Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.914
Filter
1.
Cancer Immunol Immunother ; 73(8): 148, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38832958

ABSTRACT

Immunotherapy is one of the most promising anti-cancer treatment. It involves activating the host's own immune system to eliminate cancer cells. Activation of cGAS-STING pathway is promising therapeutic approach for cancer immunotherapy. However, in human clinical trials, targeting cGAS-STING pathway results in insufficient or unsustainable anti-tumor response. To enhance its effectiveness, combination with other anti-cancer therapies seems essential to achieve synergistic systemic anti-tumor response.The aim of this study was to evaluate whether the combination of STING agonist-cGAMP with anti-vascular RGD-(KLAKLAK)2 peptide results in a better anti-tumor response in poorly immunogenic tumors with various STING protein and αvß3 integrin status.Combination therapy inhibited growth of murine breast carcinoma more effectively than melanoma. In melanoma, the administration of STING agonist alone was sufficient to obtain a satisfactory therapeutic effect. In both tumor models we have noted stimulation of innate immune response following cGAMP administration alone or in combination. The largest population of immune cells infiltrating the TME after therapy were activated NK cells. Increased infiltration of cytotoxic CD8+ T lymphocytes within the TME was only observed in melanoma tumors. However, they also expressed the "exhaustion" PD-1 receptor. In contrast, in breast carcinoma tumors each therapy caused the drop in the number of infiltrating CD8+ T cells.The obtained results indicate an additional therapeutic benefit from combining STING agonist with an anti-vascular agent. However, this effect depends on the type of tumor, the status of its microenvironment and the expression of specific proteins such as STING and αvß3 family integrin.


Subject(s)
Membrane Proteins , Animals , Mice , Membrane Proteins/agonists , Female , Humans , Oligopeptides/pharmacology , Nucleotides, Cyclic/pharmacology , Nucleotides, Cyclic/administration & dosage , Immunotherapy/methods , Mice, Inbred C57BL , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
2.
Nat Commun ; 15(1): 4012, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740774

ABSTRACT

cGAS activates innate immune responses against cytosolic double-stranded DNA. Here, by determining crystal structures of cGAS at various reaction stages, we report a unifying catalytic mechanism. apo-cGAS assumes an array of inactive conformations and binds NTPs nonproductively. Dimerization-coupled double-stranded DNA-binding then affixes the active site into a rigid lock for productive metal•substrate binding. A web-like network of protein•NTP, intra-NTP, and inter-NTP interactions ensures the stepwise synthesis of 2'-5'/3'-5'-linked cGAMP while discriminating against noncognate NTPs and off-pathway intermediates. One divalent metal is sufficient for productive substrate binding, and capturing the second divalent metal is tightly coupled to nucleotide and linkage specificities, a process which manganese is preferred over magnesium by 100-fold. Additionally, we elucidate how mouse cGAS achieves more stringent NTP and linkage specificities than human cGAS. Together, our results reveal that an adaptable, yet precise lock-and-key-like mechanism underpins cGAS catalysis.


Subject(s)
Nucleotides, Cyclic , Nucleotidyltransferases , Animals , Humans , Mice , Catalytic Domain , Crystallography, X-Ray , DNA , Models, Molecular , Nucleotides, Cyclic/genetics , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Binding , Substrate Specificity
3.
J Immunol ; 212(12): 1945-1957, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38700419

ABSTRACT

The cytosolic detection of pathogen-derived nucleic acids has evolved as an essential strategy for host innate immune defense in mammals. One crucial component in this process is the stimulator of IFN genes (STING), which acts as a vital signaling adaptor, connecting the cytosolic detection of DNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) to the downstream type I IFN signaling pathway. However, this process remains elusive in invertebrates. In this study, we present evidence demonstrating that STING, an ortholog found in a marine invertebrate (shrimp) called Litopenaeus vannamei, can directly detect DNA and initiate an IFN-like antiviral response. Unlike its homologs in other eukaryotic organisms, which exclusively function as sensors for cyclic dinucleotides, shrimp STING has the ability to bind to both double-stranded DNA and cyclic dinucleotides, including 2'3'-cGAMP. In vivo, shrimp STING can directly sense DNA nucleic acids from an infected virus, accelerate IFN regulatory factor dimerization and nuclear translocation, induce the expression of an IFN functional analog protein (Vago4), and finally establish an antiviral state. Taken together, our findings unveil a novel double-stranded DNA-STING-IKKε-IRF-Vago antiviral axis in an arthropod, providing valuable insights into the functional origins of DNA-sensing pathways in evolution.


Subject(s)
Membrane Proteins , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Penaeidae/immunology , Penaeidae/virology , Immunity, Innate/immunology , Signal Transduction/immunology , Interferons/metabolism , Interferons/immunology , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/immunology
4.
Cell Chem Biol ; 31(5): 851-861, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38723635

ABSTRACT

Ten years ago, the second messenger cGAMP was discovered as the activator of the anti-cancer STING pathway. The characterization of cGAMP's paracrine action and dominant extracellular hydrolase ENPP1 cemented cGAMP as an intercellular immunotransmitter that coordinates the innate and adaptive immune systems to fight cancer. In this Perspective, I look back at a decade of discovery of extracellular cGAMP biology and drug development aiming to supply or preserve extracellular cGAMP for cancer treatment. Reviewing our understanding of the cell type-specific regulatory mechanisms of STING agonists, including their transporters and degradation enzymes, I explain on a molecular and cellular level the successes and challenges of direct STING agonists for cancer therapy. Based on what we know now, I propose new ways to stimulate the STING pathway in a manner that is not only cancer specific, but also cell type specific to fully harness the anti-cancer effect of cGAMP while avoiding collateral damage.


Subject(s)
Membrane Proteins , Neoplasms , Nucleotides, Cyclic , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Membrane Proteins/metabolism , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Animals
5.
Cell Rep ; 43(5): 114209, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38749434

ABSTRACT

2'3'-Cyclic guanosine monophosphate (GMP)-AMP (cGAMP) is a second messenger synthesized upon detection of cytosolic double-stranded DNA (dsDNA) and passed between cells to facilitate downstream immune signaling. Ectonucleotide pyrophosphatase phosphodiesterase I (ENPP1), an extracellular enzyme, was the only metazoan hydrolase known to regulate cGAMP levels to dampen anti-cancer immunity. Here, we uncover ENPP3 as the second and likely the only other metazoan cGAMP hydrolase under homeostatic conditions. ENPP3 has a tissue expression pattern distinct from ENPP1's and accounts for all cGAMP hydrolysis activity in ENPP1-deficient mice. Importantly, we also show that, as with ENPP1, selectively abolishing ENPP3's cGAMP hydrolysis activity results in diminished cancer growth and metastasis of certain tumor types in a stimulator of interferon genes (STING)-dependent manner. Both ENPP1 and ENPP3 are extracellular enzymes, suggesting the dominant role that extracellular cGAMP must play as a mediator of cell-cell innate immune communication. Our work demonstrates that ENPP1 and ENPP3 non-redundantly dampen extracellular cGAMP-STING signaling, pointing to ENPP3 as a target for cancer immunotherapy.


Subject(s)
Immunity, Innate , Membrane Proteins , Nucleotides, Cyclic , Phosphoric Diester Hydrolases , Pyrophosphatases , Animals , Nucleotides, Cyclic/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/genetics , Mice , Membrane Proteins/metabolism , Pyrophosphatases/metabolism , Pyrophosphatases/genetics , Humans , Mice, Inbred C57BL , Hydrolysis , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction
6.
Mol Cell ; 84(11): 2203-2213.e5, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38749421

ABSTRACT

The cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a pivotal role in innate immune responses to viral infection and inhibition of autoimmunity. Recent studies have suggested that micronuclei formed by genotoxic stress can activate innate immune signaling via the cGAS-STING pathway. Here, we investigated cGAS localization, activation, and downstream signaling from micronuclei induced by ionizing radiation, replication stress, and chromosome segregation errors. Although cGAS localized to ruptured micronuclei via binding to self-DNA, we failed to observe cGAS activation; cGAMP production; downstream phosphorylation of STING, TBK1, or IRF3; nuclear accumulation of IRF3; or expression of interferon-stimulated genes. Failure to activate the cGAS-STING pathway was observed across primary and immortalized cell lines, which retained the ability to activate the cGAS-STING pathway in response to dsDNA or modified vaccinia virus infection. We provide evidence that micronuclei formed by genotoxic insults contain histone-bound self-DNA, which we show is inhibitory to cGAS activation in cells.


Subject(s)
Chromosome Segregation , Membrane Proteins , Micronuclei, Chromosome-Defective , Nucleotides, Cyclic , Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Micronuclei, Chromosome-Defective/radiation effects , Nucleotides, Cyclic/metabolism , Phosphorylation , DNA Replication/radiation effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Immunity, Innate/radiation effects , DNA Damage , HEK293 Cells , Animals , Radiation, Ionizing , HeLa Cells
7.
ACS Chem Neurosci ; 15(8): 1652-1668, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38579109

ABSTRACT

In treating retinitis pigmentosa, a genetic disorder causing progressive vision loss, selective inhibition of rod cyclic nucleotide-gated (CNG) channels holds promise. Blocking the increased Ca2+-influx in rod photoreceptors through CNG channels can potentially delay disease progression and improve the quality of life for patients. To find inhibitors for rod CNG channels, we investigated the impact of 16 cGMP analogues on both rod and cone CNG channels using the patch-clamp technique. Although modifications at the C8 position of the guanine ring did not change the ligand efficacy, modifications at the N1 and N2 positions rendered cGMP largely ineffective in activating retinal CNG channels. Notably, PET-cGMP displayed selective potential, favoring rod over cone, whereas Rp-cGMPS showed greater efficiency in activating cone over rod CNG channels. Ligand docking and molecular dynamics simulations on cyclic nucleotide-binding domains showed comparable binding energies and binding modes for cGMP and its analogues in both rod and cone CNG channels (CNGA1 vs CNGA3 subunits). Computational experiments on CNGB1a vs CNGB3 subunits showed similar binding modes albeit with fewer amino acid interactions with cGMP due to an inactivated conformation of their C-helix. In addition, no clear correlation could be observed between the computational scores and the CNG channel efficacy values, suggesting additional factors beyond binding strength determining ligand selectivity and potency. This study highlights the importance of looking beyond the cyclic nucleotide-binding domain and toward the gating mechanism when searching for selective modulators. Future efforts in developing selective modulators for CNG channels should prioritize targeting alternative channel domains.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels , Quality of Life , Humans , Cyclic Nucleotide-Gated Cation Channels/metabolism , Ligands , Retina/metabolism , Nucleotides, Cyclic , Cyclic GMP/metabolism
8.
Pharmacol Ther ; 258: 108653, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679204

ABSTRACT

Cyclic nucleotides are important signaling molecules that play many critical physiological roles including controlling cell fate and development, regulation of metabolic processes, and responding to changes in the environment. Cyclic nucleotides are also pivotal regulators in immune signaling, orchestrating intricate processes that maintain homeostasis and defend against pathogenic threats. This review provides a comprehensive examination of the pharmacological potential of cyclic nucleotide signaling pathways within the realm of immunity. Beginning with an overview of the fundamental roles of cAMP and cGMP as ubiquitous second messengers, this review delves into the complexities of their involvement in immune responses. Special attention is given to the challenges associated with modulating these signaling pathways for therapeutic purposes, emphasizing the necessity for achieving cell-type specificity to avert unintended consequences. A major focus of the review is on the recent paradigm-shifting discoveries regarding specialized cyclic nucleotide signals in the innate immune system, notably the cGAS-STING pathway. The significance of cyclic dinucleotides, exemplified by 2'3'-cGAMP, in controlling immune responses against pathogens and cancer, is explored. The evolutionarily conserved nature of cyclic dinucleotides as antiviral agents, spanning across diverse organisms, underscores their potential as targets for innovative immunotherapies. Findings from the last several years have revealed a striking diversity of novel bacterial cyclic nucleotide second messengers which are involved in antiviral responses. Knowledge of the existence and precise identity of these molecules coupled with accurate descriptions of their associated immune defense pathways will be essential to the future development of novel antibacterial therapeutic strategies. The insights presented herein may help researchers navigate the evolving landscape of immunopharmacology as it pertains to cyclic nucleotides and point toward new avenues or lines of thinking about development of therapeutics against the pathways they regulate.


Subject(s)
Nucleotides, Cyclic , Signal Transduction , Humans , Animals , Nucleotides, Cyclic/metabolism , Immunity, Innate , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism
9.
Nat Microbiol ; 9(6): 1566-1578, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649411

ABSTRACT

The cyclic-oligonucleotide-based anti-phage signalling system (CBASS) is a type of innate prokaryotic immune system. Composed of a cyclic GMP-AMP synthase (cGAS) and CBASS-associated proteins, CBASS uses cyclic oligonucleotides to activate antiviral immunity. One major class of CBASS contains a homologue of eukaryotic ubiquitin-conjugating enzymes, which is either an E1-E2 fusion or a single E2. However, the functions of single E2s in CBASS remain elusive. Here, using biochemical, genetic, cryo-electron microscopy and mass spectrometry investigations, we discover that the E2 enzyme from Serratia marcescens regulates cGAS by imitating the ubiquitination cascade. This includes the processing of the cGAS C terminus, conjugation of cGAS to a cysteine residue, ligation of cGAS to a lysine residue, cleavage of the isopeptide bond and poly-cGASylation. The poly-cGASylation activates cGAS to produce cGAMP, which acts as an antiviral signal and leads to cell death. Thus, our findings reveal a unique regulatory role of E2 in CBASS.


Subject(s)
Nucleotidyltransferases , Ubiquitin-Conjugating Enzymes , Ubiquitination , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/chemistry , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/chemistry , Signal Transduction , Nucleotides, Cyclic/metabolism , Bacteriophages/genetics , Bacteriophages/enzymology , Ubiquitin/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Humans , Cryoelectron Microscopy , Immunity, Innate
10.
Cell Rep ; 43(5): 114122, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38652659

ABSTRACT

DNA sensing is important for antiviral immunity. The DNA sensor cGAS synthesizes 2'3'-cyclic GMP-AMP (cGAMP), a second messenger that activates STING, which induces innate immunity. cGAMP not only activates STING in the cell where it is produced but cGAMP also transfers to other cells. Transporters, channels, and pores (including SLC19A1, SLC46A2, P2X7, ABCC1, and volume-regulated anion channels (VRACs)) release cGAMP into the extracellular space and/or import cGAMP. We report that infection with multiple human viruses depletes some of these cGAMP conduits. This includes herpes simplex virus 1 (HSV-1) that targets SLC46A2, P2X7, and the VRAC subunits LRRC8A and LRRC8C for degradation. The HSV-1 protein UL56 is necessary and sufficient for these effects that are mediated at least partially by proteasomal turnover. UL56 thereby inhibits cGAMP uptake via VRAC, SLC46A2, and P2X7. Taken together, HSV-1 antagonizes intercellular cGAMP transfer. We propose that this limits innate immunity by reducing cell-to-cell communication via the immunotransmitter cGAMP.


Subject(s)
Herpesvirus 1, Human , Nucleotides, Cyclic , Herpesvirus 1, Human/physiology , Humans , Nucleotides, Cyclic/metabolism , Viral Proteins/metabolism , HEK293 Cells , Animals , Herpes Simplex/virology , Herpes Simplex/metabolism , Herpes Simplex/immunology
11.
Front Immunol ; 15: 1340001, 2024.
Article in English | MEDLINE | ID: mdl-38680492

ABSTRACT

Germinal center (GC) responses are essential for establishing protective, long-lasting immunity through the differentiation of GC B cells (BGC) and plasma cells (BPC), along with the generation of antigen-specific antibodies. Among the various pathways influencing immune responses, the STING (Stimulator of Interferon Genes) pathway has emerged as significant, especially in innate immunity, and extends its influence to adaptive responses. In this study, we examined how the STING ligand cGAMP can modulate these key elements of the adaptive immune response, particularly in enhancing GC reactions and the differentiation of BGC, BPC, and follicular helper T cells (TFH). Employing in vivo models, we evaluated various antigens and the administration of cGAMP in Alum adjuvant, investigating the differentiation of BGC, BPC, and TFH cells, along with the production of antigen-specific antibodies. cGAMP enhances the differentiation of BGC and BPC, leading to increased antigen-specific antibody production. This effect is shown to be type I Interferon-dependent, with a substantial reduction in BPC frequency upon interferon (IFN)-ß blockade. Additionally, cGAMP's influence on TFH differentiation varies over time, which may be critical for refining vaccine strategies. The findings elucidate a complex, antigen-specific influence of cGAMP on T and B cell responses, providing insights that could optimize vaccine efficacy.


Subject(s)
Cell Differentiation , Germinal Center , Membrane Proteins , Nucleotides, Cyclic , Signal Transduction , Germinal Center/immunology , Germinal Center/metabolism , Animals , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/immunology , Cell Differentiation/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Mice, Inbred C57BL , Lymphocyte Activation/immunology , Plasma Cells/immunology , Plasma Cells/metabolism
12.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(3): 252-258, 2024 Mar 12.
Article in Chinese | MEDLINE | ID: mdl-38448178

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is a DNA receptor that produces the second messenger cyclic GMP-AMP (cGAMP). cGAMP activates stimulator of interferon genes (STING), which initiates a signaling cascade leading to immune and inflammatory responses. This intricate molecular pathway plays a pivotal role in the pathogenesis and progression of diverse respiratory ailments, including respiratory infection, lung cancer, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, asthma, and acute lung injury. Consequently, the cGAS-STING signaling pathway has emerged as a promising novel therapeutic target, opening up new avenues for the diagnosis and treatment of respiratory disorders. This review focuses on recent advances in our understanding of the cGAS-STING signaling pathway and its intricate involvement in respiratory system diseases.


Subject(s)
Nucleotides, Cyclic , Respiration Disorders , Respiratory Tract Infections , Humans , Nucleotidyltransferases/genetics , Interferons
13.
Biomolecules ; 14(3)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38540769

ABSTRACT

Cyclic dinucleotides (CDNs) are cyclic molecules consisting of two nucleoside monophosphates linked by two phosphodiester bonds, which act as a second messenger and bind to the interferon gene stimulating factor (STING) to activate the downstream signaling pathway and ultimately induce interferon secretion, initiating an anti-infective immune response. Cyclic dinucleotides and their analogs are lead compounds in the immunotherapy of infectious diseases and tumors, as well as immune adjuvants with promising applications. Many agonists of pathogen recognition receptors have been developed as effective adjuvants to optimize vaccine immunogenicity and efficacy. In this work, the binding mechanism of human-derived interferon gene-stimulating protein and its isoforms with cyclic dinucleotides and their analogs was theoretically investigated using computer simulations and combined with experimental results in the hope of providing guidance for the subsequent synthesis of cyclic dinucleotide analogs.


Subject(s)
Membrane Proteins , Nucleotides, Cyclic , Humans , Membrane Proteins/metabolism , Second Messenger Systems , Interferons , Signal Transduction , Adjuvants, Immunologic
14.
Plant Physiol Biochem ; 208: 108522, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38493663

ABSTRACT

In staple crops, such as rice (Oryza sativa L.), pollen plays a crucial role in seed production. However, the molecular mechanisms underlying rice pollen germination and tube growth remain underexplored. Notably, we recently uncovered the redundant expression and mutual interaction of two rice genes encoding cyclic nucleotide-gated channels (CNGCs), OsCNGC4 and OsCNGC5, in mature pollen. Building on these findings, the current study focused on clarifying the functional roles of these two genes in pollen germination and tube growth. To overcome functional redundancy, we produced gene-edited rice plants with mutations in both genes using the CRISPR-Cas9 system. The resulting homozygous OsCNGC4 and OsCNGC5 gene-edited mutants (oscngc4/5) exhibited significantly lower pollen germination rates than the wild type (WT), along with severely reduced fertility. Transcriptome analysis of the double oscngc4/5 mutant revealed downregulation of genes related to receptor kinases, transporters, and cell wall metabolism. To identify the direct regulators of OsCNGC4, which form a heterodimer with OsCNGC5, we screened a yeast two-hybrid library containing rice cDNAs from mature anthers. Subsequently, we identified two calmodulin isoforms (CaM1-1 and CaM1-2), NETWORKED 2 A (NET2A), and proline-rich extension-like receptor kinase 13 (PERK13) proteins as interactors of OsCNGC4, suggesting its roles in regulating Ca2+ channel activity and F-actin organization. Overall, our results suggest that OsCNGC4 and OsCNGC5 may play critical roles in pollen germination and elongation by regulating the Ca2+ gradient in growing pollen tubes.


Subject(s)
Oryza , Oryza/physiology , Cyclic Nucleotide-Gated Cation Channels/genetics , Germination/genetics , Pollen/metabolism , Pollen Tube/genetics , Calmodulin/genetics , Calmodulin/metabolism , Phosphotransferases , Nucleotides, Cyclic/metabolism
15.
Elife ; 122024 Mar 22.
Article in English | MEDLINE | ID: mdl-38517938

ABSTRACT

Cyclic nucleotide binding domains (CNB) confer allosteric regulation by cAMP or cGMP to many signaling proteins, including PKA and PKG. PKA of phylogenetically distant Trypanosoma is the first exception as it is cyclic nucleotide-independent and responsive to nucleoside analogues (Bachmaier et al., 2019). Here, we show that natural nucleosides inosine, guanosine and adenosine are nanomolar affinity CNB ligands and activators of PKA orthologs of the important tropical pathogens Trypanosoma brucei, Trypanosoma cruzi, and Leishmania. The sequence and structural determinants of binding affinity, -specificity and kinase activation of PKAR were established by structure-activity relationship (SAR) analysis, co-crystal structures and mutagenesis. Substitution of two to three amino acids in the binding sites is sufficient for conversion of CNB domains from nucleoside to cyclic nucleotide specificity. In addition, a trypanosomatid-specific C-terminal helix (αD) is required for high affinity binding to CNB-B. The αD helix functions as a lid of the binding site that shields ligands from solvent. Selectivity of guanosine for CNB-B and of adenosine for CNB-A results in synergistic kinase activation at low nanomolar concentration. PKA pulldown from rapid lysis establishes guanosine as the predominant ligand in vivo in T. brucei bloodstream forms, whereas guanosine and adenosine seem to synergize in the procyclic developmental stage in the insect vector. We discuss the versatile use of CNB domains in evolution and recruitment of PKA for novel nucleoside-mediated signaling.


Subject(s)
Cyclic AMP , Purine Nucleosides , Cyclic AMP/metabolism , Nucleosides/pharmacology , Allosteric Regulation , Nucleotides, Cyclic , Guanosine , Adenosine
16.
PLoS Comput Biol ; 20(3): e1011559, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38517941

ABSTRACT

Cyclic AMP controls neuronal ion channel activity. For example hyperpolarization-activated cyclic nucleotide-gated (HCN) and M-type K+ channels are activated by cAMP. These effects have been suggested to be involved in astrocyte control of neuronal activity, for example, by controlling the action potential firing frequency. In cortical neurons, cAMP can induce mixed-mode oscillations (MMOs) consisting of small-amplitude, subthreshold oscillations separating complete action potentials, which lowers the firing frequency greatly. We extend a model of neuronal activity by including HCN and M channels, and show that it can reproduce a series of experimental results under various conditions involving and inferring with cAMP-induced activation of HCN and M channels. In particular, we find that the model can exhibit MMOs as found experimentally, and argue that both HCN and M channels are crucial for reproducing these patterns. To understand how M and HCN channels contribute to produce MMOs, we exploit the fact that the model is a three-time scale dynamical system with one fast, two slow, and two super-slow variables. We show that the MMO mechanism does not rely on the super-slow dynamics of HCN and M channel gating variables, since the model is able to produce MMOs even when HCN and M channel activity is kept constant. In other words, the cAMP-induced increase in the average activity of HCN and M channels allows MMOs to be produced by the slow-fast subsystem alone. We show that the slow-fast subsystem MMOs are due to a folded node singularity, a geometrical structure well known to be involved in the generation of MMOs in slow-fast systems. Besides raising new mathematical questions for multiple-timescale systems, our work is a starting point for future research on how cAMP signalling, for example resulting from interactions between neurons and glial cells, affects neuronal activity via HCN and M channels.


Subject(s)
Nucleotides, Cyclic , Potassium Channels , Potassium Channels/chemistry , Nucleotides, Cyclic/pharmacology , Neurons , Cyclic AMP , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Cyclic Nucleotide-Gated Cation Channels
17.
Nucleic Acids Res ; 52(6): 2761-2775, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38471818

ABSTRACT

CRISPR-Cas provides adaptive immunity in prokaryotes. Type III CRISPR systems detect invading RNA and activate the catalytic Cas10 subunit, which generates a range of nucleotide second messengers to signal infection. These molecules bind and activate a diverse range of effector proteins that provide immunity by degrading viral components and/or by disturbing key aspects of cellular metabolism to slow down viral replication. Here, we focus on the uncharacterised effector Csx23, which is widespread in Vibrio cholerae. Csx23 provides immunity against plasmids and phage when expressed in Escherichia coli along with its cognate type III CRISPR system. The Csx23 protein localises in the membrane using an N-terminal transmembrane α-helical domain and has a cytoplasmic C-terminal domain that binds cyclic tetra-adenylate (cA4), activating its defence function. Structural studies reveal a tetrameric structure with a novel fold that binds cA4 specifically. Using pulse EPR, we demonstrate that cA4 binding to the cytoplasmic domain of Csx23 results in a major perturbation of the transmembrane domain, consistent with the opening of a pore and/or disruption of membrane integrity. This work reveals a new class of cyclic nucleotide binding protein and provides key mechanistic detail on a membrane-associated CRISPR effector.


Many anti-viral defence systems generate a cyclic nucleotide signal that activates cellular defences in response to infection. Type III CRISPR systems use a specialised polymerase to make cyclic oligoadenylate (cOA) molecules from ATP. These can bind and activate a range of effector proteins that slow down viral replication. In this study, we focussed on the Csx23 effector from the human pathogen Vibrio cholerae ­ a trans-membrane protein that binds a cOA molecule, leading to anti-viral immunity. Structural studies revealed a new class of nucleotide recognition domain, where cOA binding is transmitted to changes in the trans-membrane domain, most likely resulting in membrane depolarisation. This study highlights the diversity of mechanisms for anti-viral defence via nucleotide signalling.


Subject(s)
Bacterial Proteins , CRISPR-Associated Proteins , Vibrio cholerae , Adenine Nucleotides/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nucleotides, Cyclic , Second Messenger Systems , Bacterial Proteins/metabolism , Vibrio cholerae/metabolism
18.
FEBS Lett ; 598(8): 839-863, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38453162

ABSTRACT

Cyclic nucleotides are the most diversified category of second messengers and are found in all organisms modulating diverse pathways. While cAMP and cGMP have been studied over 50 years, cyclic di-nucleotide signaling in eukaryotes emerged only recently with the anti-viral molecule 2´3´cGAMP. Recent breakthrough discoveries have revealed not only the astonishing chemical diversity of cyclic nucleotides but also surprisingly deep-rooted evolutionary origins of cyclic oligo-nucleotide signaling pathways and structural conservation of the proteins involved in their synthesis and signaling. Here we discuss how enzyme-centered approaches have paved the way for the identification of several cyclic nucleotide signals, focusing on the advantages and challenges associated with deciphering the activation mechanisms of such enzymes.


Subject(s)
Nucleotides, Cyclic , Nucleotides, Cyclic/metabolism , Humans , Animals , Signal Transduction , Cyclic GMP/metabolism , Cyclic AMP/metabolism
19.
Adv Sci (Weinh) ; 11(20): e2308131, 2024 May.
Article in English | MEDLINE | ID: mdl-38498770

ABSTRACT

To evade immune surveillance, tumor cells express ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) on the surface of their membrane, which degrades extracellular cyclic GMP-AMP (cGAMP), thereby inhibiting the cyclic GMP-AMP synthase (cGAS) stimulator of interferon gene (STING) DNA-sensing pathway. To fully understand this tumor stealth mechanism, it is essential to determine whether other forms of ENPP1 with hydrolytic cGAMP activity also are present in the tumor microenvironment to regulate this innate immune pathway. Herein, it is reported that various tumor-derived exosomes carry ENPP1, and can hydrolyze synthetic 2'3'-cGAMP and endogenous 2'3'-cGAMP produced by cells to inhibit cGAS-STING pathway in immune cells. Moreover, tumor exosomal ENPP1 also can hydrolyze 2'3'-cGAMP bound to LL-37 (an effective transporter of 2'3'-cGAMP) to inhibit STING signaling. Furthermore, high expression of ENPP1 in exosomes is observed isolated from human breast and lung cancer tissue, and tumor exosomal ENPP1 inhibited the immune infiltration of CD8+ T cells and CD4+ T cells. The results elucidate the essential function of tumor exosomal ENPP1 in the cGAS-STING pathway, furthering understanding of the crosstalk between the tumor cells and immune system.


Subject(s)
Exosomes , Membrane Proteins , Nucleotides, Cyclic , Nucleotidyltransferases , Phosphoric Diester Hydrolases , Pyrophosphatases , Signal Transduction , Nucleotides, Cyclic/metabolism , Pyrophosphatases/metabolism , Pyrophosphatases/genetics , Signal Transduction/genetics , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Exosomes/metabolism , Exosomes/genetics , Mice , Animals , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/immunology , Cell Line, Tumor , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
20.
J Cell Mol Med ; 28(4): e18139, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38334198

ABSTRACT

Platelets assume a pivotal role in the cardiovascular diseases (CVDs). Thus, targeting platelet activation is imperative for mitigating CVDs. Ginkgetin (GK), from Ginkgo biloba L, renowned for its anticancer and neuroprotective properties, remains unexplored concerning its impact on platelet activation, particularly in humans. In this investigation, we delved into the intricate mechanisms through which GK influences human platelets. At low concentrations (0.5-1 µM), GK exhibited robust inhibition of collagen and arachidonic acid (AA)-induced platelet aggregation. Intriguingly, thrombin and U46619 remained impervious to GK's influence. GK's modulatory effect extended to ATP release, P-selectin expression, intracellular calcium ([Ca2+ ]i) levels and thromboxane A2 formation. It significantly curtailed the activation of various signaling cascades, encompassing phospholipase Cγ2 (PLCγ2)/protein kinase C (PKC), phosphoinositide 3-kinase/Akt/glycogen synthase kinase-3ß and mitogen-activated protein kinases. GK's antiplatelet effect was not reversed by SQ22536 (an adenylate cyclase inhibitor) or ODQ (a guanylate cyclase inhibitor), and GK had no effect on the phosphorylation of vasodilator-stimulated phosphoproteinSer157 or Ser239 . Moreover, neither cyclic AMP nor cyclic GMP levels were significantly increased after GK treatment. In mouse studies, GK notably extended occlusion time in mesenteric vessels, while sparing bleeding time. In conclusion, GK's profound impact on platelet activation, achieved through inhibiting PLCγ2-PKC cascade, culminates in the suppression of downstream signaling and, ultimately, the inhibition of platelet aggregation. These findings underscore the promising therapeutic potential of GK in the CVDs.


Subject(s)
Biflavonoids , Nucleotides, Cyclic , Phospholipases , Humans , Animals , Mice , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Phospholipase C gamma/metabolism , Arachidonic Acid/pharmacology , Arachidonic Acid/metabolism , Phospholipases/metabolism , Phospholipases/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Activation , Blood Platelets/metabolism , Platelet Aggregation , Protein Kinase C/metabolism , Phosphorylation , Collagen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...