Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.063
Filter
Add more filters

Publication year range
1.
Cell ; 186(16): 3499-3518.e14, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37437571

ABSTRACT

Chloroplasts are eukaryotic photosynthetic organelles that drive the global carbon cycle. Despite their importance, our understanding of their protein composition, function, and spatial organization remains limited. Here, we determined the localizations of 1,034 candidate chloroplast proteins using fluorescent protein tagging in the model alga Chlamydomonas reinhardtii. The localizations provide insights into the functions of poorly characterized proteins; identify novel components of nucleoids, plastoglobules, and the pyrenoid; and reveal widespread protein targeting to multiple compartments. We discovered and further characterized cellular organizational features, including eleven chloroplast punctate structures, cytosolic crescent structures, and unexpected spatial distributions of enzymes within the chloroplast. We also used machine learning to predict the localizations of other nuclear-encoded Chlamydomonas proteins. The strains and localization atlas developed here will serve as a resource to accelerate studies of chloroplast architecture and functions.


Subject(s)
Biosynthetic Pathways , Chlamydomonas reinhardtii , Chloroplast Proteins , Chlamydomonas reinhardtii/metabolism , Chloroplast Proteins/metabolism , Chloroplasts/metabolism , Photosynthesis
2.
Cell ; 185(24): 4488-4506.e20, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36318922

ABSTRACT

When challenged by hypertonicity, dehydrated cells must recover their volume to survive. This process requires the phosphorylation-dependent regulation of SLC12 cation chloride transporters by WNK kinases, but how these kinases are activated by cell shrinkage remains unknown. Within seconds of cell exposure to hypertonicity, WNK1 concentrates into membraneless condensates, initiating a phosphorylation-dependent signal that drives net ion influx via the SLC12 cotransporters to restore cell volume. WNK1 condensate formation is driven by its intrinsically disordered C terminus, whose evolutionarily conserved signatures are necessary for efficient phase separation and volume recovery. This disorder-encoded phase behavior occurs within physiological constraints and is activated inĀ vivo by molecular crowding rather than changes in cell size. This allows kinase activity despite an inhibitory ionic milieu and permits cell volume recovery through condensate-mediated signal amplification. Thus, WNK kinases are physiological crowding sensors that phase separate to coordinate a cell volume rescue response.


Subject(s)
Protein Serine-Threonine Kinases , Phosphorylation , Cell Size
3.
Cell ; 184(16): 4154-4167.e12, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34324837

ABSTRACT

Environmental light cycles entrain circadian feeding behaviors in animals that produce rhythms in exposure to foodborne bacteria. Here, we show that the intestinal microbiota generates diurnal rhythms in innate immunity that synchronize with feeding rhythms to anticipate microbial exposure. Rhythmic expression of antimicrobial proteins was driven by daily rhythms in epithelial attachment by segmented filamentous bacteria (SFB), members of the mouse intestinal microbiota. Rhythmic SFB attachment was driven by the circadian clock through control of feeding rhythms. Mechanistically, rhythmic SFB attachment activated an immunological circuit involving group 3 innate lymphoid cells. This circuit triggered oscillations in epithelial STAT3 expression and activation that produced rhythmic antimicrobial protein expression and caused resistance to Salmonella Typhimurium infection to vary across the day-night cycle. Thus, host feeding rhythms synchronize with the microbiota to promote rhythms in intestinal innate immunity that anticipate exogenous microbial exposure.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Gastrointestinal Microbiome , Immunity, Innate , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Adhesion , Cell Adhesion , Epithelial Cells/microbiology , Feeding Behavior , Intestine, Small/microbiology , Intestine, Small/ultrastructure , Lymphocytes/metabolism , Mice, Inbred C57BL , Muramidase/metabolism , Pancreatitis-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Salmonella Infections, Animal/microbiology , Signal Transduction
4.
Cell ; 179(1): 205-218.e21, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522888

ABSTRACT

The molecular chaperone HSP90 facilitates the folding of several client proteins, including innate immune receptors and protein kinases. HSP90 is an essential component of plant and animal immunity, yet pathogenic strategies that directly target the chaperone have not been described. Here, we identify the HopBF1 family of bacterial effectors as eukaryotic-specific HSP90 protein kinases. HopBF1 adopts a minimal protein kinase fold that is recognized by HSP90 as a host client. As a result, HopBF1 phosphorylates HSP90 to completely inhibit the chaperone's ATPase activity. We demonstrate that phosphorylation of HSP90 prevents activation of immune receptors that trigger the hypersensitive response in plants. Consequently, HopBF1-dependent phosphorylation of HSP90 is sufficient to induce severe disease symptoms in plants infected with the bacterial pathogen, Pseudomonas syringae. Collectively, our results uncover a family of bacterial effector kinases with toxin-like properties and reveal a previously unrecognized betrayal mechanism by which bacterial pathogens modulate host immunity.


Subject(s)
Arabidopsis Proteins/metabolism , Bacterial Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Mimicry/immunology , Plant Immunity/physiology , Adenosine Triphosphatases/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/microbiology , Bacterial Proteins/chemistry , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , Host Microbial Interactions/immunology , Humans , Phosphorylation , Plasmids/genetics , Protein Binding , Protein Folding , Protein Kinases/metabolism , Pseudomonas syringae/metabolism , Saccharomyces cerevisiae/metabolism
5.
Immunity ; 57(8): 1769-1779.e4, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38901428

ABSTRACT

Many infections, including malaria, are associated with an increase in autoantibodies (AAbs). Prior studies have reported an association between genetic markers of susceptibility to autoimmune disease and resistance to malaria, but the underlying mechanisms are unclear. Here, we performed a longitudinal study of children and adults (nĀ = 602) in Mali and found that high levels of plasma AAbs before the malaria season independently predicted a reduced risk of clinical malaria in children during the ensuing malaria season. Baseline AAb seroprevalence increased with age and asymptomatic Plasmodium falciparum infection. We found that AAbs purified from the plasma of protected individuals inhibit the growth of blood-stage parasites and bind P.Ā falciparum proteins that mediate parasite invasion. Protected individuals had higher plasma immunoglobulin G (IgG) reactivity against 33 of the 123 antigens assessed in an autoantigen microarray. This study provides evidence in support of the hypothesis that a propensity toward autoimmunity offers a survival advantage against malaria.


Subject(s)
Autoantibodies , Immunoglobulin G , Malaria, Falciparum , Plasmodium falciparum , Humans , Plasmodium falciparum/immunology , Autoantibodies/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Child , Child, Preschool , Adult , Immunoglobulin G/immunology , Immunoglobulin G/blood , Female , Mali , Male , Adolescent , Antibodies, Protozoan/immunology , Longitudinal Studies , Infant , Antigens, Protozoan/immunology , Young Adult , Autoantigens/immunology , Seroepidemiologic Studies , Middle Aged
6.
Cell ; 175(3): 809-821.e19, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30270044

ABSTRACT

Approximately 10% of human protein kinases are believed to be inactive and named pseudokinases because they lack residues required for catalysis. Here, we show that the highly conserved pseudokinase selenoprotein-O (SelO) transfers AMP from ATP to Ser, Thr, and Tyr residues on protein substrates (AMPylation), uncovering a previously unrecognized activity for a member of the protein kinase superfamily. The crystal structure of a SelO homolog reveals a protein kinase-like fold with ATP flipped in the active site, thus providing a structural basis for catalysis. SelO pseudokinases localize to the mitochondria and AMPylate proteins involved in redox homeostasis. Consequently, SelO activity is necessary for the proper cellular response to oxidative stress. Our results suggest that AMPylation may be a more widespread post-translational modification than previously appreciated and that pseudokinases should be analyzed for alternative transferase activities.


Subject(s)
Adenosine Monophosphate/metabolism , Catalytic Domain , Protein Processing, Post-Translational , Selenoproteins/metabolism , Conserved Sequence , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Stress , Selenoproteins/chemistry
7.
Cell ; 162(3): 635-47, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232229

ABSTRACT

Recent progress in understanding the diversity of midbrain dopamine neurons has highlighted the importance--and the challenges--of defining mammalian neuronal cell types. Although neurons may be best categorized using inclusive criteria spanning biophysical properties, wiring of inputs, wiring of outputs, and activity during behavior, linking all of these measurements to cell types within the intact brains of living mammals has been difficult. Here, using an array of intact-brain circuit interrogation tools, including CLARITY, COLM, optogenetics, viral tracing, and fiber photometry, we explore the diversity of dopamine neurons within the substantia nigra pars compacta (SNc). We identify two parallel nigrostriatal dopamine neuron subpopulations differing in biophysical properties, input wiring, output wiring to dorsomedial striatum (DMS) versus dorsolateral striatum (DLS), and natural activity patterns during free behavior. Our results reveal independently operating nigrostriatal information streams, with implications for understanding the logic of dopaminergic feedback circuits and the diversity of mammalian neuronal cell types.


Subject(s)
Neural Pathways , Neurons/metabolism , Substantia Nigra/metabolism , Animals , Brain Mapping , Dopamine/metabolism , Mice , Mice, Inbred C57BL , Reward , Shock
8.
Cell ; 163(6): 1457-67, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26627735

ABSTRACT

A variety of signals finely tune insulin secretion by pancreatic Ɵ cells to prevent both hyper-and hypoglycemic states. Here, we show that post-translational regulation of the transcription factors ETV1, ETV4, and ETV5 by the ubiquitin ligase COP1 (also called RFWD2) in Ɵ cells is critical for insulin secretion. Mice lacking COP1 in Ɵ cells developed diabetes due to insulin granule docking defects that were fully rescued by genetic deletion of Etv1, Etv4, and Etv5. Genes regulated by ETV1, ETV4, or ETV5 in the absence of mouse COP1 were enriched in human diabetes-associated genes, suggesting that they also influence human Ɵ-cell pathophysiology. In normal Ɵ cells, ETV4 was stabilized upon membrane depolarization and limited insulin secretion under hyperglycemic conditions. Collectively, our data reveal that ETVs negatively regulate insulin secretion for the maintenance of normoglycemia.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/metabolism , Diabetes Mellitus/metabolism , Exocytosis , Gene Deletion , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Insulin Secretion , Mice , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
9.
Nature ; 626(8000): 881-890, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297124

ABSTRACT

The pace of human brain development is highly protracted compared with most other species1-7. The maturation of cortical neurons is particularly slow, taking months to years to develop adult functions3-5. Remarkably, such protracted timing is retained in cortical neurons derived from human pluripotent stemĀ cells (hPSCs) during in vitro differentiation or upon transplantation into the mouse brain4,8,9. Those findings suggest the presence of a cell-intrinsic clock setting the pace of neuronal maturation, although the molecular nature of this clock remains unknown. Here we identify an epigenetic developmental programme that sets the timing of human neuronal maturation. First, we developed a hPSC-based approach to synchronize the birth of cortical neurons in vitro which enabled us to define an atlas of morphological, functional and molecular maturation. We observed a slow unfolding of maturation programmes, limited by the retention of specific epigenetic factors. Loss of function of several of those factors in cortical neurons enables precocious maturation. Transient inhibition of EZH2, EHMT1 and EHMT2 or DOT1L, at progenitor stage primes newly born neurons to rapidly acquire mature properties upon differentiation. Thus our findings reveal that the rate at which human neurons mature is set well before neurogenesis through the establishment of an epigenetic barrier in progenitor cells. Mechanistically, this barrier holds transcriptional maturation programmes in a poised state that is gradually released to ensure the prolonged timeline of human cortical neuron maturation.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells , Neural Stem Cells , Neurogenesis , Neurons , Adult , Animals , Humans , Mice , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Time Factors , Transcription, Genetic
10.
Physiol Rev ; 102(1): 339-341, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34494892

ABSTRACT

During the COVID-19 pandemic, efforts have been made worldwide to develop effective therapies to address the devastating immune-mediated effects of SARS-CoV-2. With the exception of monoclonal antibody-mediated therapeutics and preventive approaches such as mass immunization, most experimental or repurposed drugs have failed in large randomized clinical trials (https://www.who.int/publications/i/item/therapeutics-and-covid-19-living-guideline). The worldwide spread of SARS-CoV-2 virus revealed specific susceptibilities to the virus among the elderly and individuals with age-related syndromes. These populations were more likely to experience a hyperimmune response characterized by a treatment-resistant acute lung pathology accompanied by multiple organ failure. These observations underscore the interplay between the virus, the biology of aging, and outcomes observed in the most severe cases of SARS-CoV-2 infection. The ectoenzyme CD38 has been implicated in the process of "inflammaging" in aged tissues. In a current publication, Horenstein et al. present evidence to support the hypothesis that CD38 plays a central role in altered immunometabolism resulting from COVID-19 infection. The authors discuss a critical but underappreciated trifecta of CD38-mediated NAD+ metabolism, aging, and COVID-19 immune response and speculate that the CD38/NAD+ axis is a promising therapeutic target for this disease.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , COVID-19/physiopathology , Membrane Glycoproteins/metabolism , SARS-CoV-2 , ADP-ribosyl Cyclase 1/genetics , Aging , Gene Expression Regulation, Enzymologic , Humans , Membrane Glycoproteins/genetics , NAD/metabolism
11.
Cell ; 157(7): 1535-51, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949967

ABSTRACT

Social interaction is a complex behavior essential for many species and is impaired in major neuropsychiatric disorders. Pharmacological studies have implicated certain neurotransmitter systems in social behavior, but circuit-level understanding of endogenous neural activity during social interaction is lacking. We therefore developed and applied a new methodology, termed fiber photometry, to optically record natural neural activity in genetically and connectivity-defined projections to elucidate the real-time role of specified pathways in mammalian behavior. Fiber photometry revealed that activity dynamics of a ventral tegmental area (VTA)-to-nucleusĀ accumbens (NAc) projection could encode and predict key features of social, but not novel object, interaction. Consistent with this observation, optogenetic control of cells specifically contributing to this projection was sufficient to modulate social behavior, which was mediated by type 1 dopamine receptor signaling downstream in the NAc. Direct observation of deep projection-specific activity in this way captures a fundamental and previously inaccessible dimension of mammalian circuit dynamics.


Subject(s)
Neural Pathways , Nucleus Accumbens/physiology , Social Behavior , Ventral Tegmental Area/physiology , Animals , Calcium Signaling , Female , Mice , Nucleus Accumbens/cytology , Photometry/methods , Receptors, Dopamine/chemistry , Receptors, Dopamine/metabolism , Reward , Rhodopsin/chemistry , Rhodopsin/metabolism , Ventral Tegmental Area/cytology
12.
Mol Cell ; 81(3): 426-441.e8, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545059

ABSTRACT

Eukaryotic genomes replicate via spatially and temporally regulated origin firing. Cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK) promote origin firing, whereas the S phase checkpoint limits firing to prevent nucleotide and RPA exhaustion. We used chemical genetics to interrogate human DDK with maximum precision, dissect its relationship with the S phase checkpoint, and identify DDK substrates. We show that DDK inhibition (DDKi) leads to graded suppression of origin firing and fork arrest. S phase checkpoint inhibition rescued origin firing in DDKi cells and DDK-depleted Xenopus egg extracts. DDKi also impairs RPA loading, nascent-strand protection, and fork restart. Via quantitative phosphoproteomics, we identify the BRCA1-associated (BRCA1-A) complex subunit MERIT40 and the cohesin accessory subunit PDS5B as DDK effectors in fork protection and restart. Phosphorylation neutralizes autoinhibition mediated by intrinsically disordered regions in both substrates. Our results reveal mechanisms through which DDK controls the duplication of large vertebrate genomes.


Subject(s)
DNA Replication , Replication Origin , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , S Phase Cell Cycle Checkpoints , Substrate Specificity , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus laevis
13.
Cell ; 155(2): 384-96, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120137

ABSTRACT

Hepatocellular carcinoma (HCC) is a slowly developing malignancy postulated to evolve from premalignant lesions in chronically damaged livers. However, it was never established that premalignant lesions actually contain tumor progenitors that give rise to cancer. Here, we describe isolationĀ and characterization of HCC progenitor cells (HcPCs) from different mouse HCC models. Unlike fully malignant HCC, HcPCs give rise to cancer only when introduced into a liver undergoing chronic damage and compensatory proliferation. Although HcPCs exhibit a similar transcriptomic profile to bipotential hepatobiliary progenitors, the latter do not give rise to tumors. Cells resembling HcPCs reside within dysplastic lesions that appear several months before HCC nodules. Unlike early hepatocarcinogenesis, which depends on paracrine IL-6 production by inflammatory cells, due to upregulation of LIN28 expression, HcPCs had acquired autocrine IL-6 signaling that stimulates their inĀ vivo growth and malignant progression. This may be a general mechanism that drives other IL-6-producing malignancies.


Subject(s)
Autocrine Communication , Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Disease Progression , Hepacivirus , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL
14.
EMBO J ; 42(11): e112590, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36912146

ABSTRACT

During development, the lymphatic vasculature forms as a second network derived chiefly from blood vessels. The transdifferentiation of embryonic venous endothelial cells (VECs) into lymphatic endothelial cells (LECs) is a key step in this process. Specification, differentiation and maintenance of LEC fate are all driven by theĀ transcription factor Prox1, yet the downstream mechanisms remain to be elucidated. We here present a single-cell transcriptomic atlas of lymphangiogenesis in zebrafish, revealing new markers and hallmarks of LEC differentiation over four developmental stages. We further profile single-cell transcriptomic and chromatin accessibility changes in zygotic prox1a mutants that are undergoing a LEC-VEC fate shift. Using maternal and zygotic prox1a/prox1b mutants, we determine the earliest transcriptomic changes directed by Prox1 during LEC specification. This work altogether reveals new downstream targets and regulatory regions of the genome controlled by Prox1 and presents evidence that Prox1 specifies LEC fate primarily by limiting blood vascular and haematopoietic fate. This extensive single-cell resource provides new mechanistic insights into the enigmatic role of Prox1 and the control of LEC differentiation in development.


Subject(s)
Lymphatic Vessels , Zebrafish , Animals , Zebrafish/genetics , Homeodomain Proteins/genetics , Tumor Suppressor Proteins/genetics , Endothelial Cells , Cells, Cultured , Cell Differentiation , Lymphangiogenesis/genetics , Transcription Factors/genetics , Single-Cell Analysis
15.
Am J Hum Genet ; 111(9): 2012-2030, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39191256

ABSTRACT

Genome analysis of individuals affected by retinitis pigmentosa (RP) identified two rare nucleotide substitutions at the same genomic location on chromosome 11 (g.61392563 [GRCh38]), 69 base pairs upstream of the start codon of the ciliopathy gene TMEM216 (c.-69G>A, c.-69G>T [GenBank: NM_001173991.3]), in individuals of South Asian and African ancestry, respectively. Genotypes included 71 homozygotes and 3 mixed heterozygotes in trans with a predicted loss-of-function allele. Haplotype analysis showed single-nucleotide variants (SNVs) common across families, suggesting ancestral alleles within the two distinct ethnic populations. Clinical phenotype analysis of 62 available individuals from 49 families indicated a similar clinical presentation with night blindness in the first decade and progressive peripheral field loss thereafter. No evident systemic ciliopathy features were noted. Functional characterization of these variants by luciferase reporter gene assay showed reduced promotor activity. Nanopore sequencing confirmed the lower transcription of the TMEM216 c.-69G>T allele in blood-derived RNA from a heterozygous carrier, and reduced expression was further recapitulated by qPCR, using both leukocytes-derived RNA of c.-69G>T homozygotes and total RNA from genome-edited hTERT-RPE1 cells carrying homozygous TMEM216 c.-69G>A. In conclusion, these variants explain a significant proportion of unsolved cases, specifically in individuals of African ancestry, suggesting that reduced TMEM216 expression might lead to abnormal ciliogenesis and photoreceptor degeneration.


Subject(s)
Pedigree , Polymorphism, Single Nucleotide , Retinitis Pigmentosa , Adult , Child , Child, Preschool , Female , Humans , Male , Young Adult , Alleles , Haplotypes , Heterozygote , Homozygote , Membrane Proteins/genetics , Phenotype , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
16.
Nature ; 591(7848): 147-151, 2021 03.
Article in English | MEDLINE | ID: mdl-33505025

ABSTRACT

Many sequence variants have been linked to complex human traits and diseases1, but deciphering their biological functions remains challenging, as most of them reside in noncoding DNA. Here we have systematically assessed the binding of 270 human transcription factors to 95,886 noncoding variants in the human genome using an ultra-high-throughput multiplex protein-DNA binding assay, termed single-nucleotide polymorphism evaluation by systematic evolution of ligands by exponential enrichment (SNP-SELEX). The resulting 828 million measurements of transcription factor-DNA interactions enable estimation of the relative affinity of these transcription factors to each variant in vitro and evaluation of the current methods to predict the effects of noncoding variants on transcription factor binding. We show that the position weight matrices of most transcription factors lack sufficient predictive power, whereas the support vector machine combined with the gapped k-mer representation show much improved performance, when assessed on results from independent SNP-SELEX experiments involving a new set of 61,020 sequence variants. We report highly predictive models for 94 human transcription factors and demonstrate their utility in genome-wide association studies and understanding of the molecular pathways involved in diverse human traits and diseases.


Subject(s)
Polymorphism, Single Nucleotide/genetics , SELEX Aptamer Technique , Support Vector Machine , Transcription Factors/metabolism , Binding Sites/genetics , Disease/genetics , Genome, Human/genetics , Humans , Ligands , Protein Binding
17.
Nature ; 595(7869): 735-740, 2021 07.
Article in English | MEDLINE | ID: mdl-34040254

ABSTRACT

The functional engagement between an enhancer and its target promoter ensures precise gene transcription1. Understanding the basis of promoter choice by enhancers has important implications for health and disease. Here we report that functional loss of a preferred promoter can release its partner enhancer to loop to and activate an alternative promoter (or alternative promoters) in the neighbourhood. We refer to this target-switching process as 'enhancer release and retargeting'. Genetic deletion, motif perturbation or mutation, and dCas9-mediated CTCF tethering reveal that promoter choice by an enhancer can be determined by the binding of CTCF at promoters, in a cohesin-dependent manner-consistent with a model of 'enhancer scanning' inside the contact domain. Promoter-associated CTCF shows a lower affinity than that at chromatin domain boundaries and often lacks a preferred motif orientation or a partnering CTCF at the cognate enhancer, suggesting properties distinct from boundary CTCF. Analyses of cancer mutations, data from the GTEx project and risk loci from genome-wide association studies, together with a focused CRISPR interference screen, reveal that enhancer release and retargeting represents an overlooked mechanism that underlies the activation of disease-susceptibility genes, as exemplified by a risk locus for Parkinson's disease (NUCKS1-RAB7L1) and three loci associated with cancer (CLPTM1L-TERT, ZCCHC7-PAX5 and PVT1-MYC).


Subject(s)
CCCTC-Binding Factor/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Promoter Regions, Genetic , CRISPR-Cas Systems , Cell Cycle Proteins/genetics , Cells, Cultured , Chromatin , Chromosomal Proteins, Non-Histone/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Humans , MCF-7 Cells , Neoplasms/genetics , Neural Stem Cells , Oncogenes , Parkinson Disease/genetics , Cohesins
18.
Proc Natl Acad Sci U S A ; 121(38): e2400781121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39259589

ABSTRACT

During homeostasis, the endoplasmic reticulum (ER) maintains productive transmembrane and secretory protein folding that is vital for proper cellular function. The ER-resident HSP70 chaperone, binding immunoglobulin protein (BiP), plays a pivotal role in sensing ER stress to activate the unfolded protein response (UPR). BiP function is regulated by the bifunctional enzyme filamentation induced by cyclic-AMP domain protein (FicD) that mediates AMPylation and deAMPylation of BiP in response to changes in ER stress. AMPylated BiP acts as a molecular rheostat to regulate UPR signaling, yet little is known about the molecular consequences of FicD loss. In this study, we investigate the role of FicD in mouse embryonic fibroblast (MEF) response to pharmacologically and metabolically induced ER stress. We find differential BiP AMPylation signatures when comparing robust chemical ER stress inducers to physiological glucose starvation stress and recovery. Wildtype MEFs respond to pharmacological ER stress by down-regulating BiP AMPylation. Conversely, BiP AMPylation in wildtype MEFs increases upon metabolic stress induced by glucose starvation. Deletion of FicD results in widespread gene expression changes under baseline growth conditions. In addition, FicD null MEFs exhibit dampened UPR signaling, altered cell stress recovery response, and unconstrained protein secretion. Taken together, our findings indicate that FicD is important for tampering UPR signaling, stress recovery, and the maintenance of secretory protein homeostasis.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Fibroblasts , Glucose , Unfolded Protein Response , Animals , Mice , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Fibroblasts/metabolism , Glucose/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Mice, Knockout , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 121(24): e2315700121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830099

ABSTRACT

Given the importance of climate in shaping species' geographic distributions, climate change poses an existential threat to biodiversity. Climate envelope modeling, the predominant approach used to quantify this threat, presumes that individuals in populations respond to climate variability and change according to species-level responses inferred from spatial occurrence data-such that individuals at the cool edge of a species' distribution should benefit from warming (the "leading edge"), whereas individuals at the warm edge should suffer (the "trailing edge"). Using 1,558 tree-ring time series of an aridland pine (Pinus edulis) collected at 977 locations across the species' distribution, we found that trees everywhere grow less in warmer-than-average and drier-than-average years. Ubiquitous negative temperature sensitivity indicates that individuals across the entire distribution should suffer with warming-the entire distribution is a trailing edge. Species-level responses to spatial climate variation are opposite in sign to individual-scale responses to time-varying climate for approximately half the species' distribution with respect to temperature and the majority of the species' distribution with respect to precipitation. These findings, added to evidence from the literature for scale-dependent climate responses in hundreds of species, suggest that correlative, equilibrium-based range forecasts may fail to accurately represent how individuals in populations will be impacted by changing climate. A scale-dependent view of the impact of climate change on biodiversity highlights the transient risk of extinction hidden inside climate envelope forecasts and the importance of evolution in rescuing species from extinction whenever local climate variability and change exceeds individual-scale climate tolerances.


Subject(s)
Climate Change , Extinction, Biological , Pinus , Pinus/physiology , Trees , Biodiversity , Forecasting/methods , Temperature , Climate Models
20.
Proc Natl Acad Sci U S A ; 121(42): e2408249121, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39388274

ABSTRACT

ATP-grasp superfamily enzymes contain a hand-like ATP-binding fold and catalyze a variety of reactions using a similar catalytic mechanism. More than 30 protein families are categorized in this superfamily, and they are involved in a plethora of cellular processes and human diseases. Here, we identify C12orf29 (RLIG1) as an atypical ATP-grasp enzyme that ligates RNA. Human RLIG1 and its homologs autoadenylate on an active site Lys residue as part of a reaction intermediate that specifically ligates RNA halves containing a 5'-phosphate and a 3'-hydroxyl. RLIG1 binds tRNA in cells and can ligate tRNA within the anticodon loop in vitro. Transcriptomic analyses of Rlig1 knockout mice revealed significant alterations in global tRNA levels in the brains of female mice, but not in those of male mice. Furthermore, crystal structures of a RLIG1 homolog from Yasminevirus bound to nucleotides revealed a minimal and atypical RNA ligase fold with a conserved active site architecture that participates in catalysis. Collectively, our results identify RLIG1 as an RNA ligase and suggest its involvement in tRNA biology.


Subject(s)
Catalytic Domain , Mice, Knockout , RNA Ligase (ATP) , RNA, Transfer , Animals , RNA, Transfer/metabolism , RNA, Transfer/genetics , Mice , RNA Ligase (ATP)/metabolism , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/chemistry , Humans , Female , Male , Crystallography, X-Ray , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL