Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.046
Filter
1.
Bioorg Med Chem Lett ; 109: 129847, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38857849

ABSTRACT

2'-5'-Adenosine linked nucleic acids are crucial components in living cells that play significant roles, including participating in antiviral defense mechanisms by facilitating the breakdown of viral genetic material. In this report, we present a chemical derivatization method employing 5-fluoro-2-pyridinoyl-imidazole as the acylation agent, a strategy that can be effectively combined with advanced analytical tools, including Nuclear Magnetic Resonance spectroscopy and Liquid Chromatography-Mass Spectrometry, to enhance the characterization and detection capabilities. This marks the first instance of a simple method designed to detect 2'-5'-adenosine linked nucleic acids. The new method is characterized by its time-saving nature, simplicity, and relative accuracy compared to previous methods.


Subject(s)
Adenosine , Acylation , Adenosine/chemistry , Adenosine/analogs & derivatives , Adenosine/analysis , Nucleic Acids/chemistry , Nucleic Acids/analysis , Imidazoles/chemistry , Molecular Structure , Magnetic Resonance Spectroscopy , Mass Spectrometry
2.
J Nanobiotechnology ; 22(1): 364, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38915007

ABSTRACT

Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.


Subject(s)
Immunotherapy , Nanomedicine , Photothermal Therapy , Tumor Microenvironment , Animals , Photothermal Therapy/methods , Immunotherapy/methods , Mice , Nanomedicine/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Neoplasms/therapy , Adenosine Triphosphate/metabolism , Adenosine/pharmacology , Adenosine/chemistry , Mice, Inbred C57BL , Apyrase/metabolism , Female , Phototherapy/methods
3.
Proc Natl Acad Sci U S A ; 121(25): e2404457121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865275

ABSTRACT

The fat mass and obesity-associated fatso (FTO) protein is a member of the Alkb family of dioxygenases and catalyzes oxidative demethylation of N6-methyladenosine (m6A), N1-methyladenosine (m1A), 3-methylthymine (m3T), and 3-methyluracil (m3U) in single-stranded nucleic acids. It is well established that the catalytic activity of FTO proceeds via two coupled reactions. The first reaction involves decarboxylation of alpha-ketoglutarate (αKG) and formation of an oxyferryl species. In the second reaction, the oxyferryl intermediate oxidizes the methylated nucleic acid to reestablish Fe(II) and the canonical base. However, it remains unclear how binding of the nucleic acid activates the αKG decarboxylation reaction and why FTO demethylates different methyl modifications at different rates. Here, we investigate the interaction of FTO with 5-mer DNA oligos incorporating the m6A, m1A, or m3T modifications using solution NMR, molecular dynamics (MD) simulations, and enzymatic assays. We show that binding of the nucleic acid to FTO activates a two-state conformational equilibrium in the αKG cosubstrate that modulates the O2 accessibility of the Fe(II) catalyst. Notably, the substrates that provide better stabilization to the αKG conformation in which Fe(II) is exposed to O2 are demethylated more efficiently by FTO. These results indicate that i) binding of the methylated nucleic acid is required to expose the catalytic metal to O2 and activate the αKG decarboxylation reaction, and ii) the measured turnover of the demethylation reaction (which is an ensemble average over the entire sample) depends on the ability of the methylated base to favor the Fe(II) state accessible to O2.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Iron , Ketoglutaric Acids , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/chemistry , Iron/metabolism , Iron/chemistry , Humans , Substrate Specificity , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry , Protein Conformation , Uracil/metabolism , Uracil/analogs & derivatives , Uracil/chemistry , Molecular Dynamics Simulation , Thymine/analogs & derivatives
4.
Nucleic Acids Res ; 52(12): 6733-6747, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38828787

ABSTRACT

Adenosine Deaminases Acting on RNA (ADARs) are enzymes that catalyze the conversion of adenosine to inosine in RNA duplexes. These enzymes can be harnessed to correct disease-causing G-to-A mutations in the transcriptome because inosine is translated as guanosine. Guide RNAs (gRNAs) can be used to direct the ADAR reaction to specific sites. Chemical modification of ADAR guide strands is required to facilitate delivery, increase metabolic stability, and increase the efficiency and selectivity of the editing reaction. Here, we show the ADAR reaction is highly sensitive to ribose modifications (e.g. 4'-C-methylation and Locked Nucleic Acid (LNA) substitution) at specific positions within the guide strand. Our studies were enabled by the synthesis of RNA containing a new, ribose-modified nucleoside analog (4'-C-methyladenosine). Importantly, the ADAR reaction is potently inhibited by LNA or 4'-C-methylation at different positions in the ADAR guide. While LNA at guide strand positions -1 and -2 block the ADAR reaction, 4'-C-methylation only inhibits at the -2 position. These effects are rationalized using high-resolution structures of ADAR-RNA complexes. This work sheds additional light on the mechanism of ADAR deamination and aids in the design of highly selective ADAR guide strands for therapeutic editing using chemically modified RNA.


Subject(s)
Adenosine Deaminase , RNA Editing , Ribose , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/chemistry , Ribose/chemistry , Ribose/metabolism , Humans , Oligonucleotides/chemistry , Oligonucleotides/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Methylation , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry , Nucleosides/chemistry , Nucleosides/metabolism , RNA/metabolism , RNA/chemistry , Inosine/metabolism , Inosine/chemistry
5.
J Mater Chem B ; 12(24): 5787-5811, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38845588

ABSTRACT

Cancer immunotherapy, as an emerging approach to cancer treatment, has tremendous potential for application. Compared to traditional methods such as surgery, chemotherapy, and radiation therapy, it has the ability to restore the patient's immune system, leading to long-term immune memory with less damage to normal tissues. However, immunotherapy has its limitations, including limited therapeutic efficacy, restricted patient populations, and inconsistent treatment responses. Finding effective immunotherapeutic approaches has become a key focus of its clinical application. The adenosine pathway is a recently discovered tumor immune regulatory signaling pathway. It can influence the metabolism and growth of tumor cells by acting through key enzymes in the adenosine pathway, thereby affecting the development of tumors. Therefore, inhibiting the adenosine pathway is an effective cancer immunotherapy. Common adenosine pathway inhibitors include small molecules and antibody proteins, and extensive preclinical trials have demonstrated their effectiveness in inhibiting tumor growth. The short half-life, low bioavailability, and single administration route of adenosine pathway inhibitors limit their clinical application. With the advent of nanotechnology, nano-delivery of adenosine pathway inhibitors has addressed these issues. Compared to traditional drugs, nano-drugs extend the drug's circulation time and improve its distribution within the body. They also offer targeting capabilities and have low toxic side effects, making them very promising for future applications. In this review, we discuss the mechanism of the adenosine pathway in tumor immune suppression, the clinical applications of adenosine pathway inhibitors, and nano-delivery based on adenosine pathway inhibitors. In the final part of this article, we also briefly discuss the technical issues and challenges currently present in nano-delivery of adenosine pathway inhibitors, with the hope of advancing the progress of adenosine inhibitor nano-drugs in clinical treatment.


Subject(s)
Adenosine , Immunotherapy , Nanoparticles , Neoplasms , Humans , Adenosine/chemistry , Adenosine/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Immunotherapy/methods , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
6.
Molecules ; 29(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38893537

ABSTRACT

ß-Thalassemia is an inherited genetic disorder associated with ß-globin chain synthesis, which ultimately becomes anemia. Adenosine-2,3-dialdehyde, by inhibiting arginine methyl transferase 5 (PRMT5), can induce fetal hemoglobin (HbF) levels. Hence, the materialization of PRMT5 inhibitors is considered a promising therapy in the management of ß-thalassemia. This study conducted a virtual screening of certain compounds similar to 5'-deoxy-5'methyladenosine (3XV) using the PubChem database. The top 10 compounds were chosen based on the best docking scores, while their interactions with the PRMT5 active site were analyzed. Further, the top two compounds demonstrating the lowest binding energy were subjected to drug-likeness analysis and pharmacokinetic property predictions, followed by molecular dynamics simulation studies. Based on the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) score and molecular interactions, (3R,4S)-2-(6-aminopurin-9-yl)-5-[(4-ethylcyclohexyl)sulfanylmethyl]oxolane-3,4-diol (TOP1) and 2-(6-Aminopurin-9-yl)-5-[(6-aminopurin-9-yl)methylsulfanylmethyl]oxolane-3,4-diol (TOP2) were identified as potential hit compounds, while TOP1 exhibited higher binding affinity and stabler binding capabilities than TOP2 during molecular dynamics simulation (MDS) analysis. Taken together, the outcomes of our study could aid researchers in identifying promising PRMT5 inhibitors. Moreover, further investigations through in vivo and in vitro experiments would unquestionably confirm that this compound could be employed as a therapeutic drug in the management of ß-thalassemia.


Subject(s)
Enzyme Inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein-Arginine N-Methyltransferases , beta-Thalassemia , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , beta-Thalassemia/drug therapy , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Drug Discovery , Protein Binding , Catalytic Domain , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/pharmacology
7.
Eur J Med Chem ; 274: 116526, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38805939

ABSTRACT

Epitranscriptomics, the field of post-translational RNA modifications, is a burgeoning domain of research that has recently received significant attention for its role in multiple diseases, including cancer. N6-methyladenosine (m6A) is the most prominent post-translational RNA modification and plays a critical role in RNA transcription, processing, translation, and metabolism. The m6A modification is controlled by three protein classes known as writers (methyltransferases), erasers (demethylases), and readers (m6A-binding proteins). Each class of m6A regulatory proteins has been implicated in cancer initiation and progression. As such, many of these proteins have been identified as potential targets for anti-cancer chemotherapeutics. In this work, we provide an overview of the role m6A-regulating proteins play in cancer and discuss the current state of small molecule therapeutics targeting these proteins.


Subject(s)
Adenosine , Antineoplastic Agents , Neoplasms , Small Molecule Libraries , Humans , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/pharmacology , Adenosine/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , RNA/metabolism , Animals , Molecular Structure , RNA Processing, Post-Transcriptional/drug effects
8.
Int J Biol Macromol ; 269(Pt 1): 131966, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697422

ABSTRACT

JAK2/STAT3/MYC axis is dysregulated in nearly 70 % of human cancers, but targeting this pathway therapeutically remains a big challenge in cancer therapy. In this study, genes associated with JAK2, STAT3, and MYC were analyzed, and potential target genes were selected. Leucine-rich PPR motif-containing protein (LRPPRC) whose function and regulation are not fully understood, emerged as one of top 3 genes in terms of RNA epigenetic modification. Here, we demonstrate LRPPRC may be an independent prognostic indicator besides JAK2, STAT3, and MYC. Mechanistically, LRPPRC impairs N6-methyladenosine (m6A) modification of JAK2, STAT3, and MYC to facilitate nuclear mRNA export and expression. Meanwhile, excess LRPPRC act as a scaffold protein binding to JAK2 and STAT3 to enhance stability of JAK2-STAT3 complex, thereby facilitating JAK2/STAT3/MYC axis activation to promote esophageal squamous cell carcinoma (ESCC) progression. Furthermore, 5,7,4'-trimethoxyflavone was verified to bind to LRPPRC, STAT3, and CDK1, dissociating LRPPRC-JAK2-STAT3 and JAK2-STAT3-CDK1 interaction, leading to impaired tumorigenesis in 4-Nitroquinoline N-oxide induced ESCC mouse models and suppressed tumor growth in ESCC patient derived xenograft mouse models. In summary, this study suggests regulation of m6A modification by LRPPRC, and identifies a novel triplex target compound, suggesting that targeting LRPPRC-mediated JAK2/STAT3/MYC axis may overcome JAK2/STAT3/MYC dependent tumor therapeutic dilemma.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Janus Kinase 2 , STAT3 Transcription Factor , Humans , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/genetics , STAT3 Transcription Factor/metabolism , Animals , Janus Kinase 2/metabolism , Mice , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , Disease Progression , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/metabolism , Adenosine/chemistry , Flavones/pharmacology , Flavones/chemistry , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/genetics , Signal Transduction/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Female , Male , Flavonoids/pharmacology , Flavonoids/chemistry , Xenograft Model Antitumor Assays , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics
9.
Int J Biol Macromol ; 270(Pt 1): 132289, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735607

ABSTRACT

S-Adenosyl-l-homocysteine hydrolase (SAHH) is a crucial enzyme that governs S-adenosyl methionine (SAM)-dependent methylation reactions within cells and regulates the intracellular concentration of SAH. Legionella pneumophila, the causative pathogen of Legionnaires' disease, encodes Lpg2021, which is the first identified dimeric SAHH in bacteria and is a promising target for drug development. Here, we report the structure of Lpg2021 in its ligand-free state and in complexes with adenine (ADE), adenosine (ADO), and 3-Deazaneplanocin A (DZNep). X-ray crystallography, isothermal titration calorimetry (ITC), and molecular docking were used to elucidate the binding mechanisms of Lpg2021 to its substrates and inhibitors. Virtual screening was performed to identify potential Lpg2021 inhibitors. This study contributes a novel perspective to the understanding of SAHH evolution and establishes a structural framework for designing specific inhibitors targeting pathogenic Legionella pneumophila SAHH.


Subject(s)
Adenosylhomocysteinase , Legionella pneumophila , Molecular Docking Simulation , Legionella pneumophila/enzymology , Substrate Specificity , Adenosylhomocysteinase/metabolism , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/chemistry , Crystallography, X-Ray , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry , Adenine/chemistry , Adenine/metabolism , Adenine/analogs & derivatives , Protein Binding , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , N-Glycosyl Hydrolases
10.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731538

ABSTRACT

Adenosine, as a water-soluble active substance, has various pharmacological effects. This study proposes a layer-by-layer assembly method of composite wall materials, using hydroxypropyl-ß-cyclodextrin as the inner wall and whey protein isolate as the outer wall, to encapsulate adenosine within the core material, aiming to enhance adenosine microcapsules' stability through intermolecular interactions. By combining isothermal titration calorimetry with molecular modeling analysis, it was determined that the core material and the inner wall and the inner wall and the outer wall interact through intermolecular forces. Adenosine and hydroxypropyl-ß-cyclodextrin form an optimal 1:1 complex through hydrophobic interactions, while hydroxypropyl-ß-cyclodextrin and whey protein isolate interact through hydrogen bonds. The embedding rate of AD/Hp-ß-CD/WPI microcapsules was 36.80%, and the 24 h retention rate under the release behavior test was 76.09%. The method of preparing adenosine microcapsules using composite wall materials is environmentally friendly and shows broad application prospects in storage and delivery systems with sustained release properties.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin , Adenosine , Capsules , Whey Proteins , Whey Proteins/chemistry , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , Capsules/chemistry , Adenosine/chemistry , Drug Compounding/methods , Hydrophobic and Hydrophilic Interactions , Drug Liberation , Models, Molecular , Hydrogen Bonding , Layer-by-Layer Nanoparticles
11.
Molecules ; 29(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38731610

ABSTRACT

Many liqueurs, including spirits infused with botanicals, are crafted not only for their taste and flavor but also for potential medicinal benefits. However, the scientific evidence supporting their medicinal effects remains limited. This study aims to verify in vitro anticancer activity and bioactive compounds in shochu spirits infused with Cordyceps militaris, a Chinese medicine. The results revealed that a bioactive fraction was eluted from the spirit extract with 40% ethanol. The infusion time impacted the inhibitory effect of the spirit extract on the proliferation of colon cancer-derived cell line HCT-116 cells, and a 21-day infusion showed the strongest inhibitory effect. Furthermore, the spirit extract was separated into four fractions, A-D, by high-performance liquid chromatography (HPLC), and Fractions B, C, and D, but not A, exerted the effects of proliferation inhibition and apoptotic induction of HCT-116 cells and HL-60 cells. Furthermore, Fractions B, C, and D were, respectively, identified as adenosine, cordycepin, and N6-(2-hydroxyethyl)-adenosine (HEA) by comprehensive chemical analyses, including proton nuclear magnetic resonance (1H-NMR), Fourier transform infrared spectroscopy (FT-IR), and electrospray ionization mass spectrometry (ESI-MS). To better understand the bioactivity mechanisms of cordycepin and HEA, the agonist and antagonist tests of the A3 adenosine receptor (A3AR) were performed. Cell viability was suppressed by cordycepin, and HEA was restored by the A3AR antagonist MR1523, suggesting that cordycepin and HEA possibly acted as agonists to activate A3ARs to inhibit cell proliferation. Molecular docking simulations revealed that both adenosine and cordycepin bound to the same pocket site of A3ARs, while HEA exhibited a different binding pattern, supporting a possible explanation for the difference in their bioactivity. Taken together, the present study demonstrated that cordycepin and HEA were major bioactive ingredients in Cordyceps militaries-infused sweet potato shochu spirits, which contributed to the in vitro anticancer activity.


Subject(s)
Apoptosis , Cell Proliferation , Cordyceps , Humans , Cordyceps/chemistry , Cell Proliferation/drug effects , HCT116 Cells , Apoptosis/drug effects , Adenosine/pharmacology , Adenosine/analogs & derivatives , Adenosine/chemistry , Deoxyadenosines/pharmacology , Deoxyadenosines/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Molecular Docking Simulation , HL-60 Cells , Chromatography, High Pressure Liquid , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor
12.
Int J Nanomedicine ; 19: 4181-4197, 2024.
Article in English | MEDLINE | ID: mdl-38766656

ABSTRACT

Purpose: The committed differentiation fate regulation has been a difficult problem in the fields of stem cell research, evidence showed that nanomaterials could promote the differentiation of stem cells into specific cell types. Layered double hydroxide (LDH) nanoparticles possess the regulation function of stem cell fate, while the underlying mechanism needs to be investigated. In this study, the process of embryonic stem cells (ESCs) differentiate to neural progenitor cells (NPCs) by magnesium aluminum LDH (MgAl-LDH) was investigated. Methods: MgAl-LDH with diameters of 30, 50, and 100 nm were synthesized and characterized, and their effects on the cytotoxicity and differentiation of NPCs were detected in vitro. Dot blot and MeRIP-qPCR were performed to detect the level of m6A RNA methylation in nanoparticles-treated cells. Results: Our work displayed that LDH nanoparticles of three different sizes were biocompatible with NPCs, and the addition of MgAl-LDH could significantly promote the process of ESCs differentiate to NPCs. 100 nm LDH has a stronger effect on promoting NPCs differentiation compared to 30 nm and 50 nm LDH. In addition, dot blot results indicated that the enhanced NPCs differentiation by MgAl-LDH was closely related to m6A RNA methylation process, and the major modification enzyme in LDH controlled NPCs differentiation may be the m6A RNA methyltransferase METTL3. The upregulated METTL3 by LDH increased the m6A level of Sox1 mRNA, enhancing its stability. Conclusion: This work reveals that MgAl-LDH nanoparticles can regulate the differentiation of ESCs into NPCs by increasing m6A RNA methylation modification of Sox1.


Subject(s)
Cell Differentiation , Nanoparticles , Neural Stem Cells , Cell Differentiation/drug effects , Animals , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Mice , Nanoparticles/chemistry , Methylation/drug effects , Hydroxides/chemistry , Hydroxides/pharmacology , Methyltransferases/metabolism , Methyltransferases/genetics , Particle Size , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/cytology , Adenosine/pharmacology , Adenosine/chemistry , Adenosine/analogs & derivatives , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacology , Magnesium Hydroxide/chemistry , Magnesium Hydroxide/pharmacology
13.
Angew Chem Int Ed Engl ; 63(24): e202402611, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38607929

ABSTRACT

METTL3, a primary methyltransferase catalyzing the RNA N6-methyladenosine (m6A) modification, has been identified as an oncogene in several cancer types and thus nominated as a potentially effective target for therapeutic inhibition. However, current options using this strategy are limited. In this study, we targeted protein-protein interactions at the METTL3-METTL14 binding interface to inhibit complex formation and subsequent catalysis of the RNA m6A modification. Among candidate peptides, RM3 exhibited the highest anti-cancer potency, inhibiting METTL3 activity while also facilitating its proteasomal degradation. We then designed a stapled peptide inhibitor (RSM3) with enhanced peptide stability and formation of the α-helical secondary structure required for METTL3 interaction. Functional and transcriptomic analysis in vivo indicated that RSM3 induced upregulation of programmed cell death-related genes while inhibiting cancer-promoting signals. Furthermore, tumor growth was significantly suppressed while apoptosis was enhanced upon RSM3 treatment, accompanied by increased METTL3 degradation, and reduced global RNA methylation levels in two in vivo tumor models. This peptide inhibitor thus exploits a mechanism distinct from other small-molecule competitive inhibitors to inhibit oncogenic METTL3 activity. Our findings collectively highlight the potential of targeting METTL3 in cancer therapies through peptide-based inhibition of complex formation and proteolytic degradation.


Subject(s)
Antineoplastic Agents , Methyltransferases , Peptides , Methyltransferases/metabolism , Methyltransferases/antagonists & inhibitors , Humans , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/metabolism , Adenosine/pharmacology , Animals , Cell Proliferation/drug effects , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Cell Line, Tumor , Apoptosis/drug effects
14.
Angew Chem Int Ed Engl ; 63(26): e202320029, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38591694

ABSTRACT

N1-methyladenosine (m1A) modification is one of the most prevalent epigenetic modifications on RNA. Given the vital role of m1A modification in RNA processing such as splicing, stability and translation, developing a precise and controllable m1A editing tool is pivotal for in-depth investigating the biological functions of m1A. In this study, we developed an abscisic acid (ABA)-inducible and reversible m1A demethylation tool (termed AI-dm1A), which targets specific transcripts by combining the chemical proximity-induction techniques with the CRISPR/dCas13b system and ALKBH3. We successfully employed AI-dm1A to selectively demethylate the m1A modifications at A8422 of MALAT1 RNA, and this demethylation process could be reversed by removing ABA. Furthermore, we validated its demethylation function on various types of cellular RNAs including mRNA, rRNA and lncRNA. Additionally, we used AI-dm1A to specifically demethylate m1A on ATP5D mRNA, which promoted ATP5D expression and enhanced the glycolysis activity of tumor cells. Conversely, by replacing the demethylase ALKBH3 with methyltransferase TRMT61A, we also developed a controllable m1A methylation tool, namely AI-m1A. Finally, we caged ABA by 4,5-dimethoxy-2-nitrobenzyl (DMNB) to achieve light-inducible m1A methylation or demethylation on specific transcripts. Collectively, our m1A editing tool enables us to flexibly study how m1A modifications on specific transcript influence biological functions and phenotypes.


Subject(s)
Adenosine , RNA Editing , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/metabolism , Humans , Abscisic Acid/pharmacology , Abscisic Acid/chemistry , Abscisic Acid/metabolism , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , RNA/metabolism , RNA/chemistry
15.
J Nanobiotechnology ; 22(1): 214, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689291

ABSTRACT

Combination of tumor immunotherapy with photothermal therapy (PTT) is a feasible tactic to overcome the drawback of immunotherapy such as poor immune response. Via triggering the immunogenic cells death (ICD), PTT can stimulate the activity of immune cells, but meanwhile, the level of adenosine is elevated via the CD73-induced decomposition of ATP which is overexpressed accompanying with the PTT process, resulting in negative feedback to impair the immune stimulation. Herein, we developed a novel biomimetic photothermal nanodrug to specifically block CD73 for inhibition of adenosine production and more efficient priming of the suppressive immune microenvironments. The nanodrug, named as AptEM@CBA, is constructed by encapsulation of photothermal agent black phosphorus quantum dots (BPQDs) and selective CD73 inhibitor α, ß-Methyleneadenosine 5'-diphosphate (AMPCP) in chitosan nanogels, which are further covered with aptamer AS1411 modified erythrocyte membrane (EM) for biomimetic camouflage. With AS1411 induced active targeting and EM induced long blood circulation time, the enrichment of the nanodrug tumor sites is promoted. The photothermal treatment promotes the maturation of dendritic cells. Meanwhile, the release of AMPCP suppress the adenosine generation via CD73 blockade, alleviating the impairment of adenosine to dendritic cells and suppressing regulatory T cells, synergically stimulate the activity of T cells. The combination of CD73 blockade with PTT, not only suppresses the growth of primary implanted tumors, but also boosts strong antitumor immunity to inhibit the growth of distal tumors, providing good potential for tumor photoimmunotherapy.


Subject(s)
5'-Nucleotidase , Adenosine Diphosphate , Adenosine , Immunotherapy , Photothermal Therapy , Animals , Humans , Mice , 5'-Nucleotidase/antagonists & inhibitors , Adenosine/chemistry , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Diphosphate/analogs & derivatives , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biomimetics/methods , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunotherapy/methods , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Photothermal Therapy/methods , Quantum Dots/chemistry , Tumor Microenvironment/drug effects , Male
16.
Nat Commun ; 15(1): 3603, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684662

ABSTRACT

The ability to sense chemical gradients and respond with directional motility and chemical activity is a defining feature of complex living systems. There is a strong interest among scientists to design synthetic systems that emulate these properties. Here, we realize and control such behaviors in a synthetic system by tailoring multivalent interactions of adenosine nucleotides with catalytic microbeads. We first show that multivalent interactions of the bead with gradients of adenosine mono-, di- and trinucleotides (AM/D/TP) control both the phoretic motion and a proton-transfer catalytic reaction, and find that both effects are diminished greatly with increasing valence of phosphates. We exploit this behavior by using enzymatic hydrolysis of ATP to AMP, which downregulates multivalent interactivity in situ. This produces a sudden increase in transport of the catalytic microbeads (a phoretic jump), which is accompanied by increased catalytic activity. Finally, we show how this enzymatic activity can be systematically tuned, leading to simultaneous in situ spatial and temporal control of the location of the microbeads, as well as the products of the reaction that they catalyze. These findings open up new avenues for utilizing multivalent interaction-mediated programming of complex chemo-mechanical behaviors into active systems.


Subject(s)
Adenosine Triphosphate , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Hydrolysis , Catalysis , Colloids/chemistry , Microspheres , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/chemistry , Adenosine/metabolism , Adenosine/chemistry
17.
ChemMedChem ; 19(13): e202400115, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38630955

ABSTRACT

The continuous deciphering of crucial biological roles of RNA modifications and their involvement in various pathological conditions, together with their key roles in the use of RNA-based therapeutics, has reignited interest in studying the occurrence and identity of non-canonical ribonucleoside structures during the past years. Discovery and structural elucidation of new modified structures is usually achieved by combination of liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) at the nucleoside level and stable isotope labeling experiments. This approach, however, has its pitfalls as demonstrated in the course of the present study: we structurally elucidated a new nucleoside structure that showed significant similarities to the family of (c)t6A modifications and was initially considered a genuine modification, but subsequently turned out to be an in vitro formed glycerol ester of t6A. This artifact is generated from ct6A during RNA hydrolysis upon addition of enzymes stored in glycerol containing buffers in a mildly alkaline milieu, and was moreover shown to undergo an intramolecular transesterification reaction. Our results demand for extra caution, not only in the discovery of new RNA modifications, but also with regard to the quantification of known modified structures, in particular chemically labile modifications, such as ct6A, that might suffer from exposure to putatively harmless reagents during the diverse steps of sample preparation.


Subject(s)
RNA , RNA/chemistry , RNA/metabolism , Esterification , Adenosine/chemistry , Adenosine/analogs & derivatives , Molecular Structure , Tandem Mass Spectrometry
18.
Biosens Bioelectron ; 256: 116276, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38599073

ABSTRACT

Fat mass and obesity-associated protein (FTO) has gained attention as the first RNA N6-methyladenosine (m6A) modification eraser due to its overexpression being associated with various cancers. In this study, an electrochemiluminescence (ECL) biosensor for the detection of demethylase FTO was developed based on DNAzyme-mediated CRISPR/Cas12a signal cascade amplification system and carboxylated carbon nitride nanosheets/phosphorus-doped nitrogen-vacancy modified carbon nitride nanosheets (C-CN/PCNV) heterojunction as the emitter. The biosensor was constructed by modifying the C-CN/PCNV heterojunction and a ferrocene-tagged probe (ssDNA-Fc) on a glassy carbon electrode. The presence of FTO removes the m6A modification on the catalytic core of DNAzyme, restoring its cleavage activity and generating activator DNA. This activator DNA further activates the trans-cleavage ability of Cas12a, leading to the cleavage of the ssDNA-Fc and the recovery of the ECL signal. The C-CN/PCNV heterojunction prevents electrode passivation and improves the electron-hole recombination, resulting in significantly enhanced ECL signal. The biosensor demonstrates high sensitivity with a low detection limit of 0.63 pM in the range from 1.0 pM to 100 nM. Furthermore, the biosensor was successfully applied to detect FTO in cancer cell lysate and screen FTO inhibitors, showing great potential in early clinical diagnosis and drug discovery.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Biosensing Techniques , CRISPR-Cas Systems , DNA, Catalytic , Electrochemical Techniques , Limit of Detection , Luminescent Measurements , Metallocenes , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Humans , DNA, Catalytic/chemistry , Electrochemical Techniques/methods , Nitriles/chemistry , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/metabolism , CRISPR-Associated Proteins/chemistry , Adenosine/analogs & derivatives , Adenosine/analysis , Adenosine/chemistry , Nanostructures/chemistry , Ferrous Compounds/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics
19.
Nucleic Acids Res ; 52(8): 4523-4540, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38477398

ABSTRACT

In archaea and eukaryotes, the evolutionarily conserved KEOPS is composed of four core subunits-Kae1, Bud32, Cgi121 and Pcc1, and a fifth Gon7/Pcc2 that is found in fungi and metazoa. KEOPS cooperates with Sua5/YRDC to catalyze the biosynthesis of tRNA N6-threonylcarbamoyladenosine (t6A), an essential modification needed for fitness of cellular organisms. Biochemical and structural characterizations of KEOPSs from archaea, yeast and humans have determined a t6A-catalytic role for Kae1 and auxiliary roles for other subunits. However, the precise molecular workings of KEOPSs still remain poorly understood. Here, we investigated the biochemical functions of A. thaliana KEOPS and determined a cryo-EM structure of A. thaliana KEOPS dimer. We show that A. thaliana KEOPS is composed of KAE1, BUD32, CGI121 and PCC1, which adopts a conserved overall arrangement. PCC1 dimerization leads to a KEOPS dimer that is needed for an active t6A-catalytic KEOPS-tRNA assembly. BUD32 participates in direct binding of tRNA to KEOPS and modulates the t6A-catalytic activity of KEOPS via its C-terminal tail and ATP to ADP hydrolysis. CGI121 promotes the binding of tRNA to KEOPS and potentiates the t6A-catalytic activity of KEOPS. These data and findings provide insights into mechanistic understanding of KEOPS machineries.


Subject(s)
Arabidopsis Proteins , Multiprotein Complexes , RNA, Plant , RNA, Transfer , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/chemistry , Cryoelectron Microscopy , Models, Molecular , Protein Binding , Protein Multimerization , RNA, Transfer/metabolism , RNA, Transfer/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Multiprotein Complexes/metabolism , RNA, Plant/chemistry , RNA, Plant/metabolism
20.
Nucleic Acids Res ; 52(10): 5950-5958, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38452198

ABSTRACT

Loss of the translational reading frame leads to misincorporation and premature termination, which can have lethal consequences. Based on structural evidence that A1503 of 16S rRNA intercalates between specific mRNA bases, we tested the possibility that it plays a role in maintenance of the reading frame by constructing ribosomes with an abasic nucleotide at position 1503. This was done by specific cleavage of 16S rRNA at position 1493 using the colicin E3 endonuclease and replacing the resulting 3'-terminal 49mer fragment with a synthetic oligonucleotide containing the abasic site using a novel splinted RNA ligation method. Ribosomes reconstituted from the abasic 1503 16S rRNA were highly active in protein synthesis but showed elevated levels of spontaneous frameshifting into the -1 reading frame. We then asked whether the residual frameshifting persisting in control ribosomes containing an intact A1503 is due to the absence of the N6-dimethyladenosine modifications at positions 1518 and 1519. Indeed, this frameshifting was rescued by site-specific methylation in vitro by the ksgA methylase. These findings thus implicate two different sites near the 3' end of 16S rRNA in maintenance of the translational reading frame, providing yet another example of a functional role for ribosomal RNA in protein synthesis.


Subject(s)
Frameshifting, Ribosomal , Protein Biosynthesis , RNA, Ribosomal, 16S , Ribosomes , RNA, Ribosomal, 16S/genetics , Ribosomes/metabolism , Ribosomes/genetics , Nucleotides/chemistry , Nucleotides/genetics , Methylation , Open Reading Frames , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/chemistry , Nucleic Acid Conformation , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...