Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.186
Filter
1.
Mil Med ; 189(Supplement_3): 268-275, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160853

ABSTRACT

INTRODUCTION: Non-compressible torso hemorrhagic (NCTH) shock is the leading cause of potentially survivable trauma on the battlefield. New hypotensive drug therapies are urgently required to resuscitate and protect the heart and brain following NCTH. Our aim was to examine the strengths and limitations of permissive hypotension and discuss the development of small-volume adenosine, lidocaine, and Mg2+ (ALM) fluid resuscitation in rats and pigs. MATERIALS AND METHODS: For review of permissive hypotension, a literature search was performed from inception up to November 2023 using PubMed, Cochrane, and Embase databases, with inclusion of animal studies, clinical trials and reviews with military and clinical relevance. For the preclinical study, adult female pigs underwent laparoscopic liver resection. After 30 minutes of bleeding, animals were resuscitated with 4 mL/kg 3% NaCl ± ALM bolus followed 60 minutes later with 4 h 3 mL/kg/h 0.9% NaCl ± ALM drip (n = 10 per group), then blood transfusion. Mean arterial pressure (MAP) and cardiac output (CO) were continuously measured via a left ventricular pressure catheter and pulmonary artery catheter, respectively. Systemic vascular resistance (SVR) was calculated using the formula: 80 × (MAP - CVP)/CI. Oxygen delivery was calculated as the product of CO and arterial oxygen content. RESULTS: Targeting a MAP of ∼50 mmHg can be harmful or beneficial, depending on how CO and SVR are regulated. A theoretical example shows that for the same MAP of 50 mmHg, a higher CO and lower SVR can lead to a nearly 2-fold increase in O2 supply. We further show that in animal models of NCTH, 3% NaCl ALM bolus and 0.9% NaCl ALM drip induce a hypotensive, high flow, vasodilatory state with maintained tissue O2 supply and neuroprotection. ALM therapy increases survival by resuscitating the heart, reducing internal bleeding by correcting coagulopathy, and decreasing secondary injury. CONCLUSIONS: In rat and pig models of NCTH, small-volume ALM therapy resuscitates at hypotensive pressures by increasing CO and reducing SVR. This strategy is associated with heart and brain protection and maintained tissue O2 delivery. Translational studies are required to determine reproducibility and optimal component dosing. ALM therapy may find wide utility in prehospital and far-forward military environments.


Subject(s)
Adenosine , Hypotension , Resuscitation , Animals , Swine , Resuscitation/methods , Rats , Hypotension/etiology , Hypotension/physiopathology , Adenosine/administration & dosage , Adenosine/pharmacology , Lidocaine/pharmacology , Lidocaine/therapeutic use , Lidocaine/administration & dosage , Female , Shock, Hemorrhagic/therapy , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/physiopathology
2.
Int J Rheum Dis ; 27(8): e15297, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39175261

ABSTRACT

BACKGROUND: Ferroptosis is caused by iron-dependent peroxidation of membrane phospholipids and chondrocyte ferroptosis contributes to osteoarthritis (OA) progression. Glutathione peroxidase 4 (GPX4) plays a master role in blocking ferroptosis. N6-methyladenosine (m6A) is an epigenetic modification among mRNA post-transcriptional modifications. This study investigated the effect of methyltransferase-like 14 (METTL14), the key component of the m6A methyltransferase, on chondrocyte ferroptosis via m6A modification. METHODS: An OA rat model was established through an intra-articular injection of monosodium iodoacetate in the right knee. OA cartilages in rat models were used for gene expression analysis. Primary mouse chondrocytes or ADTC5 cells were stimulated with IL-1ß or erastin. The m6A RNA methylation quantification kit was used to measure m6A level. The effect of METTL14 and GPX4 on ECM degradation and ferroptosis was investigated through western blotting, fluorescence immunostaining, propidium iodide staining, and commercially available kits. The mechanism of METTL14 action was explored through MeRIP-qPCR assays. RESULTS: METTL14 and m6A expression was upregulated in osteoarthritic cartilages and IL-1ß-induced chondrocytes. METTL14 depletion repressed the IL-1ß or erastin-stimulated ECM degradation and ferroptosis in mouse chondrocytes. METTL14 inhibited GPX4 gene through m6A methylation modification. GPX4 knockdown reversed the si-METTL14-mediated protection in IL-1ß-induced chondrocytes. CONCLUSION: METTL14 depletion inhibits ferroptosis and ECM degradation by suppressing GPX4 mRNA m6A modification in injured chondrocytes.


Subject(s)
Chondrocytes , Ferroptosis , Methyltransferases , Phospholipid Hydroperoxide Glutathione Peroxidase , Animals , Chondrocytes/drug effects , Chondrocytes/pathology , Chondrocytes/metabolism , Chondrocytes/enzymology , Ferroptosis/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Mice , Male , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/enzymology , Osteoarthritis/genetics , Osteoarthritis/chemically induced , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cells, Cultured , Disease Models, Animal , Rats , Humans , Rats, Sprague-Dawley
3.
J Nat Prod ; 87(8): 1983-1993, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39136667

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive cancer that has an effect on human health. As a first-line drug for HCC, despite its excellent efficacy, lenvatinib (Len) is prone to developing drug resistance in HCC patients. The N6-methyladenosine (m6A) modification is not only related to the development of HCC but also shows great potential in overcoming HCC resistance. Using Dot Blot, our group first screened a small molecule m6A regulator, lobeline (Lob), from a library of 390 compounds (mostly natural products). In vitro experiments demonstrated that Lob could significantly enhance the sensitivity to Len of Len-resistant HCC (HCC/Len) and inhibit migration of resistant cells. In Len-resistant cell-derived and patient-derived xenograft models, Lob could reverse the resistant phenotype, with reductions in tumor volume of 68% and 60%, respectively. Furthermore, MeRIP-m6A sequencing results indicated that the underlying molecular mechanism of Lob reversal of HCC drug resistance was related to UBE3B. Taken together, this study highlighted that Lob, a plant derived natural product, could reverse the resistance of HCC to Len by regulating the m6A levels. It is hoped that this will provide a pharmacological research basis for the clinical treatment of HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Drug Resistance, Neoplasm , Liver Neoplasms , Phenylurea Compounds , Quinolines , Quinolines/pharmacology , Quinolines/chemistry , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Humans , Drug Resistance, Neoplasm/drug effects , Animals , Phenylurea Compounds/pharmacology , Mice , Cell Line, Tumor , Adenosine/analogs & derivatives , Adenosine/pharmacology , Molecular Structure , Biological Products/pharmacology , Biological Products/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
4.
J Cell Mol Med ; 28(16): e70019, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39164798

ABSTRACT

Knee osteoarthritis (KOA) is a chronic joint disease that significantly affects the health of the elderly. As an herbal remedy, Gubi decoction (GBD) has been traditionally used for the treatment of osteoarthritis-related syndromes. However, the anti-KOA efficacy and mechanism of GBD remain unclear. This study aimed to experimentally investigate the anti-KOA efficacy and the underlying mechanism of GBD. The medial meniscus (DMM) mice model and IL-1ß-stimulated chondrocytes were, respectively, constructed as in vivo and in vitro models of KOA to evaluate the osteoprotective effect and molecular mechanism of GBD. The UPLC-MS/MS analysis showed that GBD mainly contained pinoresinol diglucoside, rehmannioside D, hesperidin, liquiritin, baohuoside I, glycyrrhizic acid, kaempferol and tangeretin. Animal experiment showed that GBD could alleviate articular cartilage destruction and recover histopathological alterations in DMM mice. In addition, GBD inhibited chondrocyte apoptosis and restored DMM-induced dysregulated autophagy evidenced by the upregulation of ATG7 and LC3 II/LC3 I but decreased P62 level. Mechanistically, METTL3-mediated m6A modification decreased the expression of ATG7 in DMM mice, as it could be significantly attenuated by GBD. METTL3 overexpression significantly counteracted the protective effect of GBD on chondrocyte autophagy. Further research showed that GBD promoted proteasome-mediated ubiquitination degradation of METLL3. Our findings suggest that GBD could act as a protective agent against KOA. The protective effect of GBD may result from its promotion on chondrocyte autophagy by suppressing METTL3-dependent ATG7 m6A methylation.


Subject(s)
Autophagy-Related Protein 7 , Autophagy , Chondrocytes , Methyltransferases , Osteoarthritis, Knee , Animals , Chondrocytes/metabolism , Chondrocytes/drug effects , Autophagy/drug effects , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/drug therapy , Mice , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein 7/genetics , Methyltransferases/metabolism , Methylation/drug effects , Male , Drugs, Chinese Herbal/pharmacology , Disease Models, Animal , Apoptosis/drug effects , Mice, Inbred C57BL , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/metabolism , Humans , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/pathology
5.
Biomolecules ; 14(7)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062531

ABSTRACT

DZNep (3-deazaneplanocin A) is commonly used to reduce lysine methylation. DZNep inhibits S-adenosyl-l-homocysteine hydrolase (AHCY), preventing the conversion of S-adenosyl-l-homocysteine (SAH) into L-homocysteine. As a result, the SAM-to-SAH ratio decreases, an indicator of the methylation potential within a cell. Many studies have characterized the impact of DZNep on histone lysine methylation or in specific cell or disease contexts, but there has yet to be a study looking at the potential downstream impact of DZNep treatment on proteins other than histones. Recently, protein thermal stability has provided a new dimension for studying the mechanism of action of small-molecule inhibitors. In addition to ligand binding, post-translational modifications and protein-protein interactions impact thermal stability. Here, we sought to characterize the protein thermal stability changes induced by DZNep treatment in HEK293T cells using the Protein Integral Solubility Alteration (PISA) assay. DZNep treatment altered the thermal stability of 135 proteins, with over half previously reported to be methylated at lysine residues. In addition to thermal stability, we identify changes in transcript and protein abundance after DZNep treatment to distinguish between direct and indirect impacts on thermal stability. Nearly one-third of the proteins with altered thermal stability had no changes at the transcript or protein level. Of these thermally altered proteins, CDK6 had a stabilized methylated peptide, while its unmethylated counterpart was unaltered. Multiple methyltransferases were among the proteins with thermal stability alteration, including DNMT1, potentially due to changes in the SAM/SAH levels. This study systematically evaluates DZNep's impact on the transcriptome, the proteome, and the thermal stability of proteins.


Subject(s)
Adenosine , Protein Stability , Humans , HEK293 Cells , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Protein Stability/drug effects , Methylation , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/metabolism , Temperature
6.
J Mater Sci Mater Med ; 35(1): 43, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073623

ABSTRACT

Collagen hydrogel has been shown promise as an inducer for chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), contributing to the repair of cartilage defects. However, the precise molecular mechanism underlying this phenomenon remains poorly elucidated. Here, we induced chondrogenic differentiation of BMSCs using collagen hydrogel and identified 4451 differentially expressed genes (DEGs) through transcriptomic sequencing. Our analysis revealed that DEGs were enriched in the focal adhesion pathway, with a notable decrease in expression levels in the collagen hydrogel group compared to the control group. Protein-protein interaction network analysis suggested that actinin alpha 1 (ACTN1) and actinin alpha 4 (ACTN4), two proteins also involved in cytoskeletal recombination, may be crucial in collagen hydrogel-induced chondrogenic differentiation of BMSCs. Additionally, we found that N6-methyladenosine RNA methylation (m6A) modification was involved in collagen hydrogel-mediated chondrogenic differentiation, with fat mass and obesity-associated protein (FTO) implicated in regulating the expression of ACTN1 and ACTN4. These findings suggest that collagen hydrogel might regulate focal adhesion and actin cytoskeletal signaling pathways through down-regulation of ACTN1 and ACTN4 mRNA via FTO-mediated m6A modification, ultimately driving chondrogenic differentiation of BMSCs. In conclusion, our study provides valuable insights into the molecular mechanisms of collagen hydrogel-induced chondrogenic differentiation of BMSCs, which may aid in developing more effective strategies for cartilage regeneration.


Subject(s)
Cell Differentiation , Chondrogenesis , Collagen , Gene Expression Profiling , Hydrogels , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Chondrogenesis/drug effects , Chondrogenesis/genetics , Cell Differentiation/drug effects , Hydrogels/chemistry , Collagen/chemistry , Animals , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Transcriptome/drug effects , Actinin/metabolism , Actinin/genetics , Cells, Cultured , Methylation , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Rats
7.
EMBO Rep ; 25(8): 3547-3573, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009832

ABSTRACT

The COVID-19 pandemic reminded us of the urgent need for new antivirals to control emerging infectious diseases and potential future pandemics. Immunotherapy has revolutionized oncology and could complement the use of antivirals, but its application to infectious diseases remains largely unexplored. Nucleoside analogs are a class of agents widely used as antiviral and anti-neoplastic drugs. Their antiviral activity is generally based on interference with viral nucleic acid replication or transcription. Based on our previous work and computer modeling, we hypothesize that antiviral adenosine analogs, like remdesivir, have previously unrecognized immunomodulatory properties which contribute to their therapeutic activity. In the case of remdesivir, we here show that these properties are due to its metabolite, GS-441524, acting as an Adenosine A2A Receptor antagonist. Our findings support a new rationale for the design of next-generation antiviral agents with dual - immunomodulatory and intrinsic - antiviral properties. These compounds could represent game-changing therapies to control emerging viral diseases and future pandemics.


Subject(s)
Adenosine Monophosphate , Adenosine , Alanine , Antiviral Agents , COVID-19 , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Humans , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/chemistry , COVID-19/immunology , COVID-19/virology , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/therapeutic use , Pandemics , COVID-19 Drug Treatment , Chlorocebus aethiops , Virus Replication/drug effects , Vero Cells , Betacoronavirus/drug effects , Betacoronavirus/immunology , Receptor, Adenosine A2A/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology
8.
Bioorg Chem ; 150: 107530, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38852310

ABSTRACT

The Asp-tRNAAsn/Glu-tRNAGln amidotransferase (GatCAB) has been proposed as a novel antibacterial drug target due to its indispensability in prominent human pathogens. While several inhibitors with in vitro activity have been identified, none have been demonstrated to have potent activity against live bacteria. In this work, seven non-hydrolyzable transition state mimics of GatCAB were synthesized and tested as the transamidase inhibitors against GatCAB from the human pathogen Helicobacter pylori. Notably, the methyl sulfone analog of glutamyl-adenosine significantly reduced GatCAB's transamination rate. Additionally, four lipid-conjugates of these mimics displayed antibacterial activity against Bacillus subtilis, likely due to enhanced cell permeability. Inhibitory activity against GatCAB in live bacteria was confirmed using a sensitive gain-of-function dual luciferase reporter in Mycobacterium bovis-BCG. Only the lipid-conjugated methyl sulfone analog exhibited a significant increase in mistranslation rate, highlighting its cell permeability and inhibitory potential. This study provides insights for developing urgently needed novel antibacterial agents amidst emerging antimicrobial drug resistance.


Subject(s)
Anti-Bacterial Agents , Enzyme Inhibitors , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Bacillus subtilis/drug effects , Molecular Structure , Dose-Response Relationship, Drug , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Adenosine/chemical synthesis , Helicobacter pylori/drug effects , Helicobacter pylori/enzymology , Nitrogenous Group Transferases/antagonists & inhibitors , Nitrogenous Group Transferases/metabolism , Humans
9.
J Physiol Sci ; 74(1): 33, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867187

ABSTRACT

Hibernation and torpor are not passive responses caused by external temperature drops and fasting but are active brain functions that lower body temperature. A population of neurons in the preoptic area was recently identified as such active torpor-regulating neurons. We hypothesized that the other hypothermia-inducing maneuvers would also activate these neurons. To test our hypothesis, we first refined the previous observations, examined the brain regions explicitly activated during the falling phase of body temperature using c-Fos expression, and confirmed the preoptic area. Next, we observed long-lasting hypothermia by reactivating torpor-tagged Gq-expressing neurons using the activity tagging and DREADD systems. Finally, we found that about 40-60% of torpor-tagged neurons were activated by succeeding isoflurane anesthesia and by icv administration of an adenosine A1 agonist. Isoflurane-induced and central adenosine-induced hypothermia is, at least in part, an active process mediated by the torpor-regulating neurons in the preoptic area.


Subject(s)
Adenosine , Isoflurane , Neurons , Preoptic Area , Animals , Preoptic Area/drug effects , Preoptic Area/metabolism , Isoflurane/pharmacology , Isoflurane/administration & dosage , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Body Temperature/drug effects , Body Temperature/physiology , Hypothermia/chemically induced , Hypothermia/metabolism , Torpor/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism
10.
Biol Pharm Bull ; 47(6): 1113-1118, 2024.
Article in English | MEDLINE | ID: mdl-38839362

ABSTRACT

Motile cilia in the ependymal cells that line the brain ventricles play pivotal roles in cerebrospinal fluid (CSF) flow in well-defined directions. However, the substances and pathways which regulate their beating have not been well studied. Here, we used primary cultured cells derived from neonatal mouse brain that possess motile cilia and found that adenosine (ADO) stimulates ciliary beating by increasing the ciliary beat frequency (CBF) in a concentration-dependent manner, with the ED50 value being 5 µM. Ciliary beating stimulated by ADO was inhibited by A2B receptor (A2BR) antagonist MRS1754 without any inhibition by antagonists of other ADO receptor subtypes. The expression of A2BR on the cilia was also confirmed by immunofluorescence. The values of CBF were also increased by forskolin, which is an activator of adenylate cyclase, whereas they were not further increased by the addition of ADO. Furthermore, ciliary beating was not stimulated by ADO in the presence of a protein kinase A (PKA) inhibitors. These results altogether suggest that ADO stimulates ciliary beating through A2BR on the cilia, and activation of PKA.


Subject(s)
Adenosine , Animals, Newborn , Brain , Cilia , Cyclic AMP-Dependent Protein Kinases , Receptor, Adenosine A2B , Animals , Cilia/drug effects , Cilia/metabolism , Cilia/physiology , Receptor, Adenosine A2B/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Adenosine/pharmacology , Brain/metabolism , Brain/drug effects , Mice , Cells, Cultured , Signal Transduction/drug effects , Adenosine A2 Receptor Antagonists/pharmacology , Colforsin/pharmacology , Ependyma/metabolism , Ependyma/cytology
11.
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
12.
Phytomedicine ; 130: 155537, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38823344

ABSTRACT

BACKGROUND: Aberrant activation of autophagy in triple-negative breast cancer (TNBC) has led researchers to investigate potential therapeutic strategies targeting this process. The regulation of autophagy is significantly influenced by METTL3. Our previous research has shown that the Panax ginseng-derived compound, 20(R)-panaxatriol (PT), has potential as an anti-tumor agent. However, it remains unclear whether PT can modulate autophagy through METTL3 to exert its anti-tumor effects. OBJECTIVE: Our objective is to investigate whether PT can regulate autophagy in TNBC cells and elucidate the molecular mechanisms. STUDY DESIGN: For in vitro experiments, we employed SUM-159-PT and MDA-MB-231 cells. While in vivo experiments involved BALB/c nude mice and NOD/SCID mice. METHODS: In vitro, TNBC cells were treated with PT, and cell lines with varying expression levels of METTL3 were established. We assessed the impact on tumor cell activity and autophagy by analyzing autophagic flux, Western Blot (WB), and methylation levels. In vivo, subcutaneous transplantation models were established in BALB/c nude and NOD/SCID mice to observe the effect of PT on TNBC growth. HE staining and immunofluorescence were employed to analyze histopathological changes in tumor tissues. MeRIP-seq and dual-luciferase reporter gene assays were used to identify key downstream targets. Additionally, the silencing of STIP1 Homology And U-Box Containing Protein 1 (STUB1) explored PT's effects. The mechanism of PT's action on STUB1 via METTL3 was elucidated through mRNA stability assays, mRNA alternative splicing analysis, and nuclear-cytoplasmic mRNA separation. RESULTS: In both in vivo and in vitro experiments, it was discovered that PT significantly upregulates the expression of METTL3, leading to autophagy inhibition and therapeutic effects in TNBC. Simultaneously, through MeRIP-seq analysis and dual-luciferase reporter gene assays, we have demonstrated that PT modulates STUB1 via METTL3, influencing autophagy in TNBC cells. Furthermore, intriguingly, PT extends the half-life of STUB1 mRNA by enhancing its methylation modification, thereby enhancing its stability. CONCLUSION: In summary, our research reveals that PT increases STUB1 m6A modification through a METTL3-mediated mechanism in TNBC cells, inhibiting autophagy and further accentuating its anti-tumor properties. Our study provides novel mechanistic insights into TNBC pathogenesis and potential drug targets for TNBC.


Subject(s)
Autophagy , Methyltransferases , Mice, Inbred BALB C , Mice, Nude , Triple Negative Breast Neoplasms , Ubiquitin-Protein Ligases , Animals , Triple Negative Breast Neoplasms/drug therapy , Humans , Autophagy/drug effects , Female , Cell Line, Tumor , Methyltransferases/metabolism , Ubiquitin-Protein Ligases/metabolism , Mice, SCID , Mice, Inbred NOD , Mice , Antineoplastic Agents, Phytogenic/pharmacology , Xenograft Model Antitumor Assays , Panax/chemistry , Adenosine/analogs & derivatives , Adenosine/pharmacology
13.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892291

ABSTRACT

Bone regeneration remains a significant clinical challenge, often necessitating surgical approaches when healing bone defects and fracture nonunions. Within this context, the modulation of adenosine signaling pathways has emerged as a promising therapeutic option, encouraging osteoblast activation and tempering osteoclast differentiation. A literature review of the PubMed database with relevant keywords was conducted. The search criteria involved in vitro or in vivo models, with clear methodological descriptions. Only studies that included the use of indirect adenosine agonists, looking at the effects of bone regeneration, were considered relevant according to the eligibility criteria. A total of 29 articles were identified which met the inclusion and exclusion criteria, and they were reviewed to highlight the preclinical translation of adenosine agonists. While preclinical studies demonstrate the therapeutic potential of adenosine signaling in bone regeneration, its clinical application remains unrealized, underscoring the need for further clinical trials. To date, only large, preclinical animal models using indirect adenosine agonists have been successful in stimulating bone regeneration. The adenosine receptors (A1, A2A, A2B, and A3) stimulate various pathways, inducing different cellular responses. Specifically, indirect adenosine agonists act to increase the extracellular concentration of adenosine, subsequently agonizing the respective adenosine receptors. The agonism of each receptor is dependent on its expression on the cell surface, the extracellular concentration of adenosine, and its affinity for adenosine. This comprehensive review analyzed the multitude of indirect agonists currently being studied preclinically for bone regeneration, discussing the mechanisms of each agonist, their cellular responses in vitro, and their effects on bone formation in vivo.


Subject(s)
Bone Regeneration , Purinergic P1 Receptor Agonists , Receptors, Purinergic P1 , Bone Regeneration/drug effects , Humans , Animals , Receptors, Purinergic P1/metabolism , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/metabolism , Signal Transduction/drug effects , Translational Research, Biomedical
14.
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
15.
Nutrients ; 16(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892725

ABSTRACT

Xanthohumol (Xn) is an antioxidant flavonoid mainly extracted from hops (Humulus lupulus), one of the main ingredients of beer. As with other bioactive compounds, their therapeutic potential against different diseases has been tested, one of which is Alzheimer's disease (AD). Adenosine is a neuromodulatory nucleoside that acts through four different G protein-coupled receptors: A1 and A3, which inhibit the adenylyl cyclases (AC) pathway, and A2A and A2B, which stimulate this activity, causing either a decrease or an increase, respectively, in the release of excitatory neurotransmitters such as glutamate. This adenosinergic pathway, which is altered in AD, could be involved in the excitotoxicity process. Therefore, the aim of this work is to describe the effect of Xn on the adenosinergic pathway using cell lines. For this purpose, two different cellular models, rat glioma C6 and human neuroblastoma SH-SY5Y, were exposed to a non-cytotoxic 10 µM Xn concentration. Adenosine A1 and A2A, receptor levels, and activities related to the adenosine pathway, such as adenylate cyclase, protein kinase A, and 5'-nucleotidase, were analyzed. The adenosine A1 receptor was significantly increased after Xn exposure, while no changes in A2A receptor membrane levels or AC activity were reported. Regarding 5'-nucleotidases, modulation of their activity by Xn was noted since CD73, the extracellular membrane attached to 5'-nucleotidase, was significantly decreased in the C6 cell line. In conclusion, here we describe a novel pathway in which the bioactive flavonoid Xn could have potentially beneficial effects on AD as it increases membrane A1 receptors while modulating enzymes related to the adenosine pathway in cell cultures.


Subject(s)
Adenosine , Flavonoids , Glioma , Humulus , Neuroblastoma , Propiophenones , Receptor, Adenosine A1 , Humans , Flavonoids/pharmacology , Rats , Propiophenones/pharmacology , Animals , Adenosine/metabolism , Adenosine/pharmacology , Cell Line, Tumor , Humulus/chemistry , Neuroblastoma/metabolism , Neuroblastoma/drug therapy , Glioma/metabolism , Glioma/drug therapy , Receptor, Adenosine A1/metabolism , Signal Transduction/drug effects , Adenylyl Cyclases/metabolism , Receptor, Adenosine A2A/metabolism
16.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928239

ABSTRACT

Aging (senescence) is an unavoidable biological process that results in visible manifestations in all cutaneous tissues, including scalp skin and hair follicles. Previously, we evaluated the molecular function of adenosine in promoting alopecia treatment in vitro. To elucidate the differences in the molecular mechanisms between minoxidil (MNX) and adenosine, gene expression changes in dermal papilla cells were examined. The androgen receptor (AR) pathway was identified as a candidate target of adenosine for hair growth, and the anti-androgenic activity of adenosine was examined in vitro. In addition, ex vivo examination of human hair follicle organ cultures revealed that adenosine potently elongated the anagen stage. According to the severity of alopecia, the ratio of the two peaks (terminal hair area/vellus hair area) decreased continuously. We further investigated the adenosine hair growth promoting effect in vivo to examine the hair thickness growth effects of topical 5% MNX and the adenosine complex (0.75% adenosine, 1% penthenol, and 2% niacinamide; APN) in vivo. After 4 months of administration, both the MNX and APN group showed significant increases in hair density (MNX + 5.01% (p < 0.01), APN + 6.20% (p < 0.001)) and thickness (MNX + 5.14% (p < 0.001), APN + 10.32% (p < 0.001)). The inhibition of AR signaling via adenosine could have contributed to hair thickness growth. We suggest that the anti-androgenic effect of adenosine, along with the evaluation of hair thickness distribution, could help us to understand hair physiology and to investigate new approaches for drug development.


Subject(s)
Adenosine , Alopecia , Hair Follicle , Hair , Minoxidil , Receptors, Androgen , Signal Transduction , Alopecia/drug therapy , Alopecia/metabolism , Alopecia/pathology , Humans , Male , Receptors, Androgen/metabolism , Adenosine/metabolism , Adenosine/pharmacology , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/growth & development , Signal Transduction/drug effects , Minoxidil/pharmacology , Female , Animals , Hair/growth & development , Hair/drug effects , Hair/metabolism
17.
Oncogene ; 43(31): 2405-2420, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38914663

ABSTRACT

Gemcitabine resistance is a major obstacle to the effectiveness of chemotherapy in pancreatic ductal adenocarcinoma (PDAC). Therefore, new strategies are needed to sensitize cancer cells to gemcitabine. Here, we constructed gemcitabine-resistant PDAC cells and analyzed them with RNA-sequence. Employing an integrated approach involving bioinformatic analyses from multiple databases, TGFB2 is identified as a crucial gene in gemcitabine-resistant PDAC and is significantly associated with poor gemcitabine therapeutic response. The patient-derived xenograft (PDX) model further substantiates the gradual upregulation of TGFB2 expression during gemcitabine-induced resistance. Silencing TGFB2 expression can enhance the chemosensitivity of gemcitabine against PDAC. Mechanistically, TGFB2, post-transcriptionally stabilized by METTL14-mediated m6A modification, can promote lipid accumulation and the enhanced triglyceride accumulation drives gemcitabine resistance by lipidomic profiling. TGFB2 upregulates the lipogenesis regulator sterol regulatory element binding factor 1 (SREBF1) and its downstream lipogenic enzymes via PI3K-AKT signaling. Moreover, SREBF1 is responsible for TGFB2-mediated lipogenesis to promote gemcitabine resistance in PDAC. Importantly, TGFB2 inhibitor imperatorin combined with gemcitabine shows synergistic effects in gemcitabine-resistant PDAC PDX model. This study sheds new light on an avenue to mitigate PDAC gemcitabine resistance by targeting TGFB2 and lipid metabolism and develops the potential of imperatorin as a promising chemosensitizer in clinical translation.


Subject(s)
Adenosine , Carcinoma, Pancreatic Ductal , Deoxycytidine , Drug Resistance, Neoplasm , Gemcitabine , Lipid Metabolism , Pancreatic Neoplasms , Transforming Growth Factor beta2 , Humans , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/genetics , Drug Resistance, Neoplasm/genetics , Animals , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Mice , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Metabolic Reprogramming , Sterol Regulatory Element Binding Protein 1
18.
Cell Mol Life Sci ; 81(1): 276, 2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38909325

ABSTRACT

N6-methyladenosine (m6A) is one of the most prevalent and conserved RNA modifications. It controls several biological processes, including the biogenesis and function of circular RNAs (circRNAs), which are a class of covalently closed-single stranded RNAs. Several studies have revealed that proteotoxic stress response induction could be a relevant anticancer therapy in Acute Myeloid Leukemia (AML). Furthermore, a strong molecular interaction between the m6A mRNA modification factors and the suppression of the proteotoxic stress response has emerged. Since the proteasome inhibition leading to the imbalance in protein homeostasis is strictly linked to the stress response induction, we investigated the role of Bortezomib (Btz) on m6A regulation and in particular its impact on the modulation of m6A-modified circRNAs expression. Here, we show that treating AML cells with Btz downregulated the expression of the m6A regulator WTAP at translational level, mainly because of increased oxidative stress. Indeed, Btz treatment promoted oxidative stress, with ROS generation and HMOX-1 activation and administration of the reducing agent N-acetylcysteine restored WTAP expression. Additionally, we identified m6A-modified circRNAs modulated by Btz treatment, including circHIPK3, which is implicated in protein folding and oxidative stress regulation. These results highlight the intricate molecular networks involved in oxidative and ER stress induction in AML cells following proteotoxic stress response, laying the groundwork for future therapeutic strategies targeting these pathways.


Subject(s)
Adenosine , Leukemia, Myeloid, Acute , Oxidative Stress , RNA, Circular , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/drug therapy , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Oxidative Stress/drug effects , Bortezomib/pharmacology , Cell Line, Tumor , Reactive Oxygen Species/metabolism , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Protein Serine-Threonine Kinases , Intracellular Signaling Peptides and Proteins
19.
J Hypertens ; 42(6): 1027-1038, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38690904

ABSTRACT

OBJECTIVE: Reno-renal reflexes are disturbed in cardiovascular and hypertensive conditions when elevated levels of pro-inflammatory mediators/cytokines are present within the kidney. We hypothesised that exogenously administered inflammatory cytokines tumour necrosis factor alpha (TNF-α) and interleukin (IL)-1ß modulate the renal sympatho-excitatory response to chemical stimulation of renal pelvic sensory nerves. METHODS: In anaesthetised rats, intrarenal pelvic infusions of vehicle [0.9% sodium chloride (NaCl)], TNF-α (500 and 1000 ng/kg) and IL-1ß (1000 ng/kg) were maintained for 30 min before chemical activation of renal pelvic sensory receptors was performed using randomized intrarenal pelvic infusions of hypertonic NaCl, potassium chloride (KCl), bradykinin, adenosine and capsaicin. RESULTS: The increase in renal sympathetic nerve activity (RSNA) in response to intrarenal pelvic hypertonic NaCl was enhanced during intrapelvic TNF-α (1000 ng/kg) and IL-1ß infusions by almost 800% above vehicle with minimal changes in mean arterial pressure (MAP) and heart rate (HR). Similarly, the RSNA response to intrarenal pelvic adenosine in the presence of TNF-α (500 ng/kg), but not IL-1ß, was almost 200% above vehicle but neither MAP nor HR were changed. There was a blunted sympatho-excitatory response to intrapelvic bradykinin in the presence of TNF-α (1000 ng/kg), but not IL-1ß, by almost 80% below vehicle, again without effect on either MAP or HR. CONCLUSION: The renal sympatho-excitatory response to renal pelvic chemoreceptor stimulation is modulated by exogenous TNF-α and IL-1ß. This suggests that inflammatory mediators within the kidney can play a significant role in modulating the renal afferent nerve-mediated sympatho-excitatory response.


Subject(s)
Interleukin-1beta , Kidney , Sympathetic Nervous System , Tumor Necrosis Factor-alpha , Animals , Interleukin-1beta/pharmacology , Rats , Kidney/innervation , Kidney/drug effects , Male , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Rats, Sprague-Dawley , Heart Rate/drug effects , Bradykinin/pharmacology , Reflex/drug effects , Blood Pressure/drug effects , Adenosine/administration & dosage , Adenosine/pharmacology , Saline Solution, Hypertonic/administration & dosage , Saline Solution, Hypertonic/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL