Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.580
Filter
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042493

ABSTRACT

Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.


Subject(s)
Acetylcysteine , Cyclic AMP , Guanine Nucleotide Exchange Factors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Thionucleotides , Animals , Child , Humans , Mice , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP/therapeutic use , DNA/drug effects , Guanine Nucleotide Exchange Factors/agonists , Mice, Inbred NOD , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Male , Female , Child, Preschool , Thionucleotides/pharmacology , Thionucleotides/therapeutic use , DNA Damage , Drug Therapy, Combination
2.
Front Immunol ; 12: 753472, 2021.
Article in English | MEDLINE | ID: mdl-34899704

ABSTRACT

When compartmentally mislocalized within cells, nucleic acids can be exceptionally immunostimulatory and can even trigger the immune-mediated elimination of cancer. Specifically, the accumulation of double-stranded DNA in the cytosol can efficiently promote antitumor immunity by activating the cGAMP synthase (cGAS) / stimulator of interferon genes (STING) cellular signaling pathway. Targeting this cytosolic DNA sensing pathway with interferon stimulatory DNA (ISD) is therefore an attractive immunotherapeutic strategy for the treatment of cancer. However, the therapeutic activity of ISD is limited by several drug delivery barriers, including susceptibility to deoxyribonuclease degradation, poor cellular uptake, and inefficient cytosolic delivery. Here, we describe the development of a nucleic acid immunotherapeutic, NanoISD, which overcomes critical delivery barriers that limit the activity of ISD and thereby promotes antitumor immunity through the pharmacological activation of cGAS at the forefront of the STING pathway. NanoISD is a nanoparticle formulation that has been engineered to confer deoxyribonuclease resistance, enhance cellular uptake, and promote endosomal escape of ISD into the cytosol, resulting in potent activation of the STING pathway via cGAS. NanoISD mediates the local production of proinflammatory cytokines via STING signaling. Accordingly, the intratumoral administration of NanoISD induces the infiltration of natural killer cells and T lymphocytes into murine tumors. The therapeutic efficacy of NanoISD is demonstrated in preclinical tumor models by attenuated tumor growth, prolonged survival, and an improved response to immune checkpoint blockade therapy.


Subject(s)
DNA , Drug Delivery Systems , Nanoparticles , Nucleotidyltransferases , Animals , Female , Humans , Mice , Colonic Neoplasms/therapy , Cytokines/biosynthesis , Cytokines/genetics , DNA/administration & dosage , DNA/chemical synthesis , DNA/pharmacology , DNA/therapeutic use , Drug Screening Assays, Antitumor , Endosomes/physiology , Immunotherapy/methods , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Membrane Proteins/physiology , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/immunology , Nucleotidyltransferases/drug effects , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , Thionucleotides/pharmacology , Tumor Microenvironment/drug effects
3.
Chem Biol Interact ; 347: 109624, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34416244

ABSTRACT

Thiopurines, immunomodulating drugs used in the management of different chronic autoimmune conditions and as anti-leukemic agents, may exert in some cases gastrointestinal toxicity. Moreover, since these agents are administered orally, they are absorbed across the gastrointestinal tract epithelium. On these premises, cellular and molecular events occurring in intestinal cells may be important to understand thiopurine effects. However, quantitative information on the biotransformation of thiopurines in intestinal tissues is still limited. To shed light on biotransformation processes specific of the intestinal tissue, in this study thiopurine metabolites concentrations were analyzed by an in vitro model of human healthy colon, the HCEC cell line, upon exposure to cytotoxic concentrations of azathioprine or mercaptopurine; the investigation was carried out using an innovative mass spectrometry method, that allowed the simultaneous quantification of 11 mono-, di-, and triphosphate thionucleotides. Among the 11 metabolites evaluated, TIMP, TGMP, TGDP, TGTP, MeTIMP, MeTIDP and MeTITP were detectable in HCEC cells treated with azathioprine or mercaptopurine, considering two different incubation times before the addition of the drugs (4 and 48 h). Different associations between metabolites concentrations and cytotoxicity were detected. In particular, the cytotoxicity was dependent on the TGMP, TGDP, TGTP and MeTITP concentrations after the 4 h incubation before the addition of thiopurines. This may be an indication that, to study the association between thiopurine metabolite concentrations and the cytotoxicity activity in vitro, short growth times before treatment should be used. Moreover, for the first time our findings highlight the strong correlation between cytotoxicity and thiopurine pharmacokinetics in HCEC intestinal cells in vitro suggesting that these cells could be a suitable in vitro model for studying thiopurine intestinal cytotoxicity.


Subject(s)
Antimetabolites/pharmacology , Intestines/drug effects , Purine Nucleotides/pharmacology , Thionucleotides/pharmacology , Antimetabolites/pharmacokinetics , Antimetabolites/toxicity , Cell Count , Cell Line , Cell Survival/drug effects , Humans , Purine Nucleotides/pharmacokinetics , Purine Nucleotides/toxicity , Thionucleotides/pharmacokinetics , Thionucleotides/toxicity
4.
Gene ; 800: 145842, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34274479

ABSTRACT

Treatment of serum-starved quiescent human cells with fetal bovine serum (FBS), epidermal growth factor (EGF), or the phorbol ester (12-O-tetradecanoylphorbol-13-acetate, TPA) activates the RAS-MAPK pathway which initiates a transcriptional program which drives cells toward proliferation. Stimulation of the RAS-MAPK pathway activates mitogen- and stress-activated kinases (MSK) 1 and 2, which phosphorylate histone H3 at S10 (H3S10ph) or S28 (H3S28ph) (nucleosomal response) located at the regulatory regions of immediate-early genes, setting in motion a series of chromatin remodeling events that result in transcription initiation. To investigate immediate-early genes regulated by the MSK, we have completed transcriptome analyses (RNA sequencing) of human normal fibroblast cells (CCD-1070Sk) stimulated with EGF or TPA ± H89, a potent MSK/PKA inhibitor. The induction of many immediate-early genes was independent of MSK activity. However, the induction of immediate-early genes attenuated with H89 also had reduced induction with the PKA inhibitor, Rp-cAMPS. Several EGF-induced genes, coding for transcriptional repressors, were further upregulated with H89 but not with Rp-cAMPS, suggesting a role for MSK in modulating the induction level of these genes.


Subject(s)
Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Mitogens/pharmacology , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblasts/physiology , Gene Expression Profiling , Genes, Immediate-Early/drug effects , Humans , Isoquinolines/pharmacology , Reproducibility of Results , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thionucleotides/pharmacology
5.
Pharmacol Res Perspect ; 9(4): e00831, 2021 08.
Article in English | MEDLINE | ID: mdl-34288585

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common cancer in children and adolescents. Although the 5-year survival rate is high, some patients respond poorly to chemotherapy or have recurrence in locations such as the testis. The blood-testis barrier (BTB) can prevent complete eradication by limiting chemotherapeutic access and lead to testicular relapse unless a chemotherapeutic is a substrate of drug transporters present at this barrier. Equilibrative nucleoside transporter (ENT) 1 and ENT2 facilitate the movement of substrates across the BTB. Clofarabine is a nucleoside analog used to treat relapsed or refractory ALL. This study investigated the role of ENTs in the testicular disposition of clofarabine. Pharmacological inhibition of the ENTs by 6-nitrobenzylthioinosine (NBMPR) was used to determine ENT contribution to clofarabine transport in primary rat Sertoli cells, in human Sertoli cells, and across the rat BTB. The presence of NBMPR decreased clofarabine uptake by 40% in primary rat Sertoli cells (p = .0329) and by 53% in a human Sertoli cell line (p = .0899). Rats treated with 10 mg/kg intraperitoneal (IP) injection of the NBMPR prodrug, 6-nitrobenzylthioinosine 5'-monophosphate (NBMPR-P), or vehicle, followed by an intravenous (IV) bolus 10 mg/kg dose of clofarabine, showed a trend toward a lower testis concentration of clofarabine than vehicle (1.81 ± 0.59 vs. 2.65 ± 0.92 ng/mg tissue; p = .1160). This suggests that ENTs could be important for clofarabine disposition. Clofarabine may be capable of crossing the human BTB, and its potential use as a first-line treatment to avoid testicular relapse should be considered.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Clofarabine/pharmacokinetics , Equilibrative Nucleoside Transporter 1/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Testis/metabolism , Animals , Biological Transport , Cells, Cultured , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative-Nucleoside Transporter 2/antagonists & inhibitors , Humans , Lamivudine/blood , Lamivudine/pharmacokinetics , Lamivudine/pharmacology , Male , Rats, Sprague-Dawley , Telomerase/genetics , Thioinosine/analogs & derivatives , Thioinosine/blood , Thioinosine/pharmacokinetics , Thioinosine/pharmacology , Thionucleotides/blood , Thionucleotides/pharmacokinetics , Thionucleotides/pharmacology
6.
ACS Appl Mater Interfaces ; 13(9): 10760-10767, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33621039

ABSTRACT

Exosomes are natural delivery vehicles because of their original feature such as low immunogenicity, excellent biocompatibility, and migration capability. Engineering exosomes with appropriate ligands are effective approaches to improve the low cellular uptake efficiency of exosomes. However, current strategies face considerable challenges due to the tedious and labor-intensive operational process. Here, we designed a novel peptides-equipped exosomes platform which can be assembled under convenient and mild reaction condition. Cell-penetrating peptides (CPPs) was conjugated on HepG2 cells-derived exosomes surface which can not only enhance the penetrating capacity of exosomes but also assist exosomes in loading antisense oligonucleotides (ASOs). The cellular uptake mechanism was investigated and we compared the difference between natural exosomes and modified exosomes. The resulting nanosystem demonstrated a preferential tropism for cells that are parented to their source tumor cells and could remarkably increase the cellular delivery of G3139 with efficient downregulation of antiapoptotic Bcl-2. This work developed a rapid strategy for intracellular delivery of nucleic acids, thus providing more possibilities toward personalized cancer medicine.


Subject(s)
Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Exosomes/chemistry , Oligodeoxyribonucleotides, Antisense/pharmacology , Thionucleotides/pharmacology , Cell-Penetrating Peptides/metabolism , Down-Regulation/drug effects , Drug Carriers/metabolism , Exosomes/metabolism , Gene Silencing/drug effects , Hep G2 Cells , Humans , Oligodeoxyribonucleotides, Antisense/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Thionucleotides/genetics
7.
Int J Mol Sci ; 22(2)2021 Jan 10.
Article in English | MEDLINE | ID: mdl-33435130

ABSTRACT

Endothelial cells and astrocytes preferentially express metabotropic P2Y nucleotide receptors, which are involved in the maintenance of vascular and neural function. Among these, P2Y1 and P2Y2 receptors appear as main actors, since their stimulation induces intracellular calcium mobilization and activates signaling cascades linked to cytoskeletal reorganization. In the present work, we have analyzed, by means of atomic force microscopy (AFM) in force spectroscopy mode, the mechanical response of human umbilical vein endothelial cells (HUVEC) and astrocytes upon 2MeSADP and UTP stimulation. This approach allows for simultaneous measurement of variations in factors such as Young's modulus, maximum adhesion force and rupture event formation, which reflect the potential changes in both the stiffness and adhesiveness of the plasma membrane. The largest effect was observed in both endothelial cells and astrocytes after P2Y2 receptor stimulation with UTP. Such exposure to UTP doubled the Young's modulus and reduced both the adhesion force and the number of rupture events. In astrocytes, 2MeSADP stimulation also had a remarkable effect on AFM parameters. Additional studies performed with the selective P2Y1 and P2Y13 receptor antagonists revealed that the 2MeSADP-induced mechanical changes were mediated by the P2Y13 receptor, although they were negatively modulated by P2Y1 receptor stimulation. Hence, our results demonstrate that AFM can be a very useful tool to evaluate functional native nucleotide receptors in living cells.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Astrocytes/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2/metabolism , Thionucleotides/metabolism , Uridine Triphosphate/metabolism , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Astrocytes/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Microscopy, Atomic Force , Signal Transduction , Thionucleotides/pharmacology , Uridine Triphosphate/pharmacology
8.
Biochim Biophys Acta Rev Cancer ; 1875(2): 188500, 2021 04.
Article in English | MEDLINE | ID: mdl-33385484

ABSTRACT

Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.


Subject(s)
Clusterin/genetics , Clusterin/metabolism , Neoplasms/metabolism , Clusterin/antagonists & inhibitors , Drug Synergism , Drug Therapy , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Thionucleotides/pharmacology , Thionucleotides/therapeutic use
9.
Retrovirology ; 17(1): 27, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859220

ABSTRACT

BACKGROUND: Adult T-cell leukemia lymphoma (ATLL) is a chemotherapy-resistant malignancy with a median survival of less than one year that will afflict between one hundred thousand and one million individuals worldwide who are currently infected with human T-cell leukemia virus type 1. Recurrent somatic mutations in host genes have exposed the T-cell receptor pathway through nuclear factor κB to interferon regulatory factor 4 (IRF4) as an essential driver for this malignancy. We sought to determine if IRF4 represents a therapeutic target for ATLL and to identify downstream effectors and biomarkers of IRF4 signaling in vivo. RESULTS: ATLL cell lines, particularly Tax viral oncoprotein-negative cell lines, that most closely resemble ATLL in humans, were sensitive to dose- and time-dependent inhibition by a next-generation class of IRF4 antisense oligonucleotides (ASOs) that employ constrained ethyl residues that mediate RNase H-dependent RNA degradation. ATLL cell lines were also sensitive to lenalidomide, which repressed IRF4 expression. Both ASOs and lenalidomide inhibited ATLL proliferation in vitro and in vivo. To identify biomarkers of IRF4-mediated CD4 + T-cell expansion in vivo, transcriptomic analysis identified several genes that encode key regulators of ATLL, including interleukin 2 receptor subunits α and ß, KIT ligand, cytotoxic T-lymphocyte-associated protein 4, and thymocyte selection-associated high mobility group protein TOX 2. CONCLUSIONS: These data support the pursuit of IRF4 as a therapeutic target in ATLL with the use of either ASOs or lenalidomide.


Subject(s)
HTLV-I Infections/metabolism , Interferon Regulatory Factors/metabolism , Leukemia-Lymphoma, Adult T-Cell/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Products, tax/metabolism , HTLV-I Infections/drug therapy , HTLV-I Infections/pathology , Human T-lymphotropic virus 1 , Humans , Interferon Regulatory Factors/genetics , Lenalidomide/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Mice , Oligonucleotides, Antisense/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Thionucleotides/pharmacology
10.
Int J Mol Sci ; 21(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580317

ABSTRACT

A new approach to improve the effectiveness of acute myeloid leukemia (AML) treatment is to use the properties of purinergic signaling molecules secreted into the bone marrow milieu in response to leukemic cell growth. Therefore, our study aimed to evaluate the effects of extracellular adenine nucleotides and adenosine on the growth and death parameters in the leukemic THP-1 cell line. Cells were exposed to ATP, ADP, AMP, adenosine and nonhydrolyzable analogues of ATP and ADP (ATPγS and ADPßS) in a 1-1000 µM broad concentration range. The basal mRNA expression of the P1 and P2 receptors was evaluated by real-time PCR. Changes in the processes of cell growth and death were assessed by flow cytometry analysis of proliferation, cell cycle and apoptosis. Chemotaxis toward stromal cell-derived factor-1 (SDF-1) was performed using the modified Boyden chamber assay, and chemokine receptor type 4 (CXCR4) surface expression was quantified by flow cytometry. We indicated several antileukemic actions. High micromolar concentrations (100-1000 µM) of extracellular adenine nucleotides and adenosine inhibit the growth of cells by arresting the cell cycle and/or inducing apoptosis. ATP is characterized by the highest potency and widest range of effects, and is responsible for the cell cycle arrest and the apoptosis induction. Compared to ATP, the effect of ADP is slightly weaker. Adenosine mostly has a cytotoxic effect, with the induction of apoptosis. The last studied nucleotide, AMP, demonstrated only a weak cytotoxic effect without affecting the cell cycle. In addition, cell migration towards SDF-1 was inhibited by low micromolar concentrations (10 µM). One of the reasons for this action of ATPγS and adenosine was a reduction in CXCR4 surface expression, but this only partially explains the mechanism of antimigratory action. In summary, extracellular adenine nucleotides and adenosine inhibit THP-1 cell growth, cause death of cells and modulate the functioning of the SDF-1/CXCR4 axis. Thus, they negatively affect the processes that are responsible for the progression of AML and the difficulties in AML treatment.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adenosine/pharmacology , Leukemia, Myeloid, Acute/pathology , Thionucleotides/pharmacology , Affinity Labels , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Tumor Cells, Cultured
11.
Int J Mol Sci ; 21(11)2020 May 30.
Article in English | MEDLINE | ID: mdl-32486261

ABSTRACT

Platelet G protein-coupled receptors (GPCRs) regulate platelet function by mediating the response to various agonists, including adenosine diphosphate (ADP), thromboxane A2, and thrombin. Although GPCR kinases (GRKs) are considered to have the crucial roles in most GPCR functions, little is known regarding the regulation of GPCR signaling and mechanisms of GPCR desensitization by GRKs in platelets. In this study, we investigated the functional role of GRK6 and the molecular basis for regulation of specific GPCR desensitization by GRK6 in platelets. We used GRK6 knockout mice to evaluate the functional role of GRK6 in platelet activation. Platelet aggregation, dense- and -granule secretion, and fibrinogen receptor activation induced by 2-MeSADP, U46619, thrombin, and AYPGKF were significantly potentiated in GRK6-/- platelets compared to the wild-type (WT) platelets. However, collagen-related peptide (CRP)-induced platelet aggregation and secretion were not affected in GRK6-/- platelets. Interestingly, platelet aggregation induced by co-stimulation of serotonin and epinephrine which activate Gq-coupled 5HT2A and Gz-coupled 2A adrenergic receptors, respectively, was not affected in GRK6-/- platelets, suggesting that GRK6 was involved in specific GPCR regulation. In addition, platelet aggregation in response to the second challenge of ADP and AYPGKF was restored in GRK6-/- platelets whereas re-stimulation of the agonist failed to induce aggregation in WT platelets, indicating that GRK6 contributed to P2Y1, P2Y12, and PAR4 receptor desensitization. Furthermore, 2-MeSADP-induced Akt phosphorylation and AYPGKF-induced Akt, extracellular signal-related kinase (ERK), and protein kinase Cδ (PKC) phosphorylation were significantly potentiated in GRK6-/- platelets. Finally, GRK6-/- mice exhibited an enhanced and stable thrombus formation after FeCl3 injury to the carotid artery and shorter tail bleeding times, indicating that GRK6-/- mice were more susceptible to thrombosis and hemostasis. We conclude that GRK6 plays an important role in regulating platelet functional responses and thrombus formation through selective GPCR desensitization.


Subject(s)
Blood Platelets/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression Regulation , Platelet Activation , Receptors, G-Protein-Coupled/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Female , Hemostatics , Male , Mice , Mice, Knockout , Oligopeptides/pharmacology , Phosphorylation , Platelet Aggregation , Thionucleotides/pharmacology , Thrombin/metabolism , Thromboxane A2/metabolism
12.
Eur Neurol ; 83(2): 195-212, 2020.
Article in English | MEDLINE | ID: mdl-32474563

ABSTRACT

INTRODUCTION: Spinal cord injury (SCI) causes most severe motor and sensory dysfunctions. In Chinese traditional medicine, the agonist of a purinergic receptor is believed to have a positive effect on SCIs, and 2-Methylthio-adenosine-5'-diphosphate (2-MesADP) is a selective agonist of the P2Y purinergic receptor. METHODS: To investigate its therapeutic function and molecular mechanism in SCI, transcriptome analysis associated with weighted gene co-expression network analysis (WGCNA) was carried out at various time points after T9 crush injury. RESULTS: 2-MesADP demonstrated recovery of limb motor function at the 6 weeks after injury, accompanied by neuronal regeneration and axon remyelination at 2 and 6 weeks. Furthermore, gene profiling revealed alternated gene expression with the treatment of 2-MesADP. These genes were assigned to a total of 38 modules, followed by gene ontology analysis; of these, 18 represented neuronal apoptosis and regeneration, immune response, synaptic transmission, cell cycle, and angiogenesis. In the neuronal apoptosis and regeneration module, Nefh, NeuroD6, and Dcx in the 2-MesADP group were noticed due to their interesting expression pattern. The gene expression patterns of Mag, Mog, and Cnp, which played key roles in myelination, were significantly changed with the treatment of 2-MesADP. Wnt signal pathway was the most important pathway in 2-MesADP treatment for acute SCI. CONCLUSION: 2-MesADP enhanced locomotor recovery in mouse SCI by altering the expression of neuronal apoptosis and remyelination-related genes and Wnt signaling pathways.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Gene Expression Regulation/drug effects , Locomotion/physiology , Purinergic Agonists/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries , Thionucleotides/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Doublecortin Protein , Humans , Mice , Nerve Regeneration/drug effects , Recovery of Function/physiology , Remyelination/drug effects , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
13.
Purinergic Signal ; 16(1): 73-84, 2020 03.
Article in English | MEDLINE | ID: mdl-32067141

ABSTRACT

ATP is a cotransmitter released with other neurotransmitters from sympathetic nerves, where it stimulates purinergic receptors. Purinergic adenosine P1 receptors (coupled to Gi/o proteins) produce sympatho-inhibition in several autonomic effectors by prejunctional inhibition of neurotransmitter release. Similarly, signalling through P2Y12 and P2Y13 receptors coupled to Gi/o proteins is initiated by the ATP breakdown product ADP. Hence, this study has pharmacologically investigated a possible role of ADP-induced inhibition of the cardioaccelerator sympathetic drive in pithed rats, using a stable ADP analogue (ADPßS) and selective antagonists for the purinergic P2Y1, P2Y12 and P2Y13 receptors. Accordingly, male Wistar rats were pithed and: (i) pretreated i.v. with gallamine (25 mg/kg) and desipramine (50 µg/kg) for preganglionic spinal (C7-T1) stimulation of the cardioaccelerator sympathetic drive (n = 78); or (ii) prepared for receiving i.v. injections of exogenous noradrenaline (n = 12). The i.v. continuous infusions of ADPßS (10 and 30 µg/kg/min) dose-dependently inhibited the tachycardic responses to electrical sympathetic stimulation, but not those to exogenous noradrenaline. The cardiac sympatho-inhibition produced by 30 µg/kg/min ADPßS was (after i.v. administration of compounds) (i) unchanged by 1-ml/kg bidistilled water or 300-µg/kg MRS 2500 (P2Y1 receptor antagonist), (ii) abolished by 300-µg/kg PSB 0739 (P2Y12 receptor antagonist) and (iii) partially blocked by 3000-µg/kg MRS 2211 (P2Y13 receptor antagonist). Our results suggest that ADPßS induces a cardiac sympatho-inhibition that mainly involves the P2Y12 receptor subtype and, probably to a lesser extent, the P2Y13 receptor subtype. These receptors may represent therapeutic targets for treating cardiovascular pathologies, including stroke and myocardial infarctions.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Cardiovascular Physiological Phenomena/drug effects , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2/metabolism , Sympathetic Nervous System/physiology , Thionucleotides/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Male , Rats , Rats, Wistar , Sympathetic Nervous System/drug effects
14.
FEBS Lett ; 594(9): 1389-1402, 2020 05.
Article in English | MEDLINE | ID: mdl-31985825

ABSTRACT

Rap1 is a small GTPase that has been implicated in dendritic development and plasticity. In this study, we investigated the role of Rap1 in axonal growth and its activation in response to neurotrophins and myelin-associated inhibitors. We report that Rap1 is activated by brain-derived neurotrophic factor and that this activation can be blocked by myelin-associated glycoprotein (MAG) or central nervous system myelin, which also induced increases in Rap1GAP1 levels. In addition, we demonstrate that adenoviral overexpression of Rap1 enhances neurite outgrowth in the presence of MAG and myelin, while inhibition of Rap1 activity through overexpression of Rap1GAP1 blocks neurite outgrowth. These findings suggest that Rap1GAP1 negatively regulates neurite outgrowth, making it a potential therapeutic target to promote axonal regeneration.


Subject(s)
GTP Phosphohydrolases/metabolism , Myelin-Associated Glycoprotein/metabolism , Neuronal Outgrowth/physiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Bucladesine/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , GTP Phosphohydrolases/genetics , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Myelin Sheath/metabolism , Nerve Tissue Proteins , Neuronal Outgrowth/drug effects , Rats, Long-Evans , Thionucleotides/pharmacology , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism
15.
Life Sci Alliance ; 3(1)2020 01.
Article in English | MEDLINE | ID: mdl-31882444

ABSTRACT

Epigenetic variation reflects the impact of a dynamic environment on chromatin. However, it remains elusive how environmental factors influence epigenetic events. Here, we show that G protein-coupled receptors (GPCRs) alter H3K4 methylation via oscillatory intracellular cAMP. Activation of Gs-coupled receptors caused a rapid decrease of H3K4me3 by elevating cAMP, whereas stimulation of Gi-coupled receptors increased H3K4me3 by diminishing cAMP. H3K4me3 gradually recovered towards baseline levels after the removal of GPCR ligands, indicating that H3K4me3 oscillates in tandem with GPCR activation. cAMP increased intracellular labile Fe(II), the cofactor for histone demethylases, through a non-canonical cAMP target-Rap guanine nucleotide exchange factor-2 (RapGEF2), which subsequently enhanced endosome acidification and Fe(II) release from the endosome via vacuolar H+-ATPase assembly. Removing Fe(III) from the media blocked intracellular Fe(II) elevation after stimulation of Gs-coupled receptors. Iron chelators and inhibition of KDM5 demethylases abolished cAMP-mediated H3K4me3 demethylation. Taken together, these results suggest a novel function of cAMP signaling in modulating histone demethylation through labile Fe(II).


Subject(s)
Cyclic AMP/analogs & derivatives , Demethylation/drug effects , Ferrous Compounds/metabolism , Histones/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Thionucleotides/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Gene Silencing , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Ligands , Methylation/drug effects , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Rats , Receptors, G-Protein-Coupled/metabolism , Schwann Cells , Thionucleotides/pharmacology , Transfection
16.
J Neuroendocrinol ; 31(12): e12809, 2019 12.
Article in English | MEDLINE | ID: mdl-31715031

ABSTRACT

An injection of unesterified oestradiol (E2 ) facilitates receptive behaviour in E2 benzoate (EB)-primed, ovariectomised female rats when it is administered i.c.v. or systemically. The present study tested the hypothesis that inhibitors of protein kinase A (PKA), protein kinase G (PKG) or the Src/mitogen-activated protein kinase (MAPK) complex interfere with E2 facilitation of receptive behaviour. In Experiment 1, lordosis induced by i.c.v. infusion of E2 was significantly reduced by i.c.v. administration of Rp-cAMPS, a PKA inhibitor, KT5823, a PKG inhibitor, and PP2 and PD98059, Src and MAPK inhibitors, respectively, between 30 and 240 minutes after infusion. In Experiment 2, we determined whether the ventromedial hypothalamus (VMH) is one of the neural sites at which those intracellular pathways participate in lordosis behaviour induced by E2 . Administration of each of the four protein kinase inhibitors into the VMH blocked facilitation of lordosis induced by infusion of E2 also into the VMH. These data support the hypothesis that activation of several protein kinase pathways is involved in the facilitation of lordosis by E2 in EB-primed rats.


Subject(s)
Estrogen Antagonists/pharmacology , Lordosis/physiopathology , Protein Kinase Inhibitors/pharmacology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Carbazoles/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Estradiol/physiology , Female , Flavonoids/pharmacology , Infusions, Intraventricular , Lordosis/chemically induced , Male , Microinjections , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/pharmacology , Rats , Thionucleotides/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects
17.
Reprod Domest Anim ; 54(12): 1612-1620, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31549441

ABSTRACT

We aimed to elucidate whether NO acts in in vitro sperm capacitation in bovine via cGMP/PKG1 pathway. For this, cryopreserved bovine sperm were capacitated in vitro with 20 µg/ml heparin (Control) plus treatments: 1 mM L-arginine (L-arg, NO precursor), 50 µM Rp-8-Bromo-ß-phenyl-1,N2 -ethenoguanosine-3',5'-cyclic monophosphorothioate (Rp-8-Br-cGMPS, selective inhibitor of the binding site for cGMP in PKG1), 1 mM 2-Phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (PTIO, NO scavenger), and the combinations of L-arg + RP-8-Br-cGMPS and L-arg + PTIO. Sperm motility and vigour were determined by phase-contrast microscopy, capacitation status by chlortetracycline staining, and the intracellular concentration of cGMP was measured by ELISA. Data were subjected to analysis of variance and means compared with SNK test at 5% probability. Motility and vigour were lower in sperm treated with PTIO when compared to Control and other treatments (p < .05). The L-arg treatment showed the highest percentage of capacitated sperm when compared to the Control and other treatments (Rp-8-Br-cGMPS, L-arg + Rp-8-Br-cGMPS and PTIO) (69.8 ± 3.4%, 51.2 ± 3.0, 51.1 ± 2.1, 51.2 ± 3.0 and 45.5 ± 2.7, respectively) (p < .05). The capacitation ratio (%) was lower in treatments with Rp-8-Br-cGMPS, L-arg + Rp-8-Br-cGMPS and PTIO, respectively (p < .05). Lastly, cGMP concentration (pmol/ml) was lower in PTIO and L-arg + PTIO (1.3 ± 0.3 and 1.6 ± 0.4) and was higher in Rp-8-Br-cGMPS and L-arg + Rp-8-Br-cGMPS (3.7 ± 0.4 and 4.0 ± 0.5) treatments. We showed that during in vitro capacitation of cattle: (a) NO influences sperm motility and vigour; (b) NO is associated with cGMP synthesis through two independent pathways and (c) the cGMP/PKG1 pathway has a partial role in sperm capacitation and does not involve the L-arg/NO.


Subject(s)
Cyclic GMP/physiology , Nitric Oxide/pharmacology , Sperm Capacitation/drug effects , Sperm Motility/drug effects , Animals , Arginine/pharmacology , Cattle , Cryopreservation/veterinary , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic N-Oxides/pharmacology , Heparin/pharmacology , Imidazoles/pharmacology , Male , Spermatozoa/drug effects , Spermatozoa/physiology , Thionucleotides/pharmacology
18.
J Cell Physiol ; 234(10): 17473-17481, 2019 08.
Article in English | MEDLINE | ID: mdl-30825199

ABSTRACT

Advanced glycation end products (AGE) and angiotensin II were closely correlated with the progression of diabetic nephopathy (DN). Nitric oxide (NO) is a protective mediator of renal tubular hypertrophy in DN. Here, we examined the molecular mechanisms of angiotensin-converting enzyme inhibitor (ACEI) and NO signaling responsible for diminishing AGE-induced renal tubular hypertrophy. In human renal proximal tubular cells, AGE decreased NO production, inducible NOS activity, guanosine 3',5'-cyclic monophosphate (cGMP) synthesis, and cGMP-dependent protein kinase (PKG) activation. All theses effects of AGE were reversed by treatment with ACEIs (captopril and enalapril), the NO donor S-nitroso-N-acetylpenicillamine (SNAP), and the PKG activator 8-para-chlorophenylthio-cGMPs (8-pCPT-cGMPs). In addition, AGE-enhanced activation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) were clearly reduced by captopril, enalapril, SNAP, and 8-pCPT-cGMPs. The abilities of ACEIs and NO/PKG activation to inhibit AGE-induced hypertrophic growth were verified by the observation that captopril, enalapril, SNAP, and 8-pCPT-cGMPs decreased protein levels of fibronectin, p21 Waf1/Cip1 , and receptor for AGE. The results of the present study suggest that ACEIs significantly reduced AGE-increased ERK/JNK/p38 MAPK activation and renal tubular hypertrophy partly through enhancement of the NO/PKG pathway.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Glycation End Products, Advanced/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Nitric Oxide/metabolism , Captopril/pharmacology , Cell Enlargement/drug effects , Cell Line , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Enalapril/pharmacology , Enzyme Activation/drug effects , Glycation End Products, Advanced/toxicity , Humans , Hypertrophy/prevention & control , Kidney Tubules, Proximal/pathology , MAP Kinase Signaling System/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type II/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Signal Transduction/drug effects , Thionucleotides/pharmacology
19.
Neurosci Lett ; 699: 177-183, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30753912

ABSTRACT

Axon regeneration after cerebral ischemia in mammals is inadequate to restore function, illustrating the need to design better strategies for improving outcomes. Improvement of axon regeneration has been achieved through fastigial nucleus electrostimulation (FNS) in animal researches. However, the mechanisms underlying this neuroprotection remain poorly understood. Increasing the levels of the second messenger cyclic AMP (cAMP) enhances axon regeneration, making it an excellent candidate molecule that has therapeutic potential. In the present study, we examined the expression of cAMP signaling in ischemic brain tissues following focal cerebral ischemia. Adult rats were subjected to ischemia induced by middle cerebral artery occlusion (MCAO). A dipolar electrode was placed into the cerebellum to stimulate the cerebellar fastigial nucleus for 1 h after ischemia. Neurological deficits and the expressions of cAMP, PKA (protein kinase A) and ROCK (Rho-kinase) were determined. Axonal regeneration was measured by upregulation of growth-associated protein 43 (GAP43). The data indicated that FNS significantly enhanced axonal regeneration and motor function recovery after cerebral ischemia. FNS also significantly increased cAMP and PKA levels after ischemic brain injury. All the beneficial effects of FNS were blocked by Rp-cAMP, an antagonist of PKA. Our research suggested that the axonal regeneration conferred by FNS was likely achieved via the regulation of cAMP/PKA pathway.


Subject(s)
Cerebellar Nuclei/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Electric Stimulation Therapy , Infarction, Middle Cerebral Artery/therapy , Nerve Regeneration , Signal Transduction , Animals , Cerebellar Nuclei/drug effects , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , GAP-43 Protein/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Nerve Regeneration/drug effects , Rats , Recovery of Function/drug effects , Thionucleotides/pharmacology , Up-Regulation , rho-Associated Kinases/biosynthesis
20.
Biomaterials ; 197: 182-193, 2019 03.
Article in English | MEDLINE | ID: mdl-30660994

ABSTRACT

Antisense oligonucleotides (ASOs) usually contain a fully phosphorothioate (PS) backbone, which possibly interact with many genes and proteins under intracellular conditions. G3139 is an ASO that targets Bcl-2 mRNA and induces cell apoptosis. Here, we report a kind of cytidinyl-lipid combined with a cationic lipid (DNCA/CLD, molar ration, 28:3, named mix), which may interact with oligonucleotides via H-bond formation, pi-stacking and electrostatic interaction, accompanied by low zeta potentials. The IC50 value of G3139 delivered by mix-lipid reduced from above 20 µM to 0.158 µM for MCF-7/ADR, and exhibited stronger antiproliferation upon other cancer cell lines. In addition, PS modification in the 3'-half of G3139 (especially at positions 13-16) enhanced serum stability, target specificity and anticancer activity. Also, a locked nucleic acid (LNA) gapmer G3139 (LNA-G3139) showed superior antiproliferation (78.5%) and Bcl-2 mRNA suppression effects (85.5%) at 200 nM, mainly due to its high complementary RNA affinity. More apoptosis-associated targets were identified, and a lower level of non-specific protein binding (HSA) revealed that both antisense and aptamer mechanisms might simultaneously exist. A combination of a new delivery system and chemical modifications, such as in LNA-G3139, may have potential clinical application prospects in the future.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Cytidine/analogs & derivatives , Drug Carriers/chemistry , Thionucleotides/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Delivery Systems , Female , Humans , Lipids/chemistry , MCF-7 Cells , Mice, Inbred BALB C , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Thionucleotides/pharmacokinetics , Thionucleotides/pharmacology , Thionucleotides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...