Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.736
Filter
1.
Eur J Med Chem ; 275: 116638, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38950489

ABSTRACT

The cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) pathway promotes antitumor immune responses by sensing cytosolic DNA fragments leaked from nucleus and mitochondria. Herein, we designed a highly charged ruthenium photosensitizer (Ru1) with a ß-carboline alkaloid derivative as the ligand for photo-activating of the cGAS-STING pathway. Due to the formation of multiple non-covalent intermolecular interactions, Ru1 can self-assemble into carrier-free nanoparticles (NPs). By incorporating the triphenylphosphine substituents, Ru1 can target and photo-damage mitochondrial DNA (mtDNA) to cause the cytoplasmic DNA leakage to activate the cGAS-STING pathway. Finally, Ru1 NPs show potent antitumor effects and elicit intense immune responses in vivo. In conclusion, we report the first self-assembling mtDNA-targeted photosensitizer, which can effectively activate the cGAS-STING pathway, thus providing innovations for the design of new photo-immunotherapeutic agents.


Subject(s)
Antineoplastic Agents , Immunotherapy , Membrane Proteins , Nucleotidyltransferases , Photosensitizing Agents , Ruthenium , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Ruthenium/chemistry , Ruthenium/pharmacology , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Nanoparticles/chemistry , Structure-Activity Relationship , Drug Screening Assays, Antitumor , DNA, Mitochondrial/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/pathology
2.
Int J Dev Biol ; 68(2): 85-91, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39016375

ABSTRACT

The tRNA-histidine guanylyltransferase 1-like (THG1L), also known as induced in high glucose-1 (IHG-1), encodes for an essential mitochondria-associated protein highly conserved throughout evolution, that catalyses the 3'-5' addition of a guanine to the 5'-end of tRNA-histidine (tRNAHis). Previous data indicated that THG1L plays a crucial role in the regulation of mitochondrial biogenesis and dynamics, in ATP production, and is critically involved in the modulation of apoptosis, cell-cycle progression and survival, as well as in cellular stress responses and redox homeostasis. Dysregulations of THG1L expression play a central role in various pathologies, including nephropathies, and neurodevelopmental disorders often characterized by developmental delay and cerebellar ataxia. Despite the essential role of THG1L, little is known about its expression during vertebrate development. Herein, we examined the detailed spatio-temporal expression of this gene in the developing Xenopus laevis. Our results show that thg1l is maternally inherited and its temporal expression suggests a role during the earliest stages of embryogenesis. Spatially, thg1l mRNA localizes in the ectoderm and marginal zone mesoderm during early stages of development. Then, at tadpole stages, thg1l transcripts mostly localise in neural crests and their derivatives, somites, developing kidney and central nervous system, therefore largely coinciding with territories displaying intense energy metabolism during organogenesis in Xenopus.


Subject(s)
Gene Expression Regulation, Developmental , Xenopus Proteins , Xenopus laevis , Animals , Xenopus laevis/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Embryonic Development/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
PLoS Genet ; 20(7): e1011341, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38954736

ABSTRACT

The drug floxuridine (5-fluorodeoxyuridine, FUdR) is an active metabolite of 5-Fluorouracil (5-FU). It converts to 5-fluorodeoxyuridine monophosphate (FdUMP) and 5-fluorodeoxyuridine triphosphate (FdUTP), which on incorporation into the genome inhibits DNA replication. Additionally, it inhibits thymidylate synthase, causing dTMP shortage while increasing dUMP availability, which induces uracil incorporation into the genome. However, the mechanisms underlying cellular tolerance to FUdR are yet to be fully elucidated. In this study, we explored the mechanisms underlying cellular resistance to FUdR by screening for FUdR hypersensitive mutants from a collection of DT40 mutants deficient in each genomic maintenance system. We identified REV3, which is involved in translesion DNA synthesis (TLS), to be a critical factor in FUdR tolerance. Replication using a FUdR-damaged template was attenuated in REV3-/- cells, indicating that the TLS function of REV3 is required to maintain replication on the FUdR-damaged template. Notably, FUdR-exposed REV3-/- cells exhibited defective cell cycle arrest in the early S phase, suggesting that REV3 is involved in intra-S checkpoint activation. Furthermore, REV3-/- cells showed defects in Chk1 phosphorylation, which is required for checkpoint activation, but the survival of FUdR-exposed REV3-/- cells was further reduced by the inhibition of Chk1 or ATR. These data indicate that REV3 mediates DNA checkpoint activation at least through Chk1 phosphorylation, but this signal acts in parallel with ATR-Chk1 DNA damage checkpoint pathway. Collectively, we reveal a previously unappreciated role of REV3 in FUdR tolerance.


Subject(s)
DNA Damage , DNA Replication , Floxuridine , Floxuridine/pharmacology , Animals , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/genetics , S Phase Cell Cycle Checkpoints/genetics , S Phase Cell Cycle Checkpoints/drug effects , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Chickens , Humans , DNA Repair/genetics , Phosphorylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Translesion DNA Synthesis , Deoxyuridine/analogs & derivatives
4.
Int J Biol Sci ; 20(9): 3497-3514, 2024.
Article in English | MEDLINE | ID: mdl-38993569

ABSTRACT

Resistance to HER2-targeted therapy is the major cause of treatment failure in patients with HER2+ breast cancer (BC). Given the key role of immune microenvironment in tumor development, there is a lack of an ideal prognostic model that fully accounts for immune infiltration. In this study, WGCNA analysis was performed to discover the relationship between immune-related signaling and prognosis of HER2+ BC. After Herceptin-resistant BC cell lines established, transcriptional profiles of resistant cell line and RNA-sequencing data from GSE76360 cohort were analyzed for candidate genes. 85 samples of HER2+ BC from TCGA database were analyzed by the Cox regression, XGBoost and Lasso algorithm to generalize a credible immune-related prognostic index (IRPI). Correlations between the IRPI signature and tumor microenvironment were further analyzed by multiple algorithms, including single-cell RNA sequencing data analysis. Patients with high IRPI had suppressive tumor immune microenvironment and worse prognosis. The suppression of type I interferon signaling indicated by the IRPI in Herceptin-resistant HER2+ BC was validated. And we elucidated that the suppression of cGAS-STING pathway is the key determinant underlying immune escape in Herceptin-resistant BC with high IRPI. A combination of STING agonist and DS-8201 could serve as a new strategy for Herceptin-resistant HER2+ BC.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Membrane Proteins , Nucleotidyltransferases , Receptor, ErbB-2 , Trastuzumab , Tumor Microenvironment , Humans , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/immunology , Female , Trastuzumab/therapeutic use , Trastuzumab/pharmacology , Drug Resistance, Neoplasm/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Signal Transduction , Cell Line, Tumor , Prognosis , Gene Expression Regulation, Neoplastic
5.
Protein Expr Purif ; 222: 106538, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38950762

ABSTRACT

Nucleotide sugars (UDP-Sugars) are essential for the production of polysaccharides and glycoconjugates utilized in medicines, cosmetics, and food industries. The enzyme Galactose-1-phosphate uridylyltransferase (GalU; EC 2.7.7.12) is responsible for the synthesis of UDP-galactose from α-d-galactose-1-phosphate (Gal-1P) and UTP. A novel bacterial GalU (TiGalU) encoded from a thermophilic bacterium, Thermodesulfatator indicus, was successfully purified using the Ni-NTA column after being expressed in Escherichia coli. The optimal pH for recombinant TiGalU was determined to be 5.5. The optimum temperature of the enzyme was 45 °C. The activity of TiGalU was not dependent on Mg2+ and was strongly inhibited by SDS. When coupled with galactose kinase (GALK1) and ß-1,4-galactosyltransferase 1 (B4GALT1), the enzyme enabled the one-pot synthesis of Gal-ß-1,4-GlcNAc-X by utilizing galactose and UTP as substrates. This study reported the in vitro biosynthesis of Gal-ß-1,4-GlcNAc-X for the first time, providing an environmentally friendly way to biosynthesis glycosides and other polysaccharides.


Subject(s)
Escherichia coli , Recombinant Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/isolation & purification , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics , UTP-Hexose-1-Phosphate Uridylyltransferase/metabolism , UTP-Hexose-1-Phosphate Uridylyltransferase/chemistry , Gene Expression , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/chemistry , Cloning, Molecular , Galactosephosphates/metabolism , Galactosephosphates/genetics , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Galactosyltransferases/chemistry
6.
FASEB J ; 38(13): e23663, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38958986

ABSTRACT

This study delves into the unexplored realm of castration-resistant prostate cancer (CRPC) by investigating the role of TRIM28 and its intricate molecular mechanisms using high-throughput single-cell transcriptome sequencing and advanced bioinformatics analysis. Our comprehensive examination unveiled dynamic TRIM28 expression changes, particularly in immune cells such as macrophages and CD8+ T cells within CRPC. Correlation analyses with TCGA data highlighted the connection between TRIM28 and immune checkpoint expression and emphasized its pivotal influence on the quantity and functionality of immune cells. Using TRIM28 knockout mouse models, we identified differentially expressed genes and enriched pathways, unraveling the potential regulatory involvement of TRIM28 in the cGAS-STING pathway. In vitro, experiments further illuminated that TRIM28 knockout in prostate cancer cells induced a notable anti-tumor immune effect by inhibiting M2 macrophage polarization and enhancing CD8+ T cell activity. This impactful discovery was validated in an in situ transplant tumor model, where TRIM28 knockout exhibited a deceleration in tumor growth, reduced proportions of M2 macrophages, and enhanced infiltration of CD8+ T cells. In summary, this study elucidates the hitherto unknown anti-tumor immune role of TRIM28 in CRPC and unravels its potential regulatory mechanism via the cGAS-STING signaling pathway. These findings provide novel insights into the immune landscape of CRPC, offering promising directions for developing innovative therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Membrane Proteins , Mice, Knockout , Prostatic Neoplasms, Castration-Resistant , Tripartite Motif-Containing Protein 28 , Tripartite Motif-Containing Protein 28/metabolism , Tripartite Motif-Containing Protein 28/genetics , Animals , Mice , Humans , Male , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Macrophages/metabolism , Macrophages/immunology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Mice, Inbred C57BL , Signal Transduction
7.
Invest Ophthalmol Vis Sci ; 65(8): 16, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980271

ABSTRACT

Purpose: The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) stimulator of interferon gene (STING) pathway is a crucial cascade in the inflammatory response initiated by the recognition of cytosolic double-stranded DNA (dsDNA). The aim of this study was to evaluate the effect of STING inhibitor in murine choroidal neovascularization (CNV). Methods: To investigate whether the cGAS-STING pathway is activated during CNV, CNV was induced using laser photocoagulation in male C57BL/6J mice. The expression of change of cGAS and STING during CNV development was confirmed by Western-blotting. H-151, a potent STING palmitoylation antagonist, was used as a STING inhibitor. H-151 was administered intravitreally immediately after laser induction. To confirm the role of the cGAS-STING pathway in CNV formation, we evaluated CNV size and performed fundus fluorescein angiography. Results: The expression levels of cGAS and STING were significantly upregulated in the RPE-choroid complex after CNV induction, and dsDNA merged with cGAS was observed in CNV lesions. Intravitreal administration of H-151 suppressed CNV development and fluorescent leakage from neovessels. In CNV lesions, the high expression of STING and cGAS was observed in infiltrating F4/80+ macrophages. H-151 administration attenuated downstream signals of the cGAS-STING pathway, including the phosphorylation of nuclear factor-κB, and downregulated the expression of interleukin 1ß. Conclusions: These findings support that the inhibition of cGAS-STING pathway treats abnormal ocular angiogenesis.


Subject(s)
Choroidal Neovascularization , Disease Models, Animal , Membrane Proteins , Mice, Inbred C57BL , Nucleotidyltransferases , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Male , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Blotting, Western , Fluorescein Angiography , Intravitreal Injections , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Choroid/metabolism , Choroid/pathology
8.
Int J Rheum Dis ; 27(7): e15256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982864

ABSTRACT

The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.


Subject(s)
Lupus Erythematosus, Systemic , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/drug therapy , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Animals , Autoimmunity , Interferon Type I/metabolism , Interferon Type I/immunology , Molecular Targeted Therapy , Immunity, Innate
9.
Gut Microbes ; 16(1): 2379633, 2024.
Article in English | MEDLINE | ID: mdl-39024479

ABSTRACT

Gut microbiota-derived extracellular vesicles (mEVs) are reported to regulate inflammatory response by delivering bacterial products into host cells. The complement receptor of the immunoglobulin superfamily macrophages (CRIg+ Mφ) could clear invading bacteria and their derivatives. Here, we investigate the role of CRIg+ Mφ and the mechanism by which mEVs regulate intestinal inflammation. We found that it is exacerbated in IBD patients and colitis mice by mEVs' leakage from disturbed gut microbiota, enriching microbial DNA in the intestinal mucosa. CRIg+ Mφ significantly decrease in IBD patients, allowing the spread of mEVs into the mucosa. The microbial DNA within mEVs is the key trigger for inflammation and barrier function damage. The cGAS/STING pathway is crucial in mEVs-mediated inflammatory injury. Blocking cGAS/STING signaling effectively alleviates inflammation caused by mEVs leakage and CRIg+ Mφ deficiency. Microbial DNA-containing mEVs, along with CRIg+ Mφ deficiency, stimulate inflammation in IBD, with the cGAS/STING pathway playing a crucial role.


Subject(s)
DNA, Bacterial , Extracellular Vesicles , Gastrointestinal Microbiome , Inflammation , Inflammatory Bowel Diseases , Intestinal Mucosa , Macrophages , Membrane Proteins , Nucleotidyltransferases , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Animals , Mice , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/immunology , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Inflammation/microbiology , Inflammation/metabolism , DNA, Bacterial/genetics , Mice, Inbred C57BL , Male , Female , Signal Transduction , Colitis/microbiology , Colitis/pathology
10.
Molecules ; 29(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38999073

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is pivotal in immunotherapy. Several agonists and inhibitors of the cGAS-STING pathway have been developed and evaluated for the treatment of various diseases. The agonists aim to activate STING, with cyclic dinucleotides (CDNs) being the most common, while the inhibitors aim to block the enzymatic activity or DNA binding ability of cGAS. Meanwhile, non-CDN compounds and cGAS agonists are also gaining attention. The omnipresence of the cGAS-STING pathway in vivo indicates that its overactivation could lead to undesired inflammatory responses and autoimmune diseases, which underscores the necessity of developing both agonists and inhibitors of the cGAS-STING pathway. This review describes the molecular traits and roles of the cGAS-STING pathway and summarizes the development of cGAS-STING agonists and inhibitors. The information is supposed to be conducive to the design of novel drugs for targeting the cGAS-STING pathway.


Subject(s)
Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Membrane Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/agonists , Signal Transduction/drug effects , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism
11.
Front Immunol ; 15: 1403070, 2024.
Article in English | MEDLINE | ID: mdl-39015575

ABSTRACT

Background: The cGAS-STING axis-mediated type I interferon pathway is a crucial strategy for host defense against DNA virus infection. Numerous evasion strategies developed by the pseudorabies virus (PRV) counteract host antiviral immunity. To what extent PRV-encoded proteins evade the cGAS-STING signaling pathway is unknown. Methods: Using US2 stably expressing cell lines and US2-deficient PRV model, we revealed that the PRV tegument protein US2 reduces STING protein stability and downregulates STING-mediated antiviral signaling. Results: To promote K48-linked ubiquitination and STING degradation, US2 interacts with the LBD structural domain of STING and recruits the E3 ligase TRIM21. TRIM21 deficiency consistently strengthens the host antiviral immune response brought on by PRV infection. Additionally, US2-deficient PRV is less harmful in mice. Conclusions: Our study implies that PRV US2 inhibits IFN signaling by a new mechanism that selectively targets STING while successfully evading the host antiviral response. As a result, the present study reveals a novel strategy by which PRV evades host defense and offers explanations for why the Bartha-K61 classical vaccine strain failed to offer effective defense against PRV variant strains in China, indicating that US2 may be a key target for developing gene-deficient PRV vaccines.


Subject(s)
Herpesvirus 1, Suid , Immunity, Innate , Membrane Proteins , Nucleotidyltransferases , Pseudorabies , Signal Transduction , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , Signal Transduction/immunology , Herpesvirus 1, Suid/immunology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Mice , Pseudorabies/immunology , Pseudorabies/virology , Humans , Ubiquitination , Ribonucleoproteins/immunology , Ribonucleoproteins/metabolism , Immune Evasion , Host-Pathogen Interactions/immunology , HEK293 Cells
12.
Sci Rep ; 14(1): 14146, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898119

ABSTRACT

Eribulin (ERI), clinically utilized for locally advanced or metastatic breast tumors, has shown potential links to the immune system. Notably, the cGAS-STING pathway, a key component of innate immunity, has gained prominence. Yet, limited reports explore ERI's effects on the cGAS-STING pathway. Additionally, the nuclear presence of cGAS remains poorly understood. This study uniquely delves into ERI's impact on both the cytosolic cGAS-STING pathway and nuclear cGAS. ERI enhances nuclear localization of cGAS, resulting in hyper-activation of the cGAS-STING pathway in triple-negative breast cancer cells. Reduction of cGAS heightened both cell proliferation and ERI sensitivity. In clinical data using ERI in a neo-adjuvant setting, patients with low cGAS cases exhibited reduced likelihood of achieving pathological complete response after ERI treatment. These findings illuminate the potential of cGAS and IFNß as predictive biomarkers for ERI sensitivity, providing valuable insights for personalized breast cancer treatment strategies.


Subject(s)
Cell Nucleus , Furans , Ketones , Nucleotidyltransferases , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Nucleotidyltransferases/metabolism , Female , Ketones/pharmacology , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Cell Line, Tumor , Furans/pharmacology , Cell Proliferation/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Signal Transduction/drug effects , Polyether Polyketides
13.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932169

ABSTRACT

Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNß transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNß accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.


Subject(s)
Cytomegalovirus , Membrane Proteins , Myeloid Cells , Nucleotidyltransferases , Protein Serine-Threonine Kinases , Signal Transduction , Valproic Acid , Humans , Valproic Acid/pharmacology , Myeloid Cells/virology , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Signal Transduction/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/genetics , Virus Latency/drug effects , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Interferon-beta/metabolism , Interferon-beta/genetics
14.
Redox Biol ; 74: 103224, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38865904

ABSTRACT

BACKGROUND: Silicosis, characterized by interstitial lung inflammation and fibrosis, poses a significant health threat. ATII cells play a crucial role in alveolar epithelial repair and structural integrity maintenance. Inhibiting ATII cell senescence has shown promise in silicosis treatment. However, the mechanism behind silica-induced senescence remains elusive. METHODS: The study employed male C57BL/6 N mice and A549 human alveolar epithelial cells to investigate silicosis and its potential treatment. Silicosis was induced in mice via intratracheal instillation of crystalline silica particles, with honokiol administered intraperitoneally for 14 days. Silica-induced senescence in A549 cells was confirmed, and SIRT3 knockout and overexpression cell lines were generated. Various analyses were conducted, including immunoblotting, qRT-PCR, histology, and transmission electron microscopy. Statistical significance was determined using one-way ANOVA with Tukey's post-hoc test. RESULTS: This study elucidates how silica induces ATII cell senescence, emphasizing mtDNA damage. Notably, honokiol (HKL) emerges as a promising anti-senescence and anti-fibrosis agent, acting through sirt3. honokiol effectively attenuated senescence in ATII cells, dependent on sirt3 expression, while mitigating mtDNA damage. Sirt3, a class III histone deacetylase, regulates senescence and mitochondrial stress. HKL activates sirt3, protecting against pulmonary fibrosis and mitochondrial damage. Additionally, HKL downregulated cGAS expression in senescent ATII cells induced by silica, suggesting sirt3's role as an upstream regulator of the cGAS/STING signaling pathway. Moreover, honokiol treatment inhibited the activation of the NF-κB signaling pathway, associated with reduced oxidative stress and mtDNA damage. Notably, HKL enhanced the activity of SOD2, crucial for mitochondrial function, through sirt3-mediated deacetylation. Additionally, HKL promoted the deacetylation activity of sirt3, further safeguarding mtDNA integrity. CONCLUSIONS: This study uncovers a natural compound, HKL, with significant anti-fibrotic properties through activating sirt3, shedding light on silicosis pathogenesis and treatment avenues.


Subject(s)
Alveolar Epithelial Cells , Biphenyl Compounds , Cellular Senescence , Lignans , Signal Transduction , Silicosis , Sirtuin 3 , Animals , Silicosis/metabolism , Silicosis/drug therapy , Silicosis/pathology , Silicosis/etiology , Sirtuin 3/metabolism , Sirtuin 3/genetics , Cellular Senescence/drug effects , Mice , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Biphenyl Compounds/pharmacology , Humans , Lignans/pharmacology , Signal Transduction/drug effects , Male , A549 Cells , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Disease Models, Animal , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , DNA Damage/drug effects , Allyl Compounds , Phenols
15.
Eur J Pharmacol ; 977: 176716, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38849039

ABSTRACT

The immune system has a strong connection to tumors. When a tumor cell is recognized as an abnormal cell by the immune system, the immune system may initiate an immune response to kill the tumor cell. In this study, RNA sequencing was performed on multiple myeloma (MM) cells treated with the proteasome inhibitor FHND6091. The transcriptional changes induced by FHND6091 in RPMI8226 cells aligned notably with immune response activation and results indicated upregulation of cGAS-STING pathway-related genes in the FHND6091-treated group. In vivo and in vitro experiments had demonstrated that FHND6091 stimulated the immunoreaction of MM cells via activation of the cyclic guanosine monophosphate-adenosine synthase/stimulator of interferon genes (cGAS-STING) pathway. This activation resulted in the generation of type-I interferons and the mobilization of natural killer (NK) cells. Notably, FHND6091 upregulated the levels of calreticulin and the protein ligands UL16-binding protein 2/5/6, MHC class I chain-related A (MICA), and MICB on the surface of MM cells. Subsequently, upon engaging with the surface activation receptors of NK cells, these ligands triggered NK cell activation, leading to the subsequent elimination of tumor cells. Thus, our findings elucidated the mechanism whereby FHND6091 exerted its immunotherapeutic activity as a STING agonist, enhancing the killing ability of NK cells against tumor cells.


Subject(s)
Killer Cells, Natural , Membrane Proteins , Multiple Myeloma , Proteasome Inhibitors , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Proteasome Inhibitors/pharmacology , Cell Line, Tumor , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Mice , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Calreticulin/metabolism , Calreticulin/genetics , Signal Transduction/drug effects , Cytotoxicity, Immunologic/drug effects , Interferon Type I/metabolism
16.
Adv Immunol ; 162: 1-21, 2024.
Article in English | MEDLINE | ID: mdl-38866436

ABSTRACT

Double stranded DNA (dsDNA) in the cytoplasm triggers the cGAS-STING innate immune pathway to defend against pathogenic infections, tissue damage and malignant cells. Extensive structural and functional studies over the last couple of years have enabled the molecular understanding of dsDNA induced activation of the cGAS-STING signaling pathway. This review highlights recent advances in the structural characterization of key molecules in the cGAS-STING signaling axis by focusing on the mechanism of cGAS activation by dsDNA, the regulation of cGAS activity, the mechanism of STING activation by cGAMP, the molecular basis of TBK1 recruitment and activation by STING, the structural basis of IRF3 recruitment by STING, and the mechanism of IRF3 activation upon phosphorylation by TBK1. These comprehensive structural studies provide a detailed picture of the mechanism of the cGAS-STING signaling pathway, establishing a molecular framework for the development of novel therapeutic strategies targeting this pathway.


Subject(s)
DNA , Immunity, Innate , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Nucleotidyltransferases/metabolism , DNA/metabolism , DNA/immunology , Membrane Proteins/metabolism , Animals , Interferon Regulatory Factor-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Phosphorylation
17.
JCI Insight ; 9(12)2024 May 21.
Article in English | MEDLINE | ID: mdl-38912586

ABSTRACT

Immune therapy is the new frontier of cancer treatment. Therapeutic radiation is a known inducer of immune response and can be limited by immunosuppressive mediators including cyclooxygenase-2 (COX2) that is highly expressed in aggressive triple negative breast cancer (TNBC). A clinical cohort of TNBC tumors revealed poor radiation therapeutic efficacy in tumors expressing high COX2. Herein, we show that radiation combined with adjuvant NSAID (indomethacin) treatment provides a powerful combination to reduce both primary tumor growth and lung metastasis in aggressive 4T1 TNBC tumors, which occurs in part through increased antitumor immune response. Spatial immunological changes including augmented lymphoid infiltration into the tumor epithelium and locally increased cGAS/STING1 and type I IFN gene expression were observed in radiation-indomethacin-treated 4T1 tumors. Thus, radiation and adjuvant NSAID treatment shifts "immune desert phenotypes" toward antitumor M1/TH1 immune mediators in these immunologically challenging tumors. Importantly, radiation-indomethacin combination treatment improved local control of the primary lesion, reduced metastatic burden, and increased median survival when compared with radiation treatment alone. These results show that clinically available NSAIDs can improve radiation therapeutic efficacy through increased antitumor immune response and augmented local generation of cGAS/STING1 and type I IFNs.


Subject(s)
Membrane Proteins , Signal Transduction , T-Lymphocytes, Cytotoxic , Animals , Membrane Proteins/metabolism , Mice , Female , Signal Transduction/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/radiotherapy , Indomethacin/pharmacology , Indomethacin/therapeutic use , Cell Line, Tumor , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism , Cyclooxygenase 2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Mice, Inbred BALB C
18.
J Immunol ; 213(3): 347-361, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38847616

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of IFN genes (STING) pathway is instrumental to antitumor immunity, yet the underlying molecular and cellular mechanisms are complex and still unfolding. A new paradigm suggests that cancer cells' cGAS-synthesized cGAMP can be transferred to tumor-infiltrating immune cells, eliciting STING-dependent IFN-ß response for antitumor immunity. Nevertheless, how the tumor microenvironment may shape this process remains unclear. In this study, we found that extracellular ATP, an immune regulatory molecule widely present in the tumor microenvironment, can potentiate cGAMP transfer, thereby boosting the STING signaling and IFN-ß response in murine macrophages and fibroblasts. Notably, genetic ablation or chemical inhibition of murine volume-regulation anion channel LRRC8/volume-regulated anion channel (VRAC), a recently identified cGAMP transporter, abolished ATP-potentiated cGAMP transfer and STING-dependent IFN-ß response, revealing a crucial role of LRRC8/VRAC in the cross-talk of extracellular ATP and cGAMP. Mechanistically, ATP activation of the P2X family receptors triggered Ca2+ influx and K+ efflux, promoting reactive oxygen species production. Moreover, ATP-evoked K+ efflux alleviated the phosphorylation of VRAC's obligate subunit LRRC8A/SWELL1 on S174. Mutagenesis studies indicated that the phosphorylation of S174 on LRRC8A could act as a checkpoint for VRAC in the steady state and a rheostat of ATP responsiveness. In an MC38-transplanted tumor model, systemically blocking CD39 and ENPP1, hydroxylases of extracellular ATP and cGAMP, respectively, elevated antitumor NK, NKT, and CD8+ T cell responses and restrained tumor growth in mice. Altogether, this study establishes a crucial role of ATP in facilitating LRRC8/VRAC transport cGAMP in the tumor microenvironment and provides new insight into harnessing cGAMP transfer for antitumor immunity.


Subject(s)
Adenosine Triphosphate , Membrane Proteins , Nucleotides, Cyclic , Tumor Microenvironment , Animals , Nucleotides, Cyclic/metabolism , Mice , Adenosine Triphosphate/metabolism , Membrane Proteins/metabolism , Membrane Proteins/immunology , Tumor Microenvironment/immunology , Interferon-beta/metabolism , Interferon-beta/immunology , Mice, Inbred C57BL , Humans , Signal Transduction/immunology , Mice, Knockout , Cell Line, Tumor , Cations/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Macrophages/immunology , Macrophages/metabolism
19.
Cell Host Microbe ; 32(7): 1074-1088.e5, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38917809

ABSTRACT

Cyclic oligonucleotide-based signaling system (CBASS) is an antiviral system that protects bacteria from phage infection and is evolutionarily related to human cGAS-STING immunity. cGAS-STING signaling is initiated by the recognition of viral DNA, but the molecular cues activating CBASS are incompletely understood. Using a screen of 975 type I CBASS operon-phage challenges, we show that operons with distinct cGAS/DncV-like nucleotidyltransferases (CD-NTases) and CD-NTase-associated protein (Cap) effectors exhibit marked patterns of phage restriction. We find that some type I CD-NTase enzymes require a C-terminal AGS-C immunoglobulin (Ig)-like fold domain for defense against select phages. Escaper phages evade CBASS via protein-coding mutations in virion assembly proteins, and acquired resistance is largely operon specific. We demonstrate that the phage Bas13 prohead protease interacts with the CD-NTase EcCdnD12 and can induce CBASS-dependent growth arrest in cells. Our results define phage virion assembly as a determinant of type I CBASS immune evasion and support viral protein recognition as a putative mechanism of cGAS-like enzyme activation.


Subject(s)
Bacteriophages , Immune Evasion , Humans , Bacteriophages/genetics , Operon , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Signal Transduction , Membrane Proteins/metabolism , Membrane Proteins/genetics , Peptide Hydrolases/metabolism , Peptide Hydrolases/genetics
20.
Neurochem Res ; 49(8): 2228-2248, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833090

ABSTRACT

Intermittent hypoxia (IH) is the predominant pathophysiological disturbance in obstructive sleep apnea (OSA), characterized by neuronal cell death and neurocognitive impairment. We focus on the accumulated mitochondrial DNA (mtDNA) in the cytosol, which acts as a damage-associated molecular pattern (DAMP) and activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, a known trigger for immune responses and neuronal death in degenerative diseases. However, the specific role and mechanism of the mtDNA-cGAS-STING axis in IH-induced neural damage remain largely unexplored. Here, we investigated the involvement of PANoptosis, a novel type of programmed cell death linked to cytosolic mtDNA accumulation and the cGAS-STING pathway activation, in neuronal cell death induced by IH. Our study found that PANoptosis occurred in primary cultures of hippocampal neurons and HT22 cell lines exposed to IH. In addition, we discovered that during IH, mtDNA released into the cytoplasm via the mitochondrial permeability transition pore (mPTP) activates the cGAS-STING pathway, exacerbating PANoptosis-associated neuronal death. Pharmacologically inhibiting mPTP opening or depleting mtDNA significantly reduced cGAS-STING pathway activation and PANoptosis in HT22 cells under IH. Moreover, our findings indicated that the cGAS-STING pathway primarily promotes PANoptosis by modulating endoplasmic reticulum (ER) stress. Inhibiting or silencing the cGAS-STING pathway substantially reduced ER stress-mediated neuronal death and PANoptosis, while lentivirus-mediated STING overexpression exacerbated these effects. In summary, our study elucidates that cytosolic escape of mtDNA triggers cGAS-STING pathway-dependent neuronal PANoptosis in response to IH, mainly through regulating ER stress. The discovery of the novel mechanism provides theoretical support for the prevention and treatment of neuronal damage and cognitive impairment in patients with OSA.


Subject(s)
Cytosol , DNA, Mitochondrial , Membrane Proteins , Neurons , Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , DNA, Mitochondrial/metabolism , Animals , Neurons/metabolism , Cytosol/metabolism , Signal Transduction/physiology , Mice , Hypoxia/metabolism , Mice, Inbred C57BL , Cell Line , Male , Hippocampus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...