Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Nat Immunol ; 25(7): 1257-1269, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806707

ABSTRACT

The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.


Subject(s)
B7-H1 Antigen , Circadian Clocks , Immune Checkpoint Inhibitors , Myeloid-Derived Suppressor Cells , Animals , Mice , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Circadian Clocks/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Mice, Inbred C57BL , Circadian Rhythm/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/drug therapy , Tumor Microenvironment/immunology , Immune Tolerance , Humans , Female , Cell Line, Tumor , Single-Cell Analysis , Immunosuppression Therapy , Cytokines/metabolism , Male
2.
Cell ; 184(22): 5497-5500, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34587491

ABSTRACT

The scientific and clinical communities have both experienced several harsh lessons on clinical care management and drug development during the COVID-19 pandemic. Here, we discuss several key lessons learned and describe a framework within which our two communities can work together and invest in to improve future pandemic responses.


Subject(s)
COVID-19 Drug Treatment , Drug Development , Pandemics/prevention & control , Humans , Pharmaceutical Preparations
3.
Cell ; 182(1): 24-37, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32649876

ABSTRACT

Viral genomes encode transcriptional regulators that alter the expression of viral and host genes. Despite an emerging role in human diseases, a thorough annotation of human viral transcriptional regulators (vTRs) is currently lacking, limiting our understanding of their molecular features and functions. Here, we provide a comprehensive catalog of 419 vTRs belonging to 20 different virus families. Using this catalog, we characterize shared and unique cellular genes, proteins, and pathways targeted by particular vTRs and discuss the role of vTRs in human disease pathogenesis. Our study provides a unique and valuable resource for the fields of virology, genomics, and human disease genetics.


Subject(s)
Transcription, Genetic , Viral Proteins/metabolism , Epigenesis, Genetic , Humans , Models, Biological , Protein Interaction Maps , Viral Proteins/chemistry , Viral Proteins/genetics
4.
Cell ; 174(6): 1522-1536.e22, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146161

ABSTRACT

How transcription affects genome 3D organization is not well understood. We found that during influenza A (IAV) infection, rampant transcription rapidly reorganizes host cell chromatin interactions. These changes occur at the ends of highly transcribed genes, where global inhibition of transcription termination by IAV NS1 protein causes readthrough transcription for hundreds of kilobases. In these readthrough regions, elongating RNA polymerase II disrupts chromatin interactions by inducing cohesin displacement from CTCF sites, leading to locus decompaction. Readthrough transcription into heterochromatin regions switches them from the inert (B) to the permissive (A) chromatin compartment and enables transcription factor binding. Data from non-viral transcription stimuli show that transcription similarly affects cohesin-mediated chromatin contacts within gene bodies. Conversely, inhibition of transcription elongation allows cohesin to accumulate at previously transcribed intragenic CTCF sites and to mediate chromatin looping and compaction. Our data indicate that transcription elongation by RNA polymerase II remodels genome 3D architecture.


Subject(s)
Chromatin/metabolism , Genome, Human , Influenza A Virus, H5N1 Subtype/metabolism , Binding Sites , CCCTC-Binding Factor/chemistry , CCCTC-Binding Factor/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Flavonoids/pharmacology , Humans , Interferon-beta/pharmacology , Macrophages/cytology , Macrophages/metabolism , Macrophages/virology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Piperidines/pharmacology , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Cohesins
5.
Cell ; 175(7): 1931-1945.e18, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30550790

ABSTRACT

Mosquito-borne flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), are a growing public health concern. Systems-level analysis of how flaviviruses hijack cellular processes through virus-host protein-protein interactions (PPIs) provides information about their replication and pathogenic mechanisms. We used affinity purification-mass spectrometry (AP-MS) to compare flavivirus-host interactions for two viruses (DENV and ZIKV) in two hosts (human and mosquito). Conserved virus-host PPIs revealed that the flavivirus NS5 protein suppresses interferon stimulated genes by inhibiting recruitment of the transcription complex PAF1C and that chemical modulation of SEC61 inhibits DENV and ZIKV replication in human and mosquito cells. Finally, we identified a ZIKV-specific interaction between NS4A and ANKLE2, a gene linked to hereditary microcephaly, and showed that ZIKV NS4A causes microcephaly in Drosophila in an ANKLE2-dependent manner. Thus, comparative flavivirus-host PPI mapping provides biological insights and, when coupled with in vivo models, can be used to unravel pathogenic mechanisms.


Subject(s)
Dengue Virus , Dengue , Membrane Proteins , Nuclear Proteins , Viral Nonstructural Proteins , Zika Virus Infection , Zika Virus , Animals , Cell Line, Tumor , Culicidae , Dengue/genetics , Dengue/metabolism , Dengue/pathology , Dengue Virus/genetics , Dengue Virus/metabolism , Dengue Virus/pathogenicity , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Interaction Mapping , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Zika Virus/genetics , Zika Virus/metabolism , Zika Virus/pathogenicity , Zika Virus Infection/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
6.
Cell ; 169(4): 679-692.e14, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475896

ABSTRACT

The nuclear RNA exosome is an essential multi-subunit complex that controls RNA homeostasis. Congenital mutations in RNA exosome genes are associated with neurodegenerative diseases. Little is known about the role of the RNA exosome in the cellular response to pathogens. Here, using NGS and human and mouse genetics, we show that influenza A virus (IAV) ribogenesis and growth are suppressed by impaired RNA exosome activity. Mechanistically, the nuclear RNA exosome coordinates the initial steps of viral transcription with RNAPII at host promoters. The viral polymerase complex co-opts the nuclear RNA exosome complex and cellular RNAs en route to 3' end degradation. Exosome deficiency uncouples chromatin targeting of the viral polymerase complex and the formation of cellular:viral RNA hybrids, which are essential RNA intermediates that license transcription of antisense genomic viral RNAs. Our results suggest that evolutionary arms races have shaped the cellular RNA quality control machinery.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/virology , RNA Polymerase II/metabolism , A549 Cells , Animals , Chromatin Immunoprecipitation , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Humans , Mass Spectrometry , Mice , Mutation , Neurodegenerative Diseases/virology , RNA-Binding Proteins/genetics , Ribosomes/genetics , Transcription, Genetic
7.
Mol Cell ; 83(23): 4255-4271.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37995687

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancient parasitic infections and comprise sizable portions of most genomes. Although epigenetic mechanisms silence most ERVs by generating a repressive environment that prevents their expression (heterochromatin), little is known about mechanisms silencing ERVs residing in open regions of the genome (euchromatin). This is particularly important during embryonic development, where induction and repression of distinct classes of ERVs occur in short temporal windows. Here, we demonstrate that transcription-associated RNA degradation by the nuclear RNA exosome and Integrator is a regulatory mechanism that controls the productive transcription of most genes and many ERVs involved in preimplantation development. Disrupting nuclear RNA catabolism promotes dedifferentiation to a totipotent-like state characterized by defects in RNAPII elongation and decreased expression of long genes (gene-length asymmetry). Our results indicate that RNA catabolism is a core regulatory module of gene networks that safeguards RNAPII activity, ERV expression, cell identity, and developmental potency.


Subject(s)
Endogenous Retroviruses , Endogenous Retroviruses/genetics , RNA, Nuclear , Epigenesis, Genetic , Heterochromatin , Gene Expression
8.
Nat Rev Mol Cell Biol ; 19(4): 245-261, 2018 04.
Article in English | MEDLINE | ID: mdl-29184195

ABSTRACT

Multiple cell-signalling pathways converge on chromatin to induce gene expression programmes. The inducible transcriptional programmes that are established as a result of inflammatory or oncogenic signals are controlled by shared chromatin regulators. Therapeutic targeting of such chromatin dependencies has proved effective for controlling tumorigenesis and for preventing immunopathologies that are driven by overt inflammation. In this Review, we discuss how chromatin dependencies are established to regulate the expression of key oncogenes and inflammation-promoting genes and how a better mechanistic understanding of such chromatin dependencies can be leveraged to improve the magnitude, timing, duration and selectivity of cell responses with the aim of minimizing unwanted cellular and systemic effects. Recently, exciting progress has been made in cancer immunotherapy and in the development of drugs that target chromatin regulators. We discuss recent advances in clinical trials and the challenge of combining immune-cell-based therapies and epigenetic therapies to improve human health.


Subject(s)
Chromatin/genetics , Inflammation/genetics , Neoplasms/genetics , Animals , Carcinogenesis/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , Epigenesis, Genetic , Gene Expression/drug effects , Genetic Therapy , Humans , Inflammation/metabolism , Models, Genetic , Neoplasms/metabolism , Neoplasms/therapy , Signal Transduction/genetics , Transcription Factors/metabolism
9.
Mol Cell ; 81(17): 3447-3448, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34478653

ABSTRACT

Valencia-Sánchez et al. (2021) and Liu et al. (2021) provide structural and biological insights about the existence and importance of a nucleosome-like particle in a family of giant viruses.


Subject(s)
Giant Viruses , Viruses , Genome , Giant Viruses/genetics , Nucleosomes/genetics
10.
Nature ; 606(7916): 945-952, 2022 06.
Article in English | MEDLINE | ID: mdl-35732742

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.


Subject(s)
Amyotrophic Lateral Sclerosis , CD8-Positive T-Lymphocytes , Clone Cells , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clone Cells/pathology , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Knock-In Techniques , Mice , Motor Neurons/pathology , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism
11.
Mol Cell ; 78(2): 197-209.e7, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32084337

ABSTRACT

We have developed a platform for quantitative genetic interaction mapping using viral infectivity as a functional readout and constructed a viral host-dependency epistasis map (vE-MAP) of 356 human genes linked to HIV function, comprising >63,000 pairwise genetic perturbations. The vE-MAP provides an expansive view of the genetic dependencies underlying HIV infection and can be used to identify drug targets and study viral mutations. We found that the RNA deadenylase complex, CNOT, is a central player in the vE-MAP and show that knockout of CNOT1, 10, and 11 suppressed HIV infection in primary T cells by upregulating innate immunity pathways. This phenotype was rescued by deletion of IRF7, a transcription factor regulating interferon-stimulated genes, revealing a previously unrecognized host signaling pathway involved in HIV infection. The vE-MAP represents a generic platform that can be used to study the global effects of how different pathogens hijack and rewire the host during infection.


Subject(s)
Epistasis, Genetic , HIV Infections/genetics , Interferon Regulatory Factor-7/genetics , Transcription Factors/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Interferons/genetics , Mutation , Signal Transduction/genetics
12.
Nat Immunol ; 16(5): 485-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25822250

ABSTRACT

The human helicase senataxin (SETX) has been linked to the neurodegenerative diseases amyotrophic lateral sclerosis (ALS4) and ataxia with oculomotor apraxia (AOA2). Here we identified a role for SETX in controlling the antiviral response. Cells that had undergone depletion of SETX and SETX-deficient cells derived from patients with AOA2 had higher expression of antiviral mediators in response to infection than did wild-type cells. Mechanistically, we propose a model whereby SETX attenuates the activity of RNA polymerase II (RNAPII) at genes stimulated after a virus is sensed and thus controls the magnitude of the host response to pathogens and the biogenesis of various RNA viruses (e.g., influenza A virus and West Nile virus). Our data indicate a potentially causal link among inborn errors in SETX, susceptibility to infection and the development of neurologic disorders.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Influenza, Human/immunology , Orthomyxoviridae/physiology , RNA Helicases/metabolism , RNA Polymerase II/metabolism , Spinocerebellar Degenerations/genetics , West Nile Fever/immunology , West Nile virus/physiology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Cytokines/metabolism , DNA Helicases , Dogs , Down-Regulation , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Microarray Analysis , Multifunctional Enzymes , RNA Helicases/genetics , RNA Polymerase II/genetics , RNA, Small Interfering/genetics , Spinocerebellar Ataxias/congenital , Vero Cells , Virus Replication/genetics
13.
Immunity ; 49(5): 819-828.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413362

ABSTRACT

Inducing graft acceptance without chronic immunosuppression remains an elusive goal in organ transplantation. Using an experimental transplantation mouse model, we demonstrate that local macrophage activation through dectin-1 and toll-like receptor 4 (TLR4) drives trained immunity-associated cytokine production during allograft rejection. We conducted nanoimmunotherapeutic studies and found that a short-term mTOR-specific high-density lipoprotein (HDL) nanobiologic treatment (mTORi-HDL) averted macrophage aerobic glycolysis and the epigenetic modifications underlying inflammatory cytokine production. The resulting regulatory macrophages prevented alloreactive CD8+ T cell-mediated immunity and promoted tolerogenic CD4+ regulatory T (Treg) cell expansion. To enhance therapeutic efficacy, we complemented the mTORi-HDL treatment with a CD40-TRAF6-specific nanobiologic (TRAF6i-HDL) that inhibits co-stimulation. This synergistic nanoimmunotherapy resulted in indefinite allograft survival. Together, we show that HDL-based nanoimmunotherapy can be employed to control macrophage function in vivo. Our strategy, focused on preventing inflammatory innate immune responses, provides a framework for developing targeted therapies that promote immunological tolerance.


Subject(s)
Graft Survival/immunology , Immunosuppression Therapy , Inflammation/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Organ Transplantation , Allografts , Animals , Biomarkers , HMGB1 Protein/genetics , Immune Tolerance , Immunity, Innate , Immunologic Memory , Macrophages/immunology , Macrophages/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism , Vimentin/genetics
14.
Nature ; 593(7859): 362-371, 2021 05.
Article in English | MEDLINE | ID: mdl-34012080

ABSTRACT

Unlike the human genome that comprises mostly noncoding and regulatory sequences, viruses have evolved under the constraints of maintaining a small genome size while expanding the efficiency of their coding and regulatory sequences. As a result, viruses use strategies of transcription and translation in which one or more of the steps in the conventional gene-protein production line are altered. These alternative strategies of viral gene expression (also known as gene recoding) can be uniquely brought about by dedicated viral enzymes or by co-opting host factors (known as host dependencies). Targeting these unique enzymatic activities and host factors exposes vulnerabilities of a virus and provides a paradigm for the design of novel antiviral therapies. In this Review, we describe the types and mechanisms of unconventional gene and protein expression in viruses, and provide a perspective on how future basic mechanistic work could inform translational efforts that are aimed at viral eradication.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Gene Expression Regulation, Viral/drug effects , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Virus Diseases/drug therapy , Virus Diseases/virology , Animals , Frameshifting, Ribosomal/drug effects , Frameshifting, Ribosomal/genetics , Gene Expression Regulation, Viral/genetics , Genome, Viral/drug effects , Genome, Viral/genetics , Humans , RNA Splicing/drug effects , RNA Splicing/genetics
15.
Immunity ; 44(1): 46-58, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26789921

ABSTRACT

Viruses are obligate parasites and thus require the machinery of the host cell to replicate. Inhibition of host factors co-opted during active infection is a strategy hosts use to suppress viral replication and a potential pan-antiviral therapy. To define the cellular proteins and processes required for a virus during infection is thus crucial to understanding the mechanisms of virally induced disease. In this report, we generated fully infectious tagged influenza viruses and used infection-based proteomics to identify pivotal arms of cellular signaling required for influenza virus growth and infectivity. Using mathematical modeling and genetic and pharmacologic approaches, we revealed that modulation of Sec61-mediated cotranslational translocation selectively impaired glycoprotein proteostasis of influenza as well as HIV and dengue viruses and led to inhibition of viral growth and infectivity. Thus, by studying virus-human protein-protein interactions in the context of active replication, we have identified targetable host factors for broad-spectrum antiviral therapies.


Subject(s)
Host-Parasite Interactions/physiology , Influenza A virus/physiology , Influenza A virus/pathogenicity , Models, Theoretical , Virus Replication/physiology , Dengue Virus/pathogenicity , Dengue Virus/physiology , HIV/pathogenicity , HIV/physiology , Humans , Immunoprecipitation , Mass Spectrometry , Protein Folding , Proteomics
17.
Nature ; 483(7390): 428-33, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22419161

ABSTRACT

Viral infection is commonly associated with virus-driven hijacking of host proteins. Here we describe a novel mechanism by which influenza virus affects host cells through the interaction of influenza non-structural protein 1 (NS1) with the infected cell epigenome. We show that the NS1 protein of influenza A H3N2 subtype possesses a histone-like sequence (histone mimic) that is used by the virus to target the human PAF1 transcription elongation complex (hPAF1C). We demonstrate that binding of NS1 to hPAF1C depends on the NS1 histone mimic and results in suppression of hPAF1C-mediated transcriptional elongation. Furthermore, human PAF1 has a crucial role in the antiviral response. Loss of hPAF1C binding by NS1 attenuates influenza infection, whereas hPAF1C deficiency reduces antiviral gene expression and renders cells more susceptible to viruses. We propose that the histone mimic in NS1 enables the influenza virus to affect inducible gene expression selectively, thus contributing to suppression of the antiviral response.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Influenza, Human/genetics , Influenza, Human/immunology , Molecular Mimicry , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Gene Expression Regulation/immunology , Histones/chemistry , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/pathology , Influenza, Human/virology , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors , Transcription, Genetic/immunology , Viral Nonstructural Proteins/chemistry
18.
Nature ; 468(7327): 1119-23, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21068722

ABSTRACT

Interaction of pathogens with cells of the immune system results in activation of inflammatory gene expression. This response, although vital for immune defence, is frequently deleterious to the host due to the exaggerated production of inflammatory proteins. The scope of inflammatory responses reflects the activation state of signalling proteins upstream of inflammatory genes as well as signal-induced assembly of nuclear chromatin complexes that support mRNA expression. Recognition of post-translationally modified histones by nuclear proteins that initiate mRNA transcription and support mRNA elongation is a critical step in the regulation of gene expression. Here we present a novel pharmacological approach that targets inflammatory gene expression by interfering with the recognition of acetylated histones by the bromodomain and extra terminal domain (BET) family of proteins. We describe a synthetic compound (I-BET) that by 'mimicking' acetylated histones disrupts chromatin complexes responsible for the expression of key inflammatory genes in activated macrophages, and confers protection against lipopolysaccharide-induced endotoxic shock and bacteria-induced sepsis. Our findings suggest that synthetic compounds specifically targeting proteins that recognize post-translationally modified histones can serve as a new generation of immunomodulatory drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammation , Macrophages/drug effects , Acetylation/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Benzodiazepines , Cells, Cultured , Epigenomics , Genome-Wide Association Study , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inflammation/drug therapy , Inflammation/prevention & control , Kaplan-Meier Estimate , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Salmonella Infections/drug therapy , Salmonella Infections/immunology , Salmonella Infections/physiopathology , Salmonella Infections/prevention & control , Salmonella typhimurium , Sepsis/drug therapy , Sepsis/prevention & control , Shock, Septic/drug therapy , Shock, Septic/prevention & control
19.
Sci Adv ; 10(23): eadm9589, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838142

ABSTRACT

DNA replication stress (RS) is a widespread phenomenon in carcinogenesis, causing genomic instability and extensive chromatin alterations. DNA damage leads to activation of innate immune signaling, but little is known about transcriptional regulators mediating such signaling upon RS. Using a chemical screen, we identified protein arginine methyltransferase 5 (PRMT5) as a key mediator of RS-dependent induction of interferon-stimulated genes (ISGs). This response is also associated with reactivation of endogenous retroviruses (ERVs). Using quantitative mass spectrometry, we identify proteins with PRMT5-dependent symmetric dimethylarginine (SDMA) modification induced upon RS. Among these, we show that PRMT5 targets and modulates the activity of ZNF326, a zinc finger protein essential for ISG response. Our data demonstrate a role for PRMT5-mediated SDMA in the context of RS-induced transcriptional induction, affecting physiological homeostasis and cancer therapy.


Subject(s)
DNA Replication , Immunity, Innate , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Signal Transduction , Arginine/metabolism , Arginine/analogs & derivatives , Stress, Physiological , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Damage , Transcription Factors/metabolism , Transcription Factors/genetics
20.
Elife ; 122024 Jan 15.
Article in English | MEDLINE | ID: mdl-38224289

ABSTRACT

Inter-organ communication is a vital process to maintain physiologic homeostasis, and its dysregulation contributes to many human diseases. Given that circulating bioactive factors are stable in serum, occur naturally, and are easily assayed from blood, they present obvious focal molecules for therapeutic intervention and biomarker development. Recently, studies have shown that secreted proteins mediating inter-tissue signaling could be identified by 'brute force' surveys of all genes within RNA-sequencing measures across tissues within a population. Expanding on this intuition, we reasoned that parallel strategies could be used to understand how individual genes mediate signaling across metabolic tissues through correlative analyses of gene variation between individuals. Thus, comparison of quantitative levels of gene expression relationships between organs in a population could aid in understanding cross-organ signaling. Here, we surveyed gene-gene correlation structure across 18 metabolic tissues in 310 human individuals and 7 tissues in 103 diverse strains of mice fed a normal chow or high-fat/high-sucrose (HFHS) diet. Variation of genes such as FGF21, ADIPOQ, GCG, and IL6 showed enrichments which recapitulate experimental observations. Further, similar analyses were applied to explore both within-tissue signaling mechanisms (liver PCSK9) and genes encoding enzymes producing metabolites (adipose PNPLA2), where inter-individual correlation structure aligned with known roles for these critical metabolic pathways. Examination of sex hormone receptor correlations in mice highlighted the difference of tissue-specific variation in relationships with metabolic traits. We refer to this resource as gene-derived correlations across tissues (GD-CAT) where all tools and data are built into a web portal enabling users to perform these analyses without a single line of code (gdcat.org). This resource enables querying of any gene in any tissue to find correlated patterns of genes, cell types, pathways, and network architectures across metabolic organs.


Subject(s)
Proprotein Convertase 9 , Signal Transduction , Humans , Animals , Mice , Homeostasis , Adiposity
SELECTION OF CITATIONS
SEARCH DETAIL