Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Genom ; 2(11)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36465279

ABSTRACT

During pandemics, individuals exhibit differences in risk and clinical outcomes. Here, we developed single-cell high-throughput human in vitro susceptibility testing (scHi-HOST), a method for rapidly identifying genetic variants that confer resistance and susceptibility. We applied this method to influenza A virus (IAV), the cause of four pandemics since the start of the 20th century. scHi-HOST leverages single-cell RNA sequencing (scRNA-seq) to simultaneously assign genetic identity to cells in mixed infections of cell lines of European, African, and Asian origin, reveal associated genetic variants for viral burden, and identify expression quantitative trait loci. Integration of scHi-HOST with human challenge and experimental validation demonstrated that a missense variant in endoplasmic reticulum aminopeptidase 1 (ERAP1; rs27895) increased IAV burden in cells and human volunteers. rs27895 exhibits population differentiation, likely contributing to greater permissivity of cells from African populations to IAV. scHi-HOST is a broadly applicable method and resource for decoding infectious-disease genetics.

2.
Cell Host Microbe ; 30(12): 1671-1684.e9, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36084633

ABSTRACT

Chlamydia trachomatis is the leading cause of sexually transmitted bacterial infections and a major threat to women's reproductive health in particular. This obligate intracellular pathogen resides and replicates within a cellular compartment termed an inclusion, where it is sheltered by unknown mechanisms from gamma-interferon (IFNγ)-induced cell-autonomous host immunity. Through a genetic screen, we uncovered the Chlamydia inclusion membrane protein gamma resistance determinant (GarD) as a bacterial factor protecting inclusions from cell-autonomous immunity. In IFNγ-primed human cells, inclusions formed by garD loss-of-function mutants become decorated with linear ubiquitin and are eliminated. Leveraging cellular genome-wide association data, we identified the ubiquitin E3 ligase RNF213 as a candidate anti-Chlamydia protein. We demonstrate that IFNγ-inducible RNF213 facilitates the ubiquitylation and destruction of GarD-deficient inclusions. Furthermore, we show that GarD operates as a cis-acting stealth factor barring RNF213 from targeting inclusions, thus functionally defining GarD as an RNF213 antagonist essential for chlamydial growth during IFNγ-stimulated immunity.


Subject(s)
Bacterial Infections , Chlamydia Infections , Female , Humans , Chlamydia trachomatis/genetics , Genome-Wide Association Study , Chlamydia Infections/metabolism , Ubiquitination , Interferon-gamma/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , HeLa Cells , Adenosine Triphosphatases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Annu Rev Genet ; 56: 41-62, 2022 11 30.
Article in English | MEDLINE | ID: mdl-35697043

ABSTRACT

Since the identification of sickle cell trait as a heritable form of resistance to malaria, candidate gene studies, linkage analysis paired with sequencing, and genome-wide association (GWA) studies have revealed many examples of genetic resistance and susceptibility to infectious diseases. GWA studies enabled the identification of many common variants associated with small shifts in susceptibility to infectious diseases. This is exemplified by multiple loci associated with leprosy, malaria, HIV, tuberculosis, and coronavirus disease 2019 (COVID-19), which illuminate genetic architecture and implicate pathways underlying pathophysiology. Despite these successes, most of the heritability of infectious diseases remains to be explained. As the field advances, current limitations may be overcome by applying methodological innovations such as cellular GWA studies and phenome-wide association (PheWA) studies as well as by improving methodological rigor with more precise case definitions, deeper phenotyping, increased cohort diversity, and functional validation of candidate loci in the laboratory or human challenge studies.


Subject(s)
COVID-19 , Communicable Diseases , Humans , Genome-Wide Association Study , COVID-19/genetics , Communicable Diseases/genetics , Human Genetics
4.
mBio ; 13(3): e0346421, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35658533

ABSTRACT

Despite being in a golden age of bacterial epigenomics, little work has systematically examined the plasticity and functional impacts of the bacterial DNA methylome. Here, we leveraged single-molecule, real-time sequencing (SMRT-seq) to examine the m6A DNA methylome of two Salmonella enterica serovar Typhimurium strains: 14028s and a ΔmetJ mutant with derepressed methionine metabolism, grown in Luria broth or medium that simulates the intracellular environment. We found that the methylome is remarkably static: >95% of adenosine bases retain their methylation status across conditions. Integration of methylation with transcriptomic data revealed limited correlation between changes in methylation and gene expression. Further, examination of the transcriptome in ΔyhdJ bacteria lacking the m6A methylase with the most dynamic methylation pattern in our data set revealed little evidence of YhdJ-mediated gene regulation. Curiously, despite G(m6A)TC motifs being particularly resistant to change across conditions, incorporating dam mutants into our analyses revealed two examples where changes in methylation and transcription may be linked across conditions. This includes the novel finding that the ΔmetJ motility defect may be partially driven by hypermethylation of the chemotaxis gene tsr. Together, these data redefine the S. Typhimurium epigenome as a highly stable system that has rare but important roles in transcriptional regulation. Incorporating these lessons into future studies will be critical as we progress through the epigenomic era. IMPORTANCE While recent breakthroughs have enabled intense study of bacterial DNA modifications, limitations in current work have potentiated a surprisingly untested narrative that DNA methylation is a common mechanism of the bacterial response to environmental conditions. Essentially, whether epigenetic regulation of bacterial transcription is a common, generalizable phenomenon is a critical unanswered question that we address here. We found that most DNA methylation is static in Salmonella enterica serovar Typhimurium, even when the bacteria are grown under dramatically different conditions that cause broad changes in the transcriptome. Further, even when the methylation of individual bases change, these changes generally do not correlate with changes in gene expression. Finally, we demonstrate methods by which data can be stratified in order to identify coupled changes in methylation and gene expression.


Subject(s)
Epigenome , Salmonella typhimurium , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Bacterial , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Transcriptome , Virulence/genetics
5.
HGG Adv ; 3(1): 100071, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35047856

ABSTRACT

Human genetic diversity can have profound effects on health outcomes upon exposure to infectious agents. For infections with Chlamydia trachomatis (C. trachomatis), the wide range of genital and ocular disease manifestations are likely influenced by human genetic differences that regulate interactions between C. trachomatis and host cells. We leveraged this diversity in cellular responses to demonstrate the importance of variation at the Toll-like receptor 1 (TLR1), TLR6, and TLR10 locus to cytokine production in response to C. trachomatis. We determined that a single-nucleotide polymorphism (SNP) (rs1057807), located in a region that forms a loop with the TLR6 promoter, is associated with increased expression of TLR1, TLR6, and TLR10 and secreted levels of ten C. trachomatis-induced cytokines. Production of these C. trachomatis-induced cytokines is primarily dependent on MyD88 and TLR6 based on experiments using inhibitors, blocking antibodies, RNAi, and protein overexpression. Population genetic analyses further demonstrated that the mean IL-6 response of cells from two European populations were higher than the mean response of cells from three African populations and that this difference was partially attributable to variation in rs1057807 allele frequency. In contrast, a SNP associated with a different pro-inflammatory cytokine (rs2869462 associated with the chemokine CXCL10) exhibited an opposite response, underscoring the complexity of how different genetic variants contribute to an individual's immune response. This multidisciplinary study has identified a long-range chromatin interaction and genetic variation that regulates TLR6 to broaden our understanding of how human genetic variation affects the C. trachomatis-induced immune response.

6.
Sci Rep ; 10(1): 18269, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106516

ABSTRACT

Susceptibility to infectious diseases is determined by a complex interaction between host and pathogen. For infections with the obligate intracellular bacterium Chlamydia trachomatis, variation in immune activation and disease presentation are regulated by both host genetic diversity and pathogen immune evasion. Previously, we discovered a single nucleotide polymorphism (rs2869462) associated with absolute abundance of CXCL10, a pro-inflammatory T-cell chemokine. Here, we report that levels of CXCL10 change during C. trachomatis infection of cultured cells in a manner dependent on both host and pathogen. Linear modeling of cellular traits associated with CXCL10 levels identified a strong, negative correlation with bacterial burden, suggesting that C. trachomatis actively suppresses CXCL10. We identified the pathogen-encoded factor responsible for this suppression as the chlamydial protease- or proteasome-like activity factor, CPAF. Further, we applied our modeling approach to other host cytokines in response to C. trachomatis and found evidence that RANTES, another T-cell chemoattractant, is actively suppressed by Chlamydia. However, this observed suppression of RANTES is not mediated by CPAF. Overall, our results demonstrate that CPAF suppresses CXCL10 to evade the host cytokine response and that modeling of cellular infection parameters can reveal previously unrecognized facets of host-pathogen interactions.


Subject(s)
Chemokine CXCL10/genetics , Chlamydia Infections/genetics , Chlamydia trachomatis/enzymology , Endopeptidases/metabolism , Polymorphism, Single Nucleotide , Animals , Cell Line , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , Chlamydia Infections/metabolism , Chlamydia trachomatis/genetics , Chlorocebus aethiops , HeLa Cells , Humans , Models, Biological , Vero Cells
7.
PLoS Genet ; 16(6): e1008774, 2020 06.
Article in English | MEDLINE | ID: mdl-32555736

ABSTRACT

Cranial neural crest (NC) contributes to the developing vertebrate eye. By multidimensional, quantitative imaging, we traced the origin of the ocular NC cells to two distinct NC populations that differ in the maintenance of sox10 expression, Wnt signalling, origin, route, mode and destination of migration. The first NC population migrates to the proximal and the second NC cell group populates the distal (anterior) part of the eye. By analysing zebrafish pax6a/b compound mutants presenting anterior segment dysgenesis, we demonstrate that Pax6a/b guide the two NC populations to distinct proximodistal locations. We further provide evidence that the lens whose formation is pax6a/b-dependent and lens-derived TGFß signals contribute to the building of the anterior segment. Taken together, our results reveal multiple roles of Pax6a/b in the control of NC cells during development of the anterior segment.


Subject(s)
Anterior Eye Segment/metabolism , Neural Crest/metabolism , Neurogenesis , PAX6 Transcription Factor/metabolism , Zebrafish Proteins/metabolism , Animals , Anterior Eye Segment/cytology , Anterior Eye Segment/embryology , Cell Movement , Mutation , Neural Crest/cytology , Neural Crest/embryology , Neurons/cytology , Neurons/metabolism , PAX6 Transcription Factor/genetics , Signal Transduction , Transforming Growth Factor beta/metabolism , Zebrafish , Zebrafish Proteins/genetics
8.
Article in English | MEDLINE | ID: mdl-31440475

ABSTRACT

Clearance of intracellular pathogens, such as Leishmania (L.) major, depends on an immune response with well-regulated cytokine signaling. Here we describe a pathogen-mediated mechanism of evading CXCL10, a chemokine with diverse antimicrobial functions, including T cell recruitment. Infection with L. major in a human monocyte cell line induced robust CXCL10 transcription without increasing extracellular CXCL10 protein concentrations. We found that this transcriptionally independent suppression of CXCL10 is mediated by the virulence factor and protease, glycoprotein-63 (gp63). Specifically, GP63 cleaves CXCL10 after amino acid A81 at the base of a C-terminal alpha-helix. Cytokine cleavage by GP63 demonstrated specificity, as GP63 cleaved CXCL10 and its homologs, which all bind the CXCR3 receptor, but not distantly related chemokines, such as CXCL8 and CCL22. Further characterization demonstrated that CXCL10 cleavage activity by GP63 was produced by both extracellular promastigotes and intracellular amastigotes. Crucially, CXCL10 cleavage impaired T cell chemotaxis in vitro, indicating that cleaved CXCL10 cannot signal through CXCR3. Ultimately, we propose CXCL10 suppression is a convergent mechanism of immune evasion, as Salmonella enterica and Chlamydia trachomatis also suppress CXCL10. This commonality suggests that counteracting CXCL10 suppression may provide a generalizable therapeutic strategy against intracellular pathogens. Importance: Leishmaniasis, an infectious disease that annually affects over one million people, is caused by intracellular parasites that have evolved to evade the host's attempts to eliminate the parasite. Cutaneous leishmaniasis results in disfiguring skin lesions if the host immune system does not appropriately respond to infection. A family of molecules called chemokines coordinate recruitment of the immune cells required to eliminate infection. Here, we demonstrate a novel mechanism that Leishmania (L.) spp. employ to suppress host chemokines: a Leishmania-encoded protease cleaves chemokines known to recruit T cells that fight off infection. We observe that other common human intracellular pathogens, including Chlamydia trachomatis and Salmonella enterica, reduce levels of the same chemokines, suggesting a strong selective pressure to avoid this component of the immune response. Our study provides new insights into how intracellular pathogens interact with the host immune response to enhance pathogen survival.


Subject(s)
Chemokine CXCL10/antagonists & inhibitors , Immune Evasion , Immunologic Factors/antagonists & inhibitors , Leishmania major/growth & development , Leishmania major/immunology , Monocytes/immunology , Monocytes/parasitology , Cell Line , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Humans , Immunosuppression Therapy , Metalloendopeptidases/metabolism , Protein Biosynthesis , Proteolysis , Salmonella enterica/growth & development , Salmonella enterica/immunology , T-Lymphocytes/immunology , Transcription, Genetic
9.
PLoS Comput Biol ; 14(4): e1006128, 2018 04.
Article in English | MEDLINE | ID: mdl-29672531

ABSTRACT

State-of-the-art light-sheet and confocal microscopes allow recording of entire embryos in 3D and over time (3D+t) for many hours. Fluorescently labeled structures can be segmented and tracked automatically in these terabyte-scale 3D+t images, resulting in thousands of cell migration trajectories that provide detailed insights to large-scale tissue reorganization at the cellular level. Here we present EmbryoMiner, a new interactive open-source framework suitable for in-depth analyses and comparisons of entire embryos, including an extensive set of trajectory features. Starting at the whole-embryo level, the framework can be used to iteratively focus on a region of interest within the embryo, to investigate and test specific trajectory-based hypotheses and to extract quantitative features from the isolated trajectories. Thus, the new framework provides a valuable new way to quantitatively compare corresponding anatomical regions in different embryos that were manually selected based on biological prior knowledge. As a proof of concept, we analyzed 3D+t light-sheet microscopy images of zebrafish embryos, showcasing potential user applications that can be performed using the new framework.


Subject(s)
Cell Tracking/statistics & numerical data , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Movement , Computational Biology , Embryonic Development , Embryonic Stem Cells/cytology , Gastrulation , Germ Layers/cytology , Imaging, Three-Dimensional , Microscopy, Fluorescence , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Software
10.
Mol Cancer Res ; 13(5): 934-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25678598

ABSTRACT

UNLABELLED: Epithelial-to-mesenchymal transition (EMT) has been implicated in models of tumor cell migration, invasion, and metastasis. In a search for candidate therapeutic targets to reverse this process, nontumorigenic MCF10A breast epithelial cells were infected with an arrayed lentiviral kinome shRNA library and screened for either suppression or enhancement of a 26-gene EMT RNA signature. No individual kinase gene knockdown was sufficient to induce EMT. In contrast, grouped epithelial markers were induced by knockdown of multiple kinases, including mitogen activated protein kinase 7 (MAPK7). In breast cancer cells, suppression of MAPK7 increased E-cadherin (CDH1) expression and inhibited cell migration. In an orthotopic mouse model, MAPK7 suppression reduced the generation of circulating tumor cells and the appearance of lung metastases. Together, these observations raise the possibility that targeting kinases that maintain mesenchymal cell properties in cancer cells, such as MAPK7, may lessen tumor invasiveness. IMPLICATIONS: Suppression of MAPK7 induces epithelial markers, reduces generation of circulating tumor cells and appearance of lung metastases.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Mitogen-Activated Protein Kinase 7/metabolism , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Animals , Antigens, CD , Breast Neoplasms/blood , Cadherins/biosynthesis , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mitogen-Activated Protein Kinase 7/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcriptome
11.
Genes Dev ; 24(23): 2654-65, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21062900

ABSTRACT

To define the functional pathways regulating epithelial cell migration, we performed a genome-wide RNAi screen using 55,000 pooled lentiviral shRNAs targeting ∼11,000 genes, selecting for transduced cells with increased motility. A stringent validation protocol generated a set of 31 genes representing diverse pathways whose knockdown dramatically enhances cellular migration. Some of these pathways share features of epithelial-to-mesenchymal transition (EMT), and together they implicate key regulators of transcription, cellular signaling, and metabolism, as well as novel modulators of cellular trafficking, such as DLG5. In delineating downstream pathways mediating these migration phenotypes, we observed universal activation of ERKs and a profound dependence on their RSK effectors. Pharmacological inhibition of RSK dramatically suppresses epithelial cell migration induced by knockdown of all 31 genes, suggesting that convergence of diverse migratory pathways on this kinase may provide a therapeutic opportunity in disorders of cell migration, including cancer metastasis.


Subject(s)
Cell Movement/genetics , Genome-Wide Association Study , RNA Interference , Ribosomal Protein S6 Kinases/metabolism , Cell Line, Tumor , Epithelial Cells/cytology , Humans , Membrane Proteins/metabolism , Mesoderm/cytology , Reproducibility of Results , Tumor Suppressor Proteins/metabolism
12.
Genes Dev ; 23(15): 1737-42, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19651985

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) plays an important role during normal embryogenesis, and it has been implicated in cancer invasion and metastasis. Here, we report that Ladybird homeobox 1 (LBX1), a developmentally regulated homeobox gene, directs expression of the known EMT inducers ZEB1, ZEB2, Snail1, and transforming growth factor beta2 (TGFB2). In mammary epithelial cells, overexpression of LBX1 leads to morphological transformation, expression of mesenchymal markers, enhanced cell migration, increased CD44(high)/CD24(low) progenitor cell population, and tumorigenic cooperation with known oncogenes. In human breast cancer, LBX1 is up-regulated in the unfavorable estrogen receptor (ER)/progesterone (PR)/HER2 triple-negative basal-like subtype. Thus, aberrant expression of LBX1 may lead to the activation of a developmentally regulated EMT pathway in human breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Disease Progression , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Organic Cation Transport Proteins/metabolism , Transcription Factors/metabolism , Animals , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Movement , Homeodomain Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Oncogenes/physiology , Repressor Proteins/metabolism , Snail Family Transcription Factors , Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta2/metabolism , Up-Regulation , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
13.
Genes Dev ; 21(17): 2131-6, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17704304

ABSTRACT

The neural crest (NC) is a highly motile cell population that gives rise to multiple tissue lineages during vertebrate embryogenesis. Here, we identify a novel effector of the small GTPase Rap, called RADIL, and show that it is required for cell adhesion and migration. Knockdown of radil in the zebrafish model results in multiple defects in NC-derived lineages such as cartilage, pigment cells, and enteric neurons. We specifically show that these defects are primarily due to the diminished migratory capacity of NC cells. The identification of RADIL as a regulator of NC migration defines a role for the Rap pathway in this process.


Subject(s)
Carrier Proteins/physiology , Cell Adhesion/genetics , Cell Movement/genetics , Genes, vpr , Morphogenesis/genetics , Neural Crest/embryology , Zebrafish Proteins/physiology , rap GTP-Binding Proteins/physiology , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Genes, ras
SELECTION OF CITATIONS
SEARCH DETAIL
...