Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Max A. Horlbeck is active.

Publication


Featured researches published by Max A. Horlbeck.


Cell | 2013

A Systematic Mammalian Genetic Interaction Map Reveals Pathways Underlying Ricin Susceptibility

Michael C. Bassik; Martin Kampmann; Robert Jan Lebbink; Shuyi Wang; Marco Y. Hein; Ina Poser; Jimena Weibezahn; Max A. Horlbeck; Siyuan Chen; Matthias Mann; Anthony A. Hyman; Emily LeProust; Michael T. McManus; Jonathan S. Weissman

Genetic interaction (GI) maps, comprising pairwise measures of how strongly the function of one gene depends on the presence of a second, have enabled the systematic exploration of gene function in microorganisms. Here, we present a two-stage strategy to construct high-density GI maps in mammalian cells. First, we use ultracomplex pooled shRNA libraries (25 shRNAs/gene) to identify high-confidence hit genes for a given phenotype and effective shRNAs. We then construct double-shRNA libraries from these to systematically measure GIs between hits. A GI map focused on ricin susceptibility broadly recapitulates known pathways and provides many unexpected insights. These include a noncanonical role for COPI, a previously uncharacterized protein complex affecting toxin clearance, a specialized role for the ribosomal protein RPS25, and functionally distinct mammalian TRAPP complexes. The ability to rapidly generate mammalian GI maps provides a potentially transformative tool for defining gene function and designing combination therapies based on synergistic pairs.


Science | 2017

CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells

S. John Liu; Max A. Horlbeck; Seung Woo Cho; Harjus Birk; Martina Malatesta; Daniel He; Frank J. Attenello; Jacqueline E. Villalta; Min Y. Cho; Yuwen Chen; Mohammad A. Mandegar; Michael P. Olvera; Luke A. Gilbert; Bruce R. Conklin; Howard Y. Chang; Jonathan S. Weissman; Daniel A. Lim

A very focused function for lncRNAs The human genome generates many thousands of long noncoding RNAs (lncRNAs). A very small number of lncRNAs have been shown to be functional. Liu et al. carried out a large-scale CRISPR-based screen to assess the function of ∼17,000 lncRNAs in seven different human cell lines. A considerable number (∼500) of the tested lncRNAs influenced cell growth, suggesting biological function. In almost all cases, though, the function was highly cell type—specific, often limited to just one cell type. Science, this issue p. 10.1126/science.aah7111 A considerable fraction of long noncoding RNAs have highly cell type–specific biological functions. INTRODUCTION The human genome contains tens of thousands of loci that produce long noncoding RNAs (lncRNAs), transcripts that have no apparent protein-coding potential. A subset of lncRNAs have been found to play critical roles in cellular processes, organismal development, and disease. Although these examples are suggestive of the importance and diversity of lncRNAs, the vast majority of lncRNA genes have not been functionally tested. RATIONALE Because it is currently not possible to predict which lncRNA loci are functional or what function they perform, there is a need for large-scale, systematic approaches to interrogating the functional contribution of lncRNA loci. We therefore developed a genome-scale screening platform based on CRISPR-mediated interference (CRISPRi), which uses a catalytically inactive CRISPR effector protein, (d)Cas9, fused to a repressive KRAB domain and targeted by a single guide RNA (sgRNA), to inhibit gene expression. By catalyzing repressive chromatin modifications around the transcription start site (TSS) and serving as a transcriptional roadblock, CRISPRi tests a broad range of lncRNA gene functions, including the production of cis- and trans-acting RNA transcripts, cis-mediated regulation related to lncRNA transcription itself, and enhancer-like function of some lncRNA loci. RESULTS We designed a CRISPRi Non-Coding Library (CRiNCL), which targets 16,401 lncRNA genes each with 10 sgRNAs per TSS, and applied this pooled screening approach to identify lncRNA genes that modify robust cell growth. We screened seven human cell lines, including six transformed cell lines and induced pluripotent stem cells (iPSCs), and identified 499 lncRNA loci that modified cell growth upon CRISPRi targeting; 372 and 299 of these loci were distal from any protein coding gene or mapped enhancer, respectively. Extensive validation confirmed the screen results and demonstrated the robust and specific performance of CRISPRi for repressing lncRNA transcription. Remarkably, 89% of the lncRNA gene hits modified growth in just one of the cell lines tested, and no hits were common to all seven cell lines. Although nearly all of the hit genes were expressed in the cell line in which they exhibited a growth phenotype, expression alone was insufficient to explain the cell type specificity of their function. Transcriptional profiling revealed extensive gene expression changes upon CRISPRi targeting of lncRNA loci in the cells in which they modified growth, whereas targeting the same lncRNA locus in other cell lines resulted in minimal changes to the transcriptome beyond depletion of the targeted lncRNA transcript itself. CONCLUSION Our study considerably increases the number of known functional lncRNA loci. More broadly, our CRISPRi approach enables mechanistic studies of specific lncRNA functions and, when applied systematically, supports the global exploration of the complex biology contained in the lncRNA-expressing genome. Finally, in contrast to recent studies that found that essential protein-coding genes typically are required across a broad range of cell types, we show that lncRNA function is highly cell type–specific, a finding that has important implications for their involvement in both normal biology and disease. CRISPRi screening of lncRNAs in human cells. CRISPRi can precisely repress transcription of lncRNAs. The CRISPRi Non-Coding Library (CRiNCL) was generated to interrogate the function of thousands of long noncoding RNAs in seven different cell lines. Validation studies confirmed the exquisite cell type–specific function of lncRNAs. The human genome produces thousands of long noncoding RNAs (lncRNAs)—transcripts >200 nucleotides long that do not encode proteins. Although critical roles in normal biology and disease have been revealed for a subset of lncRNAs, the function of the vast majority remains untested. We developed a CRISPR interference (CRISPRi) platform targeting 16,401 lncRNA loci in seven diverse cell lines, including six transformed cell lines and human induced pluripotent stem cells (iPSCs). Large-scale screening identified 499 lncRNA loci required for robust cellular growth, of which 89% showed growth-modifying function exclusively in one cell type. We further found that lncRNA knockdown can perturb complex transcriptional networks in a cell type–specific manner. These data underscore the functional importance and cell type specificity of many lncRNAs.


Genome Biology | 2016

Single-cell analysis of long non-coding RNAs in the developing human neocortex.

Siyuan John Liu; Tomasz J. Nowakowski; Alex A. Pollen; Jan H. Lui; Max A. Horlbeck; Frank J. Attenello; Daniel He; Jonathan S. Weissman; Arnold R. Kriegstein; Aaron Diaz; Daniel A. Lim

BackgroundLong non-coding RNAs (lncRNAs) comprise a diverse class of transcripts that can regulate molecular and cellular processes in brain development and disease. LncRNAs exhibit cell type- and tissue-specific expression, but little is known about the expression and function of lncRNAs in the developing human brain. Furthermore, it has been unclear whether lncRNAs are highly expressed in subsets of cells within tissues, despite appearing lowly expressed in bulk populations.ResultsWe use strand-specific RNA-seq to deeply profile lncRNAs from polyadenylated and total RNA obtained from human neocortex at different stages of development, and we apply this reference to analyze the transcriptomes of single cells. While lncRNAs are generally detected at low levels in bulk tissues, single-cell transcriptomics of hundreds of neocortex cells reveal that many lncRNAs are abundantly expressed in individual cells and are cell type-specific. Notably, LOC646329 is a lncRNA enriched in single radial glia cells but is detected at low abundance in tissues. CRISPRi knockdown of LOC646329 indicates that this lncRNA regulates cell proliferation.ConclusionThe discrete and abundant expression of lncRNAs among individual cells has important implications for both their biological function and utility for distinguishing neural cell types.


eLife | 2016

Nucleosomes impede Cas9 access to DNA in vivo and in vitro

Max A. Horlbeck; Lea B. Witkowsky; Benjamin Guglielmi; Joseph M Replogle; Luke A. Gilbert; Jacqueline E. Villalta; Sharon E Torigoe; Robert Tijan; Jonathan S. Weissman

The prokaryotic CRISPR (clustered regularly interspaced palindromic repeats)-associated protein, Cas9, has been widely adopted as a tool for editing, imaging, and regulating eukaryotic genomes. However, our understanding of how to select single-guide RNAs (sgRNAs) that mediate efficient Cas9 activity is incomplete, as we lack insight into how chromatin impacts Cas9 targeting. To address this gap, we analyzed large-scale genetic screens performed in human cell lines using either nuclease-active or nuclease-dead Cas9 (dCas9). We observed that highly active sgRNAs for Cas9 and dCas9 were found almost exclusively in regions of low nucleosome occupancy. In vitro experiments demonstrated that nucleosomes in fact directly impede Cas9 binding and cleavage, while chromatin remodeling can restore Cas9 access. Our results reveal a critical role of eukaryotic chromatin in dictating the targeting specificity of this transplanted bacterial enzyme, and provide rules for selecting Cas9 target sites distinct from and complementary to those based on sequence properties. DOI: http://dx.doi.org/10.7554/eLife.12677.001


Cell Stem Cell | 2016

CRISPR Interference Efficiently Induces Specific and Reversible Gene Silencing in Human iPSCs

Mohammad A. Mandegar; Nathaniel Huebsch; Frolov Eb; Shin E; Annie Truong; Michael P. Olvera; Amanda H. Chan; Yuichiro Miyaoka; Holmes K; Spencer Ci; Luke M. Judge; David E. Gordon; Tilde Eskildsen; Jacqueline E. Villalta; Max A. Horlbeck; Luke A. Gilbert; Nevan J. Krogan; Søren Paludan Sheikh; Jonathan S. Weissman; Lei S. Qi; Po-Lin So; Bruce R. Conklin

Developing technologies for efficient and scalable disruption of gene expression will provide powerful tools for studying gene function, developmental pathways, and disease mechanisms. Here, we develop clustered regularly interspaced short palindromic repeat interference (CRISPRi) to repress gene expression in human induced pluripotent stem cells (iPSCs). CRISPRi, in which a doxycycline-inducible deactivated Cas9 is fused to a KRAB repression domain, can specifically and reversibly inhibit gene expression in iPSCs and iPSC-derived cardiac progenitors, cardiomyocytes, and T lymphocytes. This gene repression system is tunable and has the potential to silence single alleles. Compared with CRISPR nuclease (CRISPRn), CRISPRi gene repression is more efficient and homogenous across cell populations. The CRISPRi system in iPSCs provides a powerful platform to perform genome-scale screens in a wide range of iPSC-derived cell types, dissect developmental pathways, and model disease.


Science | 2012

ER cargo properties specify a requirement for COPII coat rigidity mediated by Sec13p.

Alenka Čopič; Catherine F. Latham; Max A. Horlbeck; Jennifer G. D’Arcangelo; Elizabeth A. Miller

A Fair COP During eukaryotic intracellular membrane traffic, how is membrane curvature imparted by the cytoplasmic proteins that form the COPII coat, which mediates vesicle budding from the endoplasmic reticulum? Čopič et al. (p. 1359, published online 2 February; see the Perspective by Silvius) dissected this process by exploiting yeast bypass-of-sec-thirteen (bst) mutants, which can survive without the otherwise essential COPII coat protein. These bst mutants appear to create a locally altered membrane that is more amenable to deformation by a Sec13-free coat. Membrane curvature of cellular vesicles is generated by altering the symmetry of the cargo and the rigidity of coat proteins. Eukaryotic secretory proteins exit the endoplasmic reticulum (ER) via transport vesicles generated by the essential coat protein complex II (COPII) proteins. The outer coat complex, Sec13-Sec31, forms a scaffold that is thought to enforce curvature. By exploiting yeast bypass-of-sec-thirteen (bst) mutants, where Sec13p is dispensable, we probed the relationship between a compromised COPII coat and the cellular context in which it could still function. Genetic and biochemical analyses suggested that Sec13p was required to generate vesicles from membranes that contained asymmetrically distributed cargoes that were likely to confer opposing curvature. Thus, Sec13p may rigidify the COPII cage and increase its membrane-bending capacity; this function could be bypassed when a bst mutation renders the membrane more deformable.


eLife | 2016

Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation

Max A. Horlbeck; Luke A. Gilbert; Jacqueline E. Villalta; Britt Adamson; Ryan A. Pak; Yuwen Chen; Alexander P. Fields; Chong Yon Park; Jacob E. Corn; Martin Kampmann; Jonathan S. Weissman

We recently found that nucleosomes directly block access of CRISPR/Cas9 to DNA (Horlbeck et al., 2016). Here, we build on this observation with a comprehensive algorithm that incorporates chromatin, position, and sequence features to accurately predict highly effective single guide RNAs (sgRNAs) for targeting nuclease-dead Cas9-mediated transcriptional repression (CRISPRi) and activation (CRISPRa). We use this algorithm to design next-generation genome-scale CRISPRi and CRISPRa libraries targeting human and mouse genomes. A CRISPRi screen for essential genes in K562 cells demonstrates that the large majority of sgRNAs are highly active. We also find CRISPRi does not exhibit any detectable non-specific toxicity recently observed with CRISPR nuclease approaches. Precision-recall analysis shows that we detect over 90% of essential genes with minimal false positives using a compact 5 sgRNA/gene library. Our results establish CRISPRi and CRISPRa as premier tools for loss- or gain-of-function studies and provide a general strategy for identifying Cas9 target sites. DOI: http://dx.doi.org/10.7554/eLife.19760.001


Nature Chemical Biology | 2016

Parallel shRNA and CRISPR-Cas9 screens enable antiviral drug target identification

Richard M Deans; David W. Morgens; Ayse Okesli; Sirika Pillay; Max A. Horlbeck; Martin Kampmann; Luke A. Gilbert; Amy Li; Roberto Mateo; Mark Smith; Jeffrey S. Glenn; Jan E. Carette; Chaitan Khosla; Michael C. Bassik

Broad spectrum antiviral drugs targeting host processes could potentially treat a wide range of viruses while reducing the likelihood of emergent resistance. Despite great promise as therapeutics, such drugs remain largely elusive. Here we use parallel genome-wide high-coverage shRNA and CRISPR-Cas9 screens to identify the cellular target and mechanism of action of GSK983, a potent broad spectrum antiviral with unexplained cytotoxicity1–3. We show that GSK983 blocks cell proliferation and dengue virus replication by inhibiting the pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase (DHODH). Guided by mechanistic insights from both genomic screens, we found that exogenous deoxycytidine markedly reduces GSK983 cytotoxicity but not antiviral activity, providing an attractive novel approach to improve the therapeutic window of DHODH inhibitors against RNA viruses. Together, our results highlight the distinct advantages and limitations of each screening method for identifying drug targets and demonstrate the utility of parallel knockdown and knockout screens for comprehensively probing drug activity.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Next-generation libraries for robust RNA interference-based genome-wide screens

Martin Kampmann; Max A. Horlbeck; Yuwen Chen; Jordan C. Tsai; Michael C. Bassik; Luke A. Gilbert; Jacqueline E. Villalta; S. Chul Kwon; Hyeshik Chang; V. Narry Kim; Jonathan S. Weissman

Significance Genetic screening is a classic approach to identify genes acting in a biological process of interest. In mammalian cells, screens are commonly based on RNA interference (RNAi), in which a short interfering RNA (siRNA) or short-hairpin RNA (shRNA) triggers degradation of cellular messenger RNAs. RNAi approaches are prone to false-positive results because of siRNA/shRNA off-target effects and false-negative results because of siRNAs/shRNAs lacking activity. We previously established that these problems can be minimized with ultracomplex shRNA libraries. Here, we present next-generation shRNA libraries targeting the human and mouse genomes, for which we improved several features to increase shRNA activity. In a pilot screen, the new library yields complementary results to clustered regularly interspaced short palindromic repeats interference (CRISPRi), an orthogonal approach we developed recently. Genetic screening based on loss-of-function phenotypes is a powerful discovery tool in biology. Although the recent development of clustered regularly interspaced short palindromic repeats (CRISPR)-based screening approaches in mammalian cell culture has enormous potential, RNA interference (RNAi)-based screening remains the method of choice in several biological contexts. We previously demonstrated that ultracomplex pooled short-hairpin RNA (shRNA) libraries can largely overcome the problem of RNAi off-target effects in genome-wide screens. Here, we systematically optimize several aspects of our shRNA library, including the promoter and microRNA context for shRNA expression, selection of guide strands, and features relevant for postscreen sample preparation for deep sequencing. We present next-generation high-complexity libraries targeting human and mouse protein-coding genes, which we grouped into 12 sublibraries based on biological function. A pilot screen suggests that our next-generation RNAi library performs comparably to current CRISPR interference (CRISPRi)-based approaches and can yield complementary results with high sensitivity and high specificity.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Versatile in vivo regulation of tumor phenotypes by dCas9-mediated transcriptional perturbation

Christian Braun; Peter M. Bruno; Max A. Horlbeck; Luke A. Gilbert; Jonathan S. Weissman; Michael T. Hemann

Significance Tumor development is accompanied by widespread genomic and transcriptional changes. The mere acquisition of this information has greatly outpaced our capability to functionally study the biological roles of altered genes. This dilemma highlights the necessity to develop technologies that facilitate a rapid functional prioritization among lists of altered genes. Here, we use catalytically dead Cas9 to specifically activate or inactivate the transcription of genes in mouse models of cancer. This approach allows us to study the impact of gene-level changes in vivo and to systematically screen for novel genetic mediators of treatment relapse. We expect that this approach can be used to systematically dissect the biological role of cancer-related genes, a process critical to identifying new cancer drug targets. Targeted transcriptional regulation is a powerful tool to study genetic mediators of cellular behavior. Here, we show that catalytically dead Cas9 (dCas9) targeted to genomic regions upstream or downstream of the transcription start site allows for specific and sustainable gene-expression level alterations in tumor cells in vitro and in syngeneic immune-competent mouse models. We used this approach for a high-coverage pooled gene-activation screen in vivo and discovered previously unidentified modulators of tumor growth and therapeutic response. Moreover, by using dCas9 linked to an activation domain, we can either enhance or suppress target gene expression simply by changing the genetic location of dCas9 binding relative to the transcription start site. We demonstrate that these directed changes in gene-transcription levels occur with minimal off-target effects. Our findings highlight the use of dCas9-mediated transcriptional regulation as a versatile tool to reproducibly interrogate tumor phenotypes in vivo.

Collaboration


Dive into the Max A. Horlbeck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuwen Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Britt Adamson

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel He

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge