Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.429
Filter
Add more filters

Publication year range
1.
Mol Cell ; 83(4): 574-588.e11, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36731470

ABSTRACT

Most eukaryotic promoter regions are divergently transcribed. As the RNA polymerase II pre-initiation complex (PIC) is intrinsically asymmetric and responsible for transcription in a single direction, it is unknown how divergent transcription arises. Here, the Saccharomyces cerevisiae Mediator complexed with a PIC (Med-PIC) was assembled on a divergent promoter and analyzed by cryoelectron microscopy. The structure reveals two distinct Med-PICs forming a dimer through the Mediator tail module, induced by a homodimeric activator protein localized near the dimerization interface. The tail dimer is associated with ∼80-bp upstream DNA, such that two flanking core promoter regions are positioned and oriented in a suitable form for PIC assembly in opposite directions. Also, cryoelectron tomography visualized the progress of the PIC assembly on the two core promoter regions, providing direct evidence for the role of the Med-PIC dimer in divergent transcription.


Subject(s)
RNA Polymerase II , Saccharomyces cerevisiae Proteins , RNA Polymerase II/metabolism , Cryoelectron Microscopy , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Mediator Complex/genetics , Transcription Initiation, Genetic
2.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751740

ABSTRACT

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Subject(s)
Osteoclasts , RNA , Humans , Mice , Animals , Co-Repressor Proteins/genetics , Osteoclasts/metabolism , RANK Ligand/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Gene Expression
3.
Mol Cell ; 82(3): 677-695.e7, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35016035

ABSTRACT

Transcription is orchestrated by thousands of transcription factors (TFs) and chromatin-associated proteins, but how these are causally connected to transcriptional activation is poorly understood. Here, we conduct an unbiased proteome-scale screen to systematically uncover human proteins that activate transcription in a natural chromatin context. By combining interaction proteomics and chemical inhibitors, we delineate the preference of these transcriptional activators for specific co-activators, highlighting how even closely related TFs can function via distinct cofactors. We also identify potent transactivation domains among the hits and use AlphaFold2 to predict and experimentally validate interaction interfaces of two activation domains with BRD4. Finally, we show that many novel activators are partners in fusion events in tumors and functionally characterize a myofibroma-associated fusion between SRF and C3orf62, a potent p300-dependent activator. Our work provides a functional catalog of potent transactivators in the human proteome and a platform for discovering transcriptional regulators at genome scale.


Subject(s)
Proteome , Proteomics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , K562 Cells , Mice , Myofibroma/genetics , Myofibroma/metabolism , NIH 3T3 Cells , Serum Response Factor/genetics , Serum Response Factor/metabolism , Transcription Factors/genetics
4.
Mol Cell ; 82(9): 1643-1659.e10, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35334231

ABSTRACT

The NADase SARM1 (sterile alpha and TIR motif containing 1) is a key executioner of axon degeneration and a therapeutic target for several neurodegenerative conditions. We show that a potent SARM1 inhibitor undergoes base exchange with the nicotinamide moiety of nicotinamide adenine dinucleotide (NAD+) to produce the bona fide inhibitor 1AD. We report structures of SARM1 in complex with 1AD, NAD+ mimetics and the allosteric activator nicotinamide mononucleotide (NMN). NMN binding triggers reorientation of the armadillo repeat (ARM) domains, which disrupts ARM:TIR interactions and leads to formation of a two-stranded TIR domain assembly. The active site spans two molecules in these assemblies, explaining the requirement of TIR domain self-association for NADase activity and axon degeneration. Our results reveal the mechanisms of SARM1 activation and substrate binding, providing rational avenues for the design of new therapeutics targeting SARM1.


Subject(s)
Armadillo Domain Proteins , NAD , Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , NAD/metabolism , NAD+ Nucleosidase/metabolism , Protein Domains
5.
Genes Dev ; 36(9-10): 618-633, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35618312

ABSTRACT

DNA damage repair systems are critical for genomic integrity. However, they must be coordinated with DNA replication and cell division to ensure accurate genomic transmission. In most bacteria, this coordination is mediated by the SOS response through LexA, which triggers a halt in cell division until repair is completed. Recently, an SOS-independent damage response system was revealed in Caulobacter crescentus. This pathway is controlled by the transcription activator, DriD, but how DriD senses and signals DNA damage is unknown. To address this question, we performed biochemical, cellular, and structural studies. We show that DriD binds a specific promoter DNA site via its N-terminal HTH domain to activate transcription of genes, including the cell division inhibitor didA A structure of the C-terminal portion of DriD revealed a WYL motif domain linked to a WCX dimerization domain. Strikingly, we found that DriD binds ssDNA between the WYL and WCX domains. Comparison of apo and ssDNA-bound DriD structures reveals that ssDNA binding orders and orients the DriD domains, indicating a mechanism for ssDNA-mediated operator DNA binding activation. Biochemical and in vivo studies support the structural model. Our data thus reveal the molecular mechanism underpinning an SOS-independent DNA damage repair pathway.


Subject(s)
Bacterial Proteins , Caulobacter crescentus , Bacterial Proteins/metabolism , Caulobacter crescentus/genetics , Caulobacter crescentus/metabolism , DNA Damage , DNA, Single-Stranded/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Immunity ; 53(4): 745-758.e4, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33010223

ABSTRACT

Innate immune responses rely on rapid and precise gene regulation mediated by accessibility of regulatory regions to transcription factors (TFs). In natural killer (NK) cells and other innate lymphoid cells, competent enhancers are primed during lineage acquisition, and formation of de novo enhancers characterizes the acquisition of innate memory in activated NK cells and macrophages. Here, we investigated how primed and de novo enhancers coordinate to facilitate high-magnitude gene induction during acute activation. Epigenomic and transcriptomic analyses of regions near highly induced genes (HIGs) in NK cells both in vitro and in a model of Toxoplasma gondii infection revealed de novo chromatin accessibility and enhancer remodeling controlled by signal-regulated TFs STATs. Acute NK cell activation redeployed the lineage-determining TF T-bet to de novo enhancers, independent of DNA-sequence-specific motif recognition. Thus, acute stimulation reshapes enhancer function through the combinatorial usage and repurposing of both lineage-determining and signal-regulated TFs to ensure an effective response.


Subject(s)
Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Killer Cells, Natural/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology
7.
Mol Cell ; 81(8): 1781-1788.e4, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33571424

ABSTRACT

Mediator is a universal adaptor for transcription control. It serves as an interface between gene-specific activator or repressor proteins and the general RNA polymerase II (pol II) transcription machinery. Previous structural studies revealed a relatively small part of Mediator and none of the gene activator-binding regions. We have determined the cryo-EM structure of the Mediator at near-atomic resolution. The structure reveals almost all amino acid residues in ordered regions, including the major targets of activator proteins, the Tail module, and the Med1 subunit of the Middle module. Comparison of Mediator structures with and without pol II reveals conformational changes that propagate across the entire Mediator, from Head to Tail, coupling activator- and pol II-interacting regions.


Subject(s)
Mediator Complex Subunit 1/metabolism , Amino Acids/genetics , Protein Conformation , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
8.
Annu Rev Microbiol ; 77: 317-339, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37285554

ABSTRACT

LysR-type transcriptional regulators (LTTRs) form one of the largest families of bacterial regulators. They are widely distributed and contribute to all aspects of metabolism and physiology. Most are homotetramers, with each subunit composed of an N-terminal DNA-binding domain followed by a long helix connecting to an effector-binding domain. LTTRs typically bind DNA in the presence or absence of a small-molecule ligand (effector). In response to cellular signals, conformational changes alter DNA interactions, contact with RNA polymerase, and sometimes contact with other proteins. Many are dual-function repressor-activators, although different modes of regulation may occur at multiple promoters. This review presents an update on the molecular basis of regulation, the complexity of regulatory schemes, and applications in biotechnology and medicine. The abundance of LTTRs reflects their versatility and importance. While a single regulatory model cannot describe all family members, a comparison of similarities and differences provides a framework for future study.


Subject(s)
Bacterial Proteins , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Bacterial Proteins/metabolism , Bacteria/genetics , Bacteria/metabolism , DNA/chemistry , Protein Binding
9.
Mol Cell ; 78(5): 890-902.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32416068

ABSTRACT

Acidic transcription activation domains (ADs) are encoded by a wide range of seemingly unrelated amino acid sequences, making it difficult to recognize features that promote their dynamic behavior, "fuzzy" interactions, and target specificity. We screened a large set of random 30-mer peptides for AD function in yeast and trained a deep neural network (ADpred) on the AD-positive and -negative sequences. ADpred identifies known acidic ADs within transcription factors and accurately predicts the consequences of mutations. Our work reveals that strong acidic ADs contain multiple clusters of hydrophobic residues near acidic side chains, explaining why ADs often have a biased amino acid composition. ADs likely use a binding mechanism similar to avidity where a minimum number of weak dynamic interactions are required between activator and target to generate biologically relevant affinity and in vivo function. This mechanism explains the basis for fuzzy binding observed between acidic ADs and targets.


Subject(s)
High-Throughput Screening Assays/methods , Transcription Factors/genetics , Transcriptional Activation/genetics , Amino Acid Sequence/genetics , Basic-Leucine Zipper Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Deep Learning , Protein Binding , Protein Domains/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology
10.
Proc Natl Acad Sci U S A ; 121(24): e2316892121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833472

ABSTRACT

The loss of function of AAA (ATPases associated with diverse cellular activities) mechanoenzymes has been linked to diseases, and small molecules that activate these proteins can be powerful tools to probe mechanisms and test therapeutic hypotheses. Unlike chemical inhibitors that can bind a single conformational state to block enzyme function, activator binding must be permissive to different conformational states needed for mechanochemistry. However, we do not know how AAA proteins can be activated by small molecules. Here, we focus on valosin-containing protein (VCP)/p97, an AAA unfoldase whose loss of function has been linked to protein aggregation-based disorders, to identify druggable sites for chemical activators. We identified VCP ATPase Activator 1 (VAA1), a compound that dose-dependently stimulates VCP ATPase activity up to ~threefold. Our cryo-EM studies resulted in structures (ranging from ~2.9 to 3.7 Å-resolution) of VCP in apo and ADP-bound states and revealed that VAA1 binds an allosteric pocket near the C-terminus in both states. Engineered mutations in the VAA1-binding site confer resistance to VAA1, and furthermore, modulate VCP activity. Mutation of a phenylalanine residue in the VCP C-terminal tail that can occupy the VAA1 binding site also stimulates ATPase activity, suggesting that VAA1 acts by mimicking this interaction. Together, our findings uncover a druggable allosteric site and a mechanism of enzyme regulation that can be tuned through small molecule mimicry.


Subject(s)
Valosin Containing Protein , Valosin Containing Protein/metabolism , Valosin Containing Protein/chemistry , Valosin Containing Protein/genetics , Allosteric Regulation , Humans , Protein Binding , Molecular Mimicry , Cryoelectron Microscopy , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Binding Sites , Allosteric Site , Models, Molecular , Protein Conformation
11.
J Cell Sci ; 137(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38264908

ABSTRACT

Activator of G-protein signaling 3 (AGS3; also known as GPSM1), a receptor-independent activator of G-protein signaling, oscillates among defined subcellular compartments and biomolecular condensates (BMCs) in a regulated manner that is likely related to the functional diversity of the protein. We determined the influence of cell stress on the cellular distribution of AGS3 and core material properties of AGS3 BMCs. Cellular stress (oxidative, pHi and thermal) induced the formation of AGS3 BMCs in HeLa and COS-7 cells, as determined by fluorescent microscopy. Oxidative stress-induced AGS3 BMCs were distinct from G3BP1 stress granules and from RNA processing BMCs defined by the P-body protein Dcp1a. Immunoblots indicated that cellular stress shifted AGS3, but not the stress granule protein G3BP1 to a membrane pellet fraction following cell lysis. The stress-induced generation of AGS3 BMCs was reduced by co-expression of the signaling protein Gαi3, but not the AGS3-binding partner DVL2. Fluorescent recovery following photobleaching of individual AGS3 BMCs indicated that there are distinct diffusion kinetics and restricted fluidity for AGS3 BMCs. These data suggest that AGS3 BMCs represent a distinct class of stress granules that serve as a previously unrecognized signal processing node.


Subject(s)
Biomolecular Condensates , Carrier Proteins , Carrier Proteins/metabolism , DNA Helicases , GTP-Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/metabolism , RNA Recognition Motif Proteins , Humans , Animals
12.
Immunity ; 46(6): 983-991.e4, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28623086

ABSTRACT

Host defense requires the specification of CD4+ helper T (Th) cells into distinct fates, including Th1 cells that preferentially produce interferon-γ (IFN-γ). IFN-γ, a member of a large family of anti-pathogenic and anti-tumor IFNs, induces T-bet, a lineage-defining transcription factor for Th1 cells, which in turn supports IFN-γ production in a feed-forward manner. Herein, we show that a cell-intrinsic role of T-bet influences how T cells perceive their secreted product in the environment. In the absence of T-bet, IFN-γ aberrantly induced a type I IFN transcriptomic program. T-bet preferentially repressed genes and pathways ordinarily activated by type I IFNs to ensure that its transcriptional response did not evoke an aberrant amplification of type I IFN signaling circuitry, otherwise triggered by its own product. Thus, in addition to promoting Th1 effector commitment, T-bet acts as a repressor in differentiated Th1 cells to prevent abberant autocrine type I IFN and downstream signaling.


Subject(s)
Autocrine Communication , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/microbiology , Th1 Cells/virology , Transcriptome
13.
Immunity ; 46(2): 220-232, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228280

ABSTRACT

Fibroblasts are major contributors to and regulators of inflammation and dominant producers of interleukin-6 (IL-6) in inflammatory diseases like rheumatoid arthritis. Yet, compared to leukocytes, the regulation of inflammatory pathways in fibroblasts is largely unknown. Here, we report that analyses of genes coordinately upregulated with IL-6 pointed to STAT4 and leukemia inhibitory factor (LIF) as potentially linked. Gene silencing revealed that STAT4 was required for IL-6 transcription. STAT4 was recruited to the IL-6 promoter after fibroblast activation, and LIF receptor (LIFR) and STAT4 formed a molecular complex that, together with JAK1 and TYK2 kinases, controlled STAT4 activation. Importantly, a positive feedback loop involving autocrine LIF, LIFR, and STAT4 drove sustained IL-6 transcription. Besides IL-6, this autorine loop also drove the production of other key inflammatory factors including IL-8, granulocyte-colony stimulating factor (G-CSF), IL-33, IL-11, IL-1α, and IL-1ß. These findings define the transcriptional regulation of fibroblast-mediated inflammation as distinct from leukocytes.


Subject(s)
Autocrine Communication/immunology , Fibroblasts/immunology , Gene Expression Regulation/immunology , Leukemia Inhibitory Factor/immunology , Receptors, OSM-LIF/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Inflammation/immunology , Interleukin-6/immunology , STAT4 Transcription Factor/immunology , Synovial Membrane/immunology , Transcriptome
14.
Circ Res ; 134(8): 954-969, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38501247

ABSTRACT

BACKGROUND: Acute ischemic stroke triggers endothelial activation that disrupts vascular integrity and increases hemorrhagic transformation leading to worsened stroke outcomes. rt-PA (recombinant tissue-type plasminogen activator) is an effective treatment; however, its use is limited due to a restricted time window and hemorrhagic transformation risk, which in part may involve activation of MMPs (matrix metalloproteinases) mediated through LOX-1 (lectin-like oxLDL [oxidized low-density lipoprotein] receptor 1). This study's overall aim was to evaluate the therapeutic potential of novel MMP-9 (matrix metalloproteinase 9) ± LOX-1 inhibitors in combination with rt-PA to improve stroke outcomes. METHODS: A rat thromboembolic stroke model was utilized to investigate the impact of rt-PA delivered 4 hours poststroke onset as well as selective MMP-9 (JNJ0966) ±LOX-1 (BI-0115) inhibitors given before rt-PA administration. Infarct size, perfusion, and hemorrhagic transformation were evaluated by 9.4-T magnetic resonance imaging, vascular and parenchymal MMP-9 activity via zymography, and neurological function was assessed using sensorimotor function testing. Human brain microvascular endothelial cells were exposed to hypoxia plus glucose deprivation/reperfusion (hypoxia plus glucose deprivation 3 hours/R 24 hours) and treated with ±tPA and ±MMP-9 ±LOX-1 inhibitors. Barrier function was assessed via transendothelial electrical resistance, MMP-9 activity was determined with zymography, and LOX-1 and barrier gene expression/levels were measured using qRT-PCR (quantitative reverse transcription PCR) and Western blot. RESULTS: Stroke and subsequent rt-PA treatment increased edema, hemorrhage, MMP-9 activity, LOX-1 expression, and worsened neurological outcomes. LOX-1 inhibition improved neurological function, reduced edema, and improved endothelial barrier integrity. Elevated MMP-9 activity correlated with increased edema, infarct volume, and decreased neurological function. MMP-9 inhibition reduced MMP-9 activity and LOX-1 expression. In human brain microvascular endothelial cells, LOX-1/MMP-9 inhibition differentially attenuated MMP-9 levels, inflammation, and activation following hypoxia plus glucose deprivation/R. CONCLUSIONS: Our findings indicate that LOX-1 inhibition and ± MMP-9 inhibition attenuate negative aspects of ischemic stroke with rt-PA therapy, thus resulting in improved neurological function. While no synergistic effect was observed with simultaneous LOX-1 and MMP-9 inhibition, a distinct interaction is evident.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Rats , Humans , Animals , Tissue Plasminogen Activator , Matrix Metalloproteinase 9/metabolism , Ischemic Stroke/drug therapy , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Stroke/drug therapy , Stroke/pathology , Hemorrhage , Edema/drug therapy , Edema/pathology , Glucose/pharmacology , Infarction/drug therapy , Hypoxia
15.
Mol Cell ; 69(4): 622-635.e6, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29429924

ABSTRACT

TIA1 and TIAL1 encode a family of U-rich element mRNA-binding proteins ubiquitously expressed and conserved in metazoans. Using PAR-CLIP, we determined that both proteins bind target sites with identical specificity in 3' UTRs and introns proximal to 5' as well as 3' splice sites. Double knockout (DKO) of TIA1 and TIAL1 increased target mRNA abundance proportional to the number of binding sites and also caused accumulation of aberrantly spliced mRNAs, most of which are subject to nonsense-mediated decay. Loss of PRKRA by mis-splicing triggered the activation of the double-stranded RNA (dsRNA)-activated protein kinase EIF2AK2/PKR and stress granule formation. Ectopic expression of PRKRA cDNA or knockout of EIF2AK2 in DKO cells rescued this phenotype. Perturbation of maturation and/or stability of additional targets further compromised cell cycle progression. Our study reveals the essential contributions of the TIA1 protein family to the fidelity of mRNA maturation, translation, and RNA-stress-sensing pathways in human cells.


Subject(s)
Cell Cycle , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Stress, Physiological , T-Cell Intracellular Antigen-1/metabolism , eIF-2 Kinase/metabolism , CRISPR-Cas Systems , Cytoplasmic Granules/metabolism , HEK293 Cells , Humans , RNA Splice Sites , RNA Splicing , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/antagonists & inhibitors , Regulatory Sequences, Ribonucleic Acid , T-Cell Intracellular Antigen-1/antagonists & inhibitors , T-Cell Intracellular Antigen-1/genetics , Uridine/metabolism , eIF-2 Kinase/genetics
16.
Proc Natl Acad Sci U S A ; 120(28): e2305085120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37399395

ABSTRACT

Chronic cutaneous wounds remain a persistent unmet medical need that decreases life expectancy and quality of life. Here, we report that topical application of PY-60, a small-molecule activator of the transcriptional coactivator Yes-associated protein (YAP), promotes regenerative repair of cutaneous wounds in pig and human models. Pharmacological YAP activation enacts a reversible pro-proliferative transcriptional program in keratinocytes and dermal cells that results in accelerated re-epithelization and regranulation of the wound bed. These results demonstrate that transient topical administration of a YAP activating agent may represent a generalizable therapeutic approach to treating cutaneous wounds.


Subject(s)
Quality of Life , Wound Healing , Humans , Animals , Swine , Wound Healing/physiology , Skin/injuries , Keratinocytes/metabolism , Administration, Cutaneous
17.
J Biol Chem ; 300(1): 105465, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37979915

ABSTRACT

Calreticulin (CRT) was originally identified as a key calcium-binding protein of the endoplasmic reticulum. Subsequently, CRT was shown to possess multiple intracellular functions, including roles in calcium homeostasis and protein folding. Recently, several extracellular functions have been identified for CRT, including roles in cancer cell invasion and phagocytosis of apoptotic and cancer cells by macrophages. In the current report, we uncover a novel function for extracellular CRT and report that CRT functions as a plasminogen-binding receptor that regulates the conversion of plasminogen to plasmin. We show that human recombinant or bovine tissue-derived CRT dramatically stimulated the conversion of plasminogen to plasmin by tissue plasminogen activator or urokinase-type plasminogen activator. Surface plasmon resonance analysis revealed that CRT-bound plasminogen (KD = 1.8 µM) with moderate affinity. Plasminogen binding and activation by CRT were inhibited by ε-aminocaproic acid, suggesting that an internal lysine residue of CRT interacts with plasminogen. We subsequently show that clinically relevant CRT variants (lacking four or eight lysines in carboxyl-terminal region) exhibited decreased plasminogen activation. Furthermore, CRT-deficient fibroblasts generated 90% less plasmin and CRT-depleted MDA MB 231 cells also demonstrated a significant reduction in plasmin generation. Moreover, treatment of fibroblasts with mitoxantrone dramatically stimulated plasmin generation by WT but not CRT-deficient fibroblasts. Our results suggest that CRT is an important cellular plasminogen regulatory protein. Given that CRT can empower cells with plasmin proteolytic activity, this discovery may provide new mechanistic insight into the established role of CRT in cancer.


Subject(s)
Calreticulin , Plasminogen , Animals , Cattle , Humans , Calreticulin/genetics , Calreticulin/isolation & purification , Calreticulin/metabolism , Fibrinolysin/metabolism , Plasminogen/genetics , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Protein Domains/genetics , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Gene Knockout Techniques , Cell Line, Tumor , Neoplasms/physiopathology
18.
J Biol Chem ; 300(7): 107472, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38879005

ABSTRACT

African swine fever virus (ASFV) causes severe disease in domestic pigs and wild boars, seriously threatening the development of the global pig industry. Type I interferon (IFN-I) is an important component of innate immunity, inducing the transcription and expression of antiviral cytokines by activating Janus-activated kinase-signal transducer and activator of transcription (STAT). However, the underlying molecular mechanisms by which ASFV antagonizes IFN-I signaling have not been fully elucidated. Therefore, using coimmunoprecipitation, confocal microscopy, and dual luciferase reporter assay methods, we investigated these mechanisms and identified a novel ASFV immunosuppressive protein, pB475L, which interacts with the C-terminal domain of STAT2. Consequently, pB475L inhibited IFN-I signaling by inhibiting STAT1 and STAT2 heterodimerization and nuclear translocation. Furthermore, we constructed an ASFV-B475L7PM mutant strain by homologous recombination, finding that ASFV-B475L7PM attenuated the inhibitory effects on IFN-I signaling compared to ASFV-WT. In summary, this study reveals a new mechanism by which ASFV impairs host innate immunity.

19.
J Biol Chem ; 300(5): 107284, 2024 May.
Article in English | MEDLINE | ID: mdl-38614208

ABSTRACT

Receptor-mediated cellular uptake of specific ligands constitutes an important step in the dynamic regulation of individual protein levels in extracellular fluids. With a focus on the inflammatory lung, we here performed a proteomics-based search for novel ligands regulated by the mannose receptor (MR), a macrophage-expressed endocytic receptor. WT and MR-deficient mice were exposed to lipopolysaccharide, after which the protein content in their lung epithelial lining fluid was compared by tandem mass tag-based mass spectrometry. More than 1200 proteins were identified in the epithelial lining fluid using this unbiased approach, but only six showed a statistically different abundance. Among these, an unexpected potential new ligand, thrombospondin-4 (TSP-4), displayed a striking 17-fold increased abundance in the MR-deficient mice. Experiments using exogenous addition of TSP-4 to MR-transfected CHO cells or MR-positive alveolar macrophages confirmed that TSP-4 is a ligand for MR-dependent endocytosis. Similar studies revealed that the molecular interaction with TSP-4 depends on both the lectin activity and the fibronectin type-II domain of MR and that a closely related member of the TSP family, TSP-5, is also efficiently internalized by the receptor. This was unlike the other members of this protein family, including TSPs -1 and -2, which are ligands for a close MR homologue known as urokinase plasminogen activator receptor-associated protein. Our study shows that MR takes part in the regulation of TSP-4, an important inflammatory component in the injured lung, and that two closely related endocytic receptors, expressed on different cell types, undertake the selective endocytosis of distinct members of the TSP family.


Subject(s)
Lectins, C-Type , Lung Injury , Mannose Receptor , Mannose-Binding Lectins , Proteomics , Receptors, Cell Surface , Thrombospondins , Animals , Mice , CHO Cells , Cricetulus , Endocytosis , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , Ligands , Lipopolysaccharides/toxicity , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Lung Injury/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mannose-Binding Lectins/metabolism , Mannose-Binding Lectins/genetics , Mice, Knockout , Proteomics/methods , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Thrombospondins/metabolism , Thrombospondins/genetics
20.
J Biol Chem ; 300(3): 105779, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38395305

ABSTRACT

The newly discovered zoonotic coronavirus swine acute diarrhea syndrome coronavirus (SADS-CoV) causes acute diarrhea, vomiting, dehydration, and high mortality rates in newborn piglets. Although SADS-CoV uses different strategies to evade the host's innate immune system, the specific mechanism(s) by which it blocks the interferon (IFN) response remains unidentified. In this study, the potential of SADS-CoV nonstructural proteins (nsp) to inhibit the IFN response was detected. The results determined that nsp1 was a potent antagonist of IFN response. SADS-CoV nsp1 efficiently inhibited signal transducer and activator of transcription 1 (STAT1) phosphorylation by inducing Janus kinase 1 (JAK1) degradation. Subsequent research revealed that nsp1 induced JAK1 polyubiquitination through K11 and K48 linkages, leading to JAK1 degradation via the ubiquitin-proteasome pathway. Furthermore, SADS-CoV nsp1 induced CREB-binding protein degradation to inhibit IFN-stimulated gene production and STAT1 acetylation, thereby inhibiting STAT1 dephosphorylation and blocking STAT1 transport out of the nucleus to receive antiviral signaling. In summary, the results revealed the novel mechanisms by which SADS-CoV nsp1 blocks the JAK-STAT signaling pathway via the ubiquitin-proteasome pathway. This study yielded valuable findings on the specific mechanism of coronavirus nsp1 in inhibiting the JAK-STAT signaling pathway and the strategies of SADS-CoV in evading the host's innate immune system.


Subject(s)
Alphacoronavirus , Coronavirus Infections , Proteasome Endopeptidase Complex , Swine Diseases , Viral Nonstructural Proteins , Animals , Acetylation , Alphacoronavirus/physiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Swine , Ubiquitins/metabolism , Swine Diseases/metabolism , Swine Diseases/virology , HEK293 Cells , Vero Cells , Humans , Chlorocebus aethiops , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL