Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.812
Filter
1.
BMJ Open Diabetes Res Care ; 12(3)2024 May 06.
Article in English | MEDLINE | ID: mdl-38719505

ABSTRACT

INTRODUCTION: There has been increasing evidence that the gut microbiota is closely related to type 2 diabetes (T2D). Metformin (Met) is often used in combination with saxagliptin (Sax) and repaglinide (Rep) for the treatment of T2D. However, little is known about the effects of these combination agents on gut microbiota in T2D. RESEARCH DESIGN AND METHODS: A T2D mouse model induced by a high-fat diet (HFD) and streptozotocin (STZ) was employed. The T2D mice were randomly divided into six groups, including sham, Met, Sax, Rep, Met+Sax and Met+Rep, for 4 weeks. Fasting blood glucose level, serum biochemical index, H&E staining of liver, Oil red O staining of liver and microbiota analysis by 16s sequencing were used to access the microbiota in the fecal samples. RESULTS: These antidiabetics effectively prevented the development of HFD/STZ-induced high blood glucose, and the combination treatment had a better effect in inhibiting lipid accumulation. All these dosing regimens restored the decreasing ratio of the phylum Bacteroidetes: Firmicutes, and increasing abundance of phylum Desulfobacterota, expect for Met. At the genus level, the antidiabetics restored the decreasing abundance of Muribaculaceae in T2D mice, but when Met was combined with Rep or Sax, the abundance of Muribaculaceae was decreased. The combined treatment could restore the reduced abundance of Prevotellaceae_UCG-001, while Met monotherapy had no such effect. In addition, the reduced Lachnospiraceae_NK4A136_group was well restored in the combination treatment groups, and the effect was much greater than that in the corresponding monotherapy group. Therefore, these dosing regimens exerted different effects on the composition of gut microbiota, which might be associated with the effect on T2D. CONCLUSIONS: Supplementation with specific probiotics may further improve the hypoglycemic effects of antidiabetics and be helpful for the development of new therapeutic drugs for T2D.


Subject(s)
Adamantane , Blood Glucose , Carbamates , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diet, High-Fat , Dipeptides , Gastrointestinal Microbiome , Hypoglycemic Agents , Metformin , Piperidines , Animals , Gastrointestinal Microbiome/drug effects , Metformin/pharmacology , Metformin/therapeutic use , Mice , Diet, High-Fat/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/microbiology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Carbamates/pharmacology , Dipeptides/pharmacology , Male , Adamantane/analogs & derivatives , Adamantane/pharmacology , Adamantane/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Blood Glucose/analysis , Blood Glucose/drug effects , Mice, Inbred C57BL , Drug Therapy, Combination , Streptozocin
2.
Chem Commun (Camb) ; 60(36): 4810-4813, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38602391

ABSTRACT

The non-canonical amino acid adamantylglycine (Ada) is introduced into peptides to allow high-affinity binding to cucurbit[7]uril (CB7). Introduction of Ada into a cell-penetrating peptide (CPP) sequence had minimal influence on the membrane transport, yet enabled up- and down-regulation of the membrane transport activity.


Subject(s)
Cell-Penetrating Peptides , Glycine , Heterocyclic Compounds, 2-Ring , Imidazolidines , Macrocyclic Compounds , Glycine/chemistry , Glycine/analogs & derivatives , Glycine/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Imidazoles/chemistry , Humans , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/metabolism , Adamantane/chemistry , Adamantane/analogs & derivatives , Cell Membrane/metabolism , Cell Membrane/chemistry , Biological Transport
3.
Phys Chem Chem Phys ; 26(17): 13420-13431, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38647171

ABSTRACT

Autoimmune inflammatory diseases, such as rheumatoid arthritis (RA) and ulcerative colitis, are associated with an uncontrolled production of cytokines leading to the pronounced inflammatory response of these disorders. Their therapy is currently focused on the inhibition of cytokine receptors, such as the Janus kinase (JAK) protein family. Tofacitinib and peficitinib are JAK inhibitors that have been recently approved to treat rheumatoid arthritis. In this study, an in-depth analysis was carried out through quantum biochemistry to understand the interactions involved in the complexes formed by JAK1 and tofacitinib or peficitinib. Computational analyses provided new insights into the binding mechanisms between tofacitinib or peficitinib and JAK1. The essential amino acid residues that support the complex are also identified and reported. Additionally, we report new interactions, such as van der Waals; hydrogen bonds; and alkyl, pi-alkyl, and pi-sulfur forces, that stabilize the complexes. The computational results revealed that peficitinib presents a similar affinity to JAK1 compared to tofacitinib based on their interaction energies.


Subject(s)
Adamantane/analogs & derivatives , Janus Kinase 1 , Niacinamide , Niacinamide/analogs & derivatives , Piperidines , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Piperidines/therapeutic use , Niacinamide/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 1/chemistry , Humans , Quantum Theory , Autoimmune Diseases/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Hydrogen Bonding , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Adamantane/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Molecular Docking Simulation
4.
Molecules ; 29(5)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38474457

ABSTRACT

This study presents a simple and energy-efficient self-assembly LAG synthetic method for novel water-soluble copper(I) complexes [Cu(terpy)(PTA)][PF6] (1) and [Cu(terpy)(PTA)2][PF6] (2). They were characterized by FT-IR, 1H, and 31P{1H} NMR spectroscopy, elemental analysis, and single-crystal/powder X-ray diffraction (for 2). The X-ray analysis of compound 2 indicates a bidentate coordination mode of terpyridine to the metal center. Variable-temperature NMR tests indicate dynamic properties for terpyridine in the case of both compounds, as well as for the PTA ligands in the case of 2. Additionally, compounds 1 and 2 exhibit interesting cytotoxic activity, which was tested on normal human dermal fibroblasts (NHDFs), human lung carcinoma (A549), human breast adenocarcinoma (MCF-7), and human cervix carcinoma (HeLa) established cell lines. In comparison to the other tested compounds, complexes 1 and 2 seem to have significantly lower IC50 values against cancer cells (A549, HeLa, MCF-7), indicating their potential as prospective anticancer agents. Moreover, both compounds show no significant toxicity towards normal skin cells (NHDFs), suggesting a certain selectivity in their action on cancer cells. Cisplatin as a reference compound also exhibited considerable cytotoxicity against cancer cells but with a low level of selectivity, which could lead to unwanted effects on normal cells. Remarkably, compounds 1 and 2 exhibit up to 30 times the cytotoxic activity of cisplatin, with a six-fold lower toxicity to normal cells. They also interact strongly with human serum albumin, suggesting potential therapeutic applications. Overall, these compounds hold significant promise as potential chemotherapeutic agents.


Subject(s)
Adamantane/analogs & derivatives , Antineoplastic Agents , Carcinoma , Coordination Complexes , Organophosphorus Compounds , Female , Humans , Cisplatin/pharmacology , Copper/chemistry , Cell Line, Tumor , Water , Prospective Studies , Spectroscopy, Fourier Transform Infrared , Coordination Complexes/chemistry , Antineoplastic Agents/pharmacology , Ligands
5.
Int Immunopharmacol ; 132: 111931, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38547769

ABSTRACT

Peficitinib is a selective Janus kinase (JAK3) inhibitor recently developed and approved for the treatment of rheumatoid arthritis in Japan. Glycolysis in macrophages could induce NOD-like receptor (NLR) family and pyrin domain-containing protein 3 (NLRP3) inflammasome activation, thus resulting in pyroptosis and acute lung injury (ALI). The aim of our study was to investigate whether Peficitinib could alleviate lipopolysaccharide (LPS)-induced ALI by inhibiting NLRP3 inflammasome activation. Wild type C57BL/6J mice were intraperitoneally injected with Peficitinib (5 or 10 mg·kg-1·day-1) for 7 consecutive days before LPS injection. The results showed that Peficitinib pretreatment significantly relieved LPS-induced pulmonary edema, inflammation, and apoptosis. NLRP3 inflammasome and glycolysis in murine lung tissues challenged with LPS were also blocked by Peficitinib. Furthermore, we found that the activation of JAK3/signal transducer and activator of transcription 3 (STAT3) was also suppressed by Peficitinib in mice with ALI. However, in Jak3 knockout mice, Peficitinib did not show obvious protective effects after LPS injection. In vitro experiments further showed that Jak3 overexpression completely abolished Peficitinib-elicited inhibitory effects on pyroptosis and glycolysis in LPS-induced RAW264.7 macrophages. Finally, we unveiled that LPS-induced activation of JAK3/STAT3 was mediated by toll-like receptor 4 (TLR4) in RAW264.7 macrophages. Collectively, our study proved that Peficitinib could protect against ALI by blocking JAK3-mediated glycolysis and pyroptosis in macrophages, which may serve as a promising candidate against ALI in the future.


Subject(s)
Acute Lung Injury , Adamantane/analogs & derivatives , Glycolysis , Janus Kinase 3 , Lipopolysaccharides , Mice, Inbred C57BL , Niacinamide , Niacinamide/analogs & derivatives , STAT3 Transcription Factor , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Janus Kinase 3/metabolism , Janus Kinase 3/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Glycolysis/drug effects , Mice , Signal Transduction/drug effects , Male , Niacinamide/pharmacology , Niacinamide/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Knockout , Acrylamides/pharmacology , Acrylamides/therapeutic use , Inflammasomes/metabolism , Pyroptosis/drug effects , Lung/pathology , Lung/drug effects , Lung/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology
7.
Int J Mol Sci ; 25(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38396999

ABSTRACT

Fibrosis is a chronic pathology resulting from excessive deposition of extracellular matrix components that leads to the loss of tissue function. Pulmonary fibrosis can follow a variety of diverse insults including ischemia, respiratory infection, or exposure to ionizing radiation. Consequently, treatments that attenuate the development of debilitating fibrosis are in desperate need across a range of conditions. Sphingolipid metabolism is a critical regulator of cell proliferation, apoptosis, autophagy, and pathologic inflammation, processes that are all involved in fibrosis. Opaganib (formerly ABC294640) is the first-in-class investigational drug targeting sphingolipid metabolism for the treatment of cancer and inflammatory diseases. Opaganib inhibits key enzymes in sphingolipid metabolism, including sphingosine kinase-2 and dihydroceramide desaturase, thereby reducing inflammation and promoting autophagy. Herein, we demonstrate in mouse models of lung damage following exposure to ionizing radiation that opaganib significantly improved long-term survival associated with reduced lung fibrosis, suppression of granulocyte infiltration, and reduced expression of IL-6 and TNFα at 180 days after radiation. These data further demonstrate that sphingolipid metabolism is a critical regulator of fibrogenesis, and specifically show that opaganib suppresses radiation-induced pulmonary inflammation and fibrosis. Because opaganib has demonstrated an excellent safety profile during clinical testing in other diseases (cancer and COVID-19), the present studies support additional clinical trials with this drug in patients at risk for pulmonary fibrosis.


Subject(s)
Adamantane/analogs & derivatives , Medical Countermeasures , Neoplasms , Pneumonia , Pulmonary Fibrosis , Pyridines , Mice , Animals , Humans , Sphingolipids/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Fibrosis , Inflammation/drug therapy
8.
Biochem Biophys Res Commun ; 703: 149685, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38373381

ABSTRACT

Ciliary beating in the airway epithelium plays an important role in preventing infection by eliminating small particles and pathogens. Stimulation of ß2 adrenergic receptor (ß2AR) increases [cAMP]i levels and strongly activates this ciliary beating. ß2AR is localized to the apical membrane of the airways by indirectly binding to ezrin, an actin-binding protein. Ezrin takes active phosphorylated and inactive dephosphorylated states at Thr-567. Previously we showed that procaterol-stimulated ciliary beating was impaired in the ezrin-knockdown mice. In this study, we examined the roles of ezrin and its phosphorylation in regulating ciliary beating by using NSC305787, an ezrin inhibitor, in normal human airway epithelial cells (NHBE). We found that NSC305787 inhibits the phosphorylation of ezrin with an IC50 of 50 µM in NHBE. Treatment with NSC305787 for 4 h or more decreased the expression of ß2AR in the cell membrane and induced vesicle- or dot-like expression of ezrin and ß2AR inside the cell. As a result, inhibition of ezrin phosphorylation by NSC305787 attenuated the effect of procaterol-induced activation of ciliary beating in both frequency and distance indices.


Subject(s)
Adamantane/analogs & derivatives , Cilia , Cytoskeletal Proteins , Procaterol , Quinolines , Mice , Humans , Animals , Cilia/metabolism , Procaterol/pharmacology , Procaterol/metabolism , Phosphorylation
9.
Diabetes Obes Metab ; 26(5): 1670-1677, 2024 May.
Article in English | MEDLINE | ID: mdl-38297915

ABSTRACT

AIM: To compare the efficacy and safety of saxagliptin/dapagliflozin and insulin glargine in people with latent autoimmune diabetes in adults (LADA). METHODS: In this phase 2b multicentre, open-label, comparator-controlled, parallel-group, non-inferiority study, we randomly assigned 33 people with LADA who had a fasting C-peptide concentration ≥0.2 nmol/L (0.6 ng/mL) to receive 1-year daily treatment with either the combination of saxagliptin (5 mg) plus dapagliflozin (10 mg) or insulin glargine (starting dose: 10 IU), both on top of metformin. The primary outcome was the 2-h mixed meal-stimulated C-peptide area under the curve (AUC), measured 12 months after randomization. Secondary outcomes were glycated haemoglobin (HbA1c) levels, change in body mass index (BMI), and hypoglycaemic events. RESULTS: In the modified intention-to-treat analysis, the primary outcome was similar in participants assigned to saxagliptin/dapagliflozin or to insulin glargine (median C-peptide AUC: 152.0 ng*min/mL [95% confidence interval {CI} 68.2; 357.4] vs. 122.2 ng*min/mL [95% CI 84.3; 255.8]; p for noninferiority = 0.0087). Participants randomized to saxagliptin/dapagliflozin lost more weight than those randomized to insulin glargine (median BMI change at the end of the study: -0.4 kg/m2 [95% CI -1.6; -0.3] vs. +0.4 kg/m2 [95% CI -0.3; +1.1]; p = 0.0076). No differences in HbA1c or in the number of participants experiencing hypoglycaemic events were found. CONCLUSIONS: Saxagliptin/dapagliflozin was non-inferior to glargine in terms of ß-cell function in this 12-month, small, phase 2b study, enrolling people with LADA with still viable endogenous insulin production. Weight loss was greater with saxagliptin/dapagliflozin, with no differences in glycaemic control or hypoglycaemic risk.


Subject(s)
Adamantane/analogs & derivatives , Benzhydryl Compounds , Diabetes Mellitus, Type 2 , Dipeptides , Glucosides , Latent Autoimmune Diabetes in Adults , Metformin , Adult , Humans , Insulin Glargine/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin , C-Peptide , Pilot Projects , Blood Glucose , Treatment Outcome , Hypoglycemic Agents/adverse effects , Metformin/therapeutic use , Double-Blind Method , Drug Therapy, Combination
10.
J Clin Invest ; 134(5)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38227384

ABSTRACT

Early-life seizures (ELSs) can cause permanent cognitive deficits and network hyperexcitability, but it is unclear whether ELSs induce persistent changes in specific neuronal populations and whether these changes can be targeted to mitigate network dysfunction. We used the targeted recombination of activated populations (TRAP) approach to genetically label neurons activated by kainate-induced ELSs in immature mice. The ELS-TRAPed neurons were mainly found in hippocampal CA1, remained uniquely susceptible to reactivation by later-life seizures, and displayed sustained enhancement in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated (AMPAR-mediated) excitatory synaptic transmission and inward rectification. ELS-TRAPed neurons, but not non-TRAPed surrounding neurons, exhibited enduring decreases in Gria2 mRNA, responsible for encoding the GluA2 subunit of the AMPARs. This was paralleled by decreased synaptic GluA2 protein expression and heightened phosphorylated GluA2 at Ser880 in dendrites, indicative of GluA2 internalization. Consistent with increased GluA2-lacking AMPARs, ELS-TRAPed neurons showed premature silent synapse depletion, impaired long-term potentiation, and impaired long-term depression. In vivo postseizure treatment with IEM-1460, an inhibitor of GluA2-lacking AMPARs, markedly mitigated ELS-induced changes in TRAPed neurons. These findings show that enduring modifications of AMPARs occur in a subpopulation of ELS-activated neurons, contributing to synaptic dysplasticity and network hyperexcitability, but are reversible with early IEM-1460 intervention.


Subject(s)
Adamantane/analogs & derivatives , Seizures , Animals , Mice , Seizures/genetics , Neurons , Hippocampus , Receptors, AMPA/genetics
11.
J Mol Model ; 30(2): 39, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38224406

ABSTRACT

CONTEXT: Mycobacterial membrane proteins Large 3 (MmpL3) is responsible for the transport of mycobacterial acids out of cell membrane to form cell wall, which is essential for the survival of Mycobacterium tuberculosis (Mtb) and has become a potent anti-tuberculosis target. SQ109 is an ethambutol (EMB) analogue, as a novel anti-tuberculosis drug, can effectively inhibit MmpL3, and has completed phase 2b-3 clinical trials. Drug resistance has always been the bottleneck problem in clinical treatment of tuberculosis. The S288T mutant of MmpL3 shows significant resistance to the inhibitor SQ109, while the specific action mechanism remains unclear. The results show that MmpL3 S288T mutation causes local conformational change with little effect on the global structure. With MmpL3 bound by SQ109 inhibitor, the distance between D710 and R715 increases resulting in H-bond destruction, but their interactions and proton transfer function are still restored. In addition, the rotation of Y44 in the S288T mutant leads to an obvious bend in the periplasmic domain channel and an increased number of contact residues, reducing substrate transport efficiency. This work not only provides a possible dual drug resistance mechanism of MmpL3 S288T mutant but also aids the development of novel anti-tuberculosis inhibitors. METHODS: In this work, molecular dynamics (MD) and quantum mechanics (QM) simulations both were performed to compare inhibitor (i.e., SQ109) recognition, motion characteristics, and H-bond energy change of MmpL3 after S288T mutation. In addition, the WT_SQ109 complex structure was obtained by molecular docking program (Autodock 4.2); Molecular Mechanics/ Poisson Boltzmann Surface Area (MM-PBSA) and Solvated Interaction Energy (SIE) methods were used to calculate the binding free energies (∆Gbind); Geometric criteria were used to analyze the changes of hydrogen bond networks.


Subject(s)
Adamantane/analogs & derivatives , Ethylenediamines , Mycobacterium tuberculosis , Protons , Molecular Docking Simulation , Ion Channels , Cell Membrane , Mycobacterium tuberculosis/genetics
12.
Mod Rheumatol ; 34(3): 453-465, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-37319017

ABSTRACT

OBJECTIVE: We evaluated remission rates and their relationship with baseline characteristics in patients with rheumatoid arthritis treated with the oral Janus kinase inhibitor peficitinib. METHODS: This post hoc analysis of data from two Phase 3 studies (RAJ3 and RAJ4) of peficitinib (100 and 150 mg/day) in Asian rheumatoid arthritis patients investigated clinical disease activity index (CDAI) remission and low disease activity rates from baseline to Week 52. CDAI, Health Assessment Questionnaire-Disability Index, and van der Heijde-modified total Sharp score remission/low disease activity rates at Week 52 were evaluated among patients achieving CDAI remission at Weeks 12/28. Logistic regression analyses explored the relationship between baseline characteristics and CDAI remission/low disease activity rates. RESULTS: CDAI remission rates increased over time in a dose-dependent manner in both peficitinib-treated groups. Most patients achieving CDAI remission at Weeks 12/28 also achieved remission at Week 52. Following the multivariate analysis of demographic and baseline characteristics, factors associated with the achievement of CDAI remission at Week 28 included male sex, low baseline prednisone dose (RAJ3 only), and low baseline Disease Activity Score 28-C-reactive protein (RAJ4 only). CONCLUSIONS: Peficitinib demonstrated persistent efficacy in clinical remission to Week 52. Baseline characteristics associated with CDAI remission were mostly consistent with previous studies using other disease-modifying antirheumatic drugs.


Subject(s)
Adamantane/analogs & derivatives , Antirheumatic Agents , Arthritis, Rheumatoid , Niacinamide/analogs & derivatives , Humans , Male , Japan , Treatment Outcome , Severity of Illness Index , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/adverse effects , Remission Induction
13.
Acta Diabetol ; 61(1): 91-97, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37691025

ABSTRACT

AIMS: Type 2 diabetes mellitus (T2DM) increases the risk of major cardiovascular events. In SAVOR-TIMI53 trial, the excess heart failure (HF) hospitalization among patients with T2DM in the saxagliptin group remains poorly understood. Our aim was to evaluate left ventricular (LV) diastolic function after 6 months of saxagliptin treatment using cardiac magnetic resonance imaging (CMR) in patients with T2DM. METHODS: In this prospective study, 16 T2DM patients without HF were prescribed saxagliptin as part of routine guideline-directed management. CMR performed at baseline and 6 months after initiation of saxagliptin treatment were evaluated in a blinded fashion. We assessed LV diastolic function by measuring LV peak filling rate with correction for end-diastolic volume (PFR/LVEDV), time to peak filling rate with correction for cardiac cycle (TPF/RR), and early diastolic strain rate parameters [global longitudinal diastolic strain rate (GLSR-E), global circumferential diastolic strain rate (GCSR-E)] by feature tracking (FT-CMR). RESULTS: Among the 16 patients (mean age of 59.9, 69% males, mean hemoglobin A1c 8.3%, mean left ventricular ejection fraction 57%), mean PFR was 314 ± 108 ml/s at baseline and did not change over 6 months (- 2.7, 95% CI - 35.6, 30.2, p = 0.86). There were also no significant changes in other diastolic parameters including PFR/EDV, TPF, TPF/RR, and GLSR-E and GCSR-E (all p > 0.50). CONCLUSION: In T2DM patients without HF receiving saxagliptin over 6 months, there were no significant subclinical changes in LV diastolic function as assessed by CMR.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2 , Dipeptides , Heart Failure , Ventricular Dysfunction, Left , Male , Humans , Middle Aged , Female , Ventricular Function, Left , Stroke Volume , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Prospective Studies , Magnetic Resonance Imaging , Heart Failure/diagnostic imaging , Heart Failure/drug therapy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology
14.
Sci Rep ; 13(1): 8435, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37225786

ABSTRACT

Glioblastomas are highly aggressive and deadly brain tumours, with a median survival time of 14-18 months post-diagnosis. Current treatment modalities are limited and only modestly increase survival time. Effective therapeutic alternatives are urgently needed. The purinergic P2X7 receptor (P2X7R) is activated within the glioblastoma microenvironment and evidence suggests it contributes to tumour growth. Studies have implicated P2X7R involvement in a range of neoplasms, including glioblastomas, although the roles of P2X7R in the tumour milieu remain unclear. Here, we report a trophic, tumour-promoting role of P2X7R activation in both patient-derived primary glioblastoma cultures and the U251 human glioblastoma cell line, and demonstrate its inhibition reduces tumour growth in vitro. Primary glioblastoma and U251 cell cultures were treated with the specific P2X7R antagonist, AZ10606120 (AZ), for 72 h. The effects of AZ treatment were also compared to cells treated with the current first-line chemotherapeutic drug, temozolomide (TMZ), and a combination of both AZ and TMZ. P2X7R antagonism by AZ significantly depleted glioblastoma cell numbers compared to untreated cells, in both primary glioblastoma and U251 cultures. Notably, AZ treatment was more effective at tumour cell killing than TMZ. No synergistic effect between AZ and TMZ was observed. AZ treatment also significantly increased lactate dehydrogenase release in primary glioblastoma cultures, suggesting AZ-induced cellular cytotoxicity. Our results reveal a trophic role of P2X7R in glioblastoma. Importantly, these data highlight the potential for P2X7R inhibition as a novel and effective alternative therapeutic approach for patients with lethal glioblastomas.


Subject(s)
Adamantane , Glioblastoma , Purinergic P2X Receptor Antagonists , Humans , Adamantane/analogs & derivatives , Adamantane/pharmacology , Aminoquinolines/pharmacology , Glioblastoma/drug therapy , Receptors, Purinergic P2X7 , Temozolomide/pharmacology , Tumor Microenvironment , Purinergic P2X Receptor Antagonists/pharmacology
15.
Niger J Physiol Sci ; 38(1): 37-46, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-38243349

ABSTRACT

Glucocorticoids have therapeutic benefits in the management of several inflammatory and immunological disorders. Despite these medicinal effects, they have the drawback of causing metabolic disorders such as hyperglycemia, insulin resistance etc., which is known to be a key indicator of metabolic syndrome. Metabolic syndrome is a major predisposing factor to type 2 diabetes mellitus and cardiomyopathy. This study was designed to compare and evaluate the effects of saxagliptin, metformin and intranasal insulin (when used singly or in combination) on dexamethasone induced insulin resistance. Fifty-six female rats were randomly assigned into eight groups. Group 1 represented the control; Group 2 was administered with dexamethasone (1mg/kg) (untreated); Group 3 received dexamethasone + intranasal insulin (2IU); Group 4 received dexamethasone + intranasal insulin + metformin (40mg/kg); Group 5; received dexamethasone + intranasal + saxagliptin (8mg/kg); Group 6 received dexamethasone + metformin (40mg/kg); Group 7 received dexamethasone + saxagliptin (8mg/kg); Group 8 received dexamethasone + saxagliptin(8mg/kg) + metformin(40mg/kg). Treatments were given for one week. At the end of the study, blood samples were collected for biochemical assays and pancreas excised for histological examination. Dexamethasone (1mg/kg) induced hyperglycemia, hyperinsulinemia, dyslipidemia, impaired glucose tolerance and disrupted the structural integrity of the pancreas. Treatment with saxagliptin, metformin and their combination significantly decreased blood glucose level, decreased LDL Level and improved glucose tolerance. The selected hypoglycemic agents used in present study ameliorate the dexamethasone induced hyperglycemia and insulin resistance of which the combination of metformin with saxagliptin showed greater efficacy.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2 , Dipeptides , Hyperglycemia , Insulin Resistance , Metabolic Syndrome , Metformin , Female , Rats , Animals , Metformin/pharmacology , Insulin , Rats, Wistar , Blood Glucose/metabolism , Hypoglycemic Agents/pharmacology , Hyperglycemia/drug therapy , Dexamethasone , Drug Therapy, Combination
16.
NEJM Evid ; 2(12): EVIDoa2300210, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38320500

ABSTRACT

Dapagliflozin or Saxagliptin in Pediatric Type 2 DiabetesDapagliflozin (a sodium-glucose co-transporter-2 inhibitor) and saxagliptin (a dipeptidyl peptidase-4 inhibitor) have both been approved by the U.S. Food and Drug Administration for the treatment of type 2 diabetes in adults but not in children. In this randomized trial of 245 pediatric patients (10 to 17 years of age) with uncontrolled type 2 diabetes, dapagliflozin but not saxagliptin significantly reduced A1C compared with placebo.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2 , Dipeptides , Glucosides , Sodium-Glucose Transporter 2 Inhibitors , United States , Adult , Humans , Child , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents , Benzhydryl Compounds , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
17.
J Pharmacol Sci ; 150(2): 74-80, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36055754

ABSTRACT

PURPOSE: Peficitinib and tofacitinib are known to suppress inflammation in rheumatoid arthritis (RA) by inhibiting Janus kinases (JAKs). However, these effects on tyrosine kinases other than JAKs have not yet been well investigated. We evaluated the effects of peficitinib and tofacitinib on platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) receptor tyrosine kinases (RTKs) and on the activation of fibroblast-like synoviocytes (FLSs) and endothelial cells, main pathological causes of RA. METHODS: Peficitinib and tofacitinib were tested in PDGF and VEGF RTK assays. We then used FLSs derived from RA patient (RA-FLSs) and human umbilical vein endothelial cells (HUVECs) to study the effects of peficitinib and tofacitinib on PDGF- and VEGF-induced signal transduction and on the activation of RA-FLSs and endothelial cell tube formation. FINDINGS: Peficitinib, not tofacitinib, inhibited both PDGF and VEGF RTKs in addition to JAKs in cell-free assay system. Peficitinib and tofacitinib attenuated PDGF- and VEGF-induced intracellular signal transduction pathways in RA-FLSs and HUVECs to varying degrees. Only peficitinib potently inhibited PDGF-induced secretion of interleukin-6, VEGF, and matrix metalloproteinase-3 in RA-FLSs, and endothelial cell tube formation by HUVECs. CONCLUSION: Peficitinib may improve RA through inhibition of PDGF and VEGF signal transduction, in addition to JAK inhibition.


Subject(s)
Arthritis, Rheumatoid , Synoviocytes , Adamantane/analogs & derivatives , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Cells, Cultured , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Janus Kinases , Niacinamide/analogs & derivatives , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Signal Transduction , Synoviocytes/pathology , Tyrosine/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors/metabolism , Vascular Endothelial Growth Factors/pharmacology
18.
Int J Biol Macromol ; 220: 1390-1401, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36116590

ABSTRACT

Dipeptidyl peptidase III (DPP III) is a cytosolic, two-domain zinc-exopeptidase. It is widely distributed in mammalian tissues, where it's involved in the final steps of normal intracellular protein degradation. However, its pronounced affinity for some bioactive peptides (angiotensins, enkephalins, and endomorphins) suggests more specific functions such as blood pressure regulation and involvement in pain regulation. We have investigated several different neuropeptides as potential substrates and inhibitors of human DPP III. The binding affinities and kinetic data determined by isothermal titration calorimetry, in combination with measurements of enzyme inhibition identified the hemorphin-related valorphin, tynorphin, S-tynorphin, and I-tynorphin as the most potent inhibitors of DPP III (actually slow substrates), whereas hemorphin-4 proved to be the best substrate of all neuropeptides examined. In addition, we have shown that the neuropeptides valorphin, Leu-valorphin-Arg, and the opioid peptide ß-casomorphin, are DPP III substrates. The molecular modelling of selected peptides shows uniform binding to the lower domain ß-strand residues of DPP III via peptide backbone atoms, but also previously unrecognized stabilizing interactions with conserved residues of the metal-binding site and catalytic machinery in the upper domain. The computational data helped explain the differences between substrates that are hydrolyzed effectively and those hydrolysed slowly by DPP III.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases , Zinc , Adamantane/analogs & derivatives , Angiotensins , Enkephalins , Humans , Opioid Peptides , Zinc/metabolism
19.
Drug Des Devel Ther ; 16: 2199-2211, 2022.
Article in English | MEDLINE | ID: mdl-35855741

ABSTRACT

The Covid-19 pandemic driven by the SARS-CoV-2 virus continues to exert extensive humanitarian and economic stress across the world. Although antivirals active against mild disease have been identified recently, new drugs to treat moderate and severe Covid-19 patients are needed. Sphingolipids regulate key pathologic processes, including viral proliferation and pathologic host inflammation. Opaganib (aka ABC294640) is a first-in-class clinical drug targeting sphingolipid metabolism for the treatment of cancer and inflammatory diseases. Recent work demonstrates that opaganib also has antiviral activity against several viruses including SARS-CoV-2. A recently completed multinational Phase 2/3 clinical trial of opaganib in patients hospitalized with Covid-19 demonstrated that opaganib can be safely administered to these patients, and more importantly, resulted in a 62% decrease in mortality in a large subpopulation of patients with moderately severe Covid-19. Furthermore, acceleration of the clearance of the virus was observed in opaganib-treated patients. Understanding the biochemical mechanism for the anti-SARS-CoV-2 activity of opaganib is essential for optimizing Covid-19 treatment protocols. Opaganib inhibits three key enzymes in sphingolipid metabolism: sphingosine kinase-2 (SK2); dihydroceramide desaturase (DES1); and glucosylceramide synthase (GCS). Herein, we describe a tripartite model by which opaganib suppresses infection and replication of SARS-CoV-2 by inhibiting SK2, DES1 and GCS. The potential impact of modulation of sphingolipid signaling on multi-organ dysfunction in Covid-19 patients is also discussed.


Subject(s)
COVID-19 Drug Treatment , Adamantane/analogs & derivatives , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , Pandemics , Pyridines , SARS-CoV-2 , Sphingolipids
20.
Nat Commun ; 13(1): 4171, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35853847

ABSTRACT

Alzheimer's disease (AD) is characterized by the brain accumulation of amyloid-ß and tau proteins. A growing body of literature suggests that epigenetic dysregulations play a role in the interplay of hallmark proteinopathies with neurodegeneration and cognitive impairment. Here, we aim to characterize an epigenetic dysregulation associated with the brain deposition of amyloid-ß and tau proteins. Using positron emission tomography (PET) tracers selective for amyloid-ß, tau, and class I histone deacetylase (HDAC I isoforms 1-3), we find that HDAC I levels are reduced in patients with AD. HDAC I PET reduction is associated with elevated amyloid-ß PET and tau PET concentrations. Notably, HDAC I reduction mediates the deleterious effects of amyloid-ß and tau on brain atrophy and cognitive impairment. HDAC I PET reduction is associated with 2-year longitudinal neurodegeneration and cognitive decline. We also find HDAC I reduction in the postmortem brain tissue of patients with AD and in a transgenic rat model expressing human amyloid-ß plus tau pathology in the same brain regions identified in vivo using PET. These observations highlight HDAC I reduction as an element associated with AD pathophysiology.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Histone Deacetylase 1 , Adamantane/analogs & derivatives , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids , Positron-Emission Tomography/methods , Rats , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...