Please login first

List of accepted submissions

 
 
Show results per page
Find papers
 
  • Open access
  • 0 Reads
Hypoglycemic activity of new S-derivatives of 6-(5-mercapto-4-methyl-4H-1,2,4-triazol-3-yl)pyrimidine-2,4(1H,3H)-dione.

A fundamental aspect of the pathophysiology of type 2 diabetes is the progressive decline in the body's response to prandial insulin, particularly in the early stages of the disease. The early-phase insulin response is significantly impaired, playing a key role in accelerating endogenous glucose production. In type 2 diabetes, endogenous glucose production persists despite prandial glucose intake. Combined with a persistent relative deficiency in insulin secretion, this leads to a predisposition to postprandial hyperglycemia.

1,2,4-Triazole is a structural motif frequently explored in the development of new compounds with potential therapeutic applications, particularly for diabetes mellitus and other metabolic disorders. These compounds can modulate enzymes involved in glucose metabolism, including AMP-activated protein kinase (AMPK).

The potential hypoglycemic activity of the new compound was evaluated by measuring changes in blood glucose levels in animals after a single administration. Male Wistar rats (180–200 g) were used as test subjects and were housed under standard vivarium conditions. The animals were divided into two groups of six (n = 6). The control group received a third-generation sulfonylurea derivative, glimepiride (4 mg/kg), a structurally related hypoglycemic drug. The test group received the experimental compound orally via gavage, either as an aqueous solution or as a finely dispersed aqueous emulsion stabilized with Tween-80. Blood samples were collected from the tail vein before administration and at 2, 4, 6, and 8 hours post-administration.

Conclusions. The study results demonstrated that 2-((5-(2,6-dioxo-1,2,3,6-tetrahydropyrimidin-4-yl)-4-ethyl-4H-1,2,4-triazol-3-yl)thio)acetamide exhibits 1.5 times greater hypoglycemic activity than glimepiride. Molecular docking analysis confirmed the compound's high affinity for the α-glucosidase enzyme (PDB ID: 2ZE0).

  • Open access
  • 0 Reads
Stabilized Curcumin in Emulsions and Hydrogel Systems: A Nutritional Strategy for Cardiovascular and Renal Protection in Hypercholesterolemia
, , , , ,

Hypercholesterolemia is a significant risk factor for cardiovascular diseases (CVDs) and has been increasingly associated with renal dysfunction. Nutritional interventions featuring bioactive compounds such as curcumin have gained attention due to their lipid-lowering, anti-inflammatory, and nephroprotective effects. A comprehensive literature search was conducted across PubMed, Scopus, and Web of Science databases, selecting randomized controlled trials (RCTs), cohort studies, and systematic reviews published between 2010 and 2024. Meta-analysis of the selected studies demonstrated that curcumin supplementation significantly reduced total cholesterol (mean difference: -15.8 mg/dL, 95% CI: -20.4 to -11.2), triglycerides (-13.2 mg/dL, 95% CI: -18.5 to -7.9), and LDL levels (-10.5 mg/dL, 95% CI: -15.7 to -5.3), while increasing HDL levels (+6.7 mg/dL, 95% CI: 3.2 to 10.2). A systematic review and meta-analysis by Emami et al. (2022) highlighted curcumin's anti-inflammatory response in CKD patients. However, curcumin’s poor aqueous solubility and rapid metabolism limit its therapeutic potential. To overcome these challenges, researchers have developed advanced delivery systems such as microencapsulation, emulsions, and hydrogel matrices, to enhance curcumin's bioavailability, stability, and controlled release. Delivery systems such as microencapsulation, nanoemulsions, and hydrogel matrices enhanced curcumin’s bioavailability and therapeutic efficacy. Studies, including on carried out by Ashraf et al. (2022), have highlighted the role of microencapsulated curcumin in modulating diet-induced hypercholesterolemia, supporting its potential as a therapeutic intervention.

Acknowledgement: Part of the work was supported by a grant of the Romanian Ministry of Education and Research, CCCDI-UEFISCDI, project number PN-IV-P1-PCE-2023-1092.

  • Open access
  • 0 Reads
Variation in White Blood Cell Counts among Individuals with Type 2 Diabetes Mellitus
,

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by insulin resistance and hyperglycemia. Emerging evidence suggests that variations in white blood cell (WBC) counts among individuals with T2DM may provide insights into the inflammatory and immune responses associated with the disease. This study aims to investigate WBC count variations in T2DM and their relationship with glycemic control and other clinical parameters.

A cross-sectional study was conducted at Colombo Medi Lab, Bambalapitiya, Sri Lanka, involving 321 participants categorized into pre-diabetic (FBS:100–125 mg/dl, Postprandial Blood Glucose: 140-199mg/dl; HbA1c: 5.7-6.4%) and diabetes (FBS>126 mg/dl; Postprandial Blood Glucose: >200mg/dl; HbA1c: ≥6.5%) groups. WBC counts and demographic data were collected, and statistical analyses were performed using Microsoft Excel and SPSS 20.0, with a significance threshold of p < 0.05.

Among the 321 participants, 171 were T2DM patients (30 pre-DM, 113 females, 58 males), while 150 were healthy individuals (95 females, 55 males). Significant differences (p < 0.05) were observed between the DM, pre-DM, and control groups in WBC count, neutrophil count, lymphocyte count, N/L ratio, MID count, and FBS.

WBC, lymphocyte, and MID counts showed a significant (p < 0.001) positive correlation with age, whereas neutrophil count, N/L ratio, and FBS showed a negative correlation. The N/L ratio and MID count positively correlated with gender, while WBC, lymphocyte count, and FBS were negatively correlated. WBC count, neutrophil count, N/L ratio, and FBS correlated positively with BMI, whereas lymphocyte and MID counts were negatively correlated.

This study highlights WBC count variations as potential biomarkers for monitoring T2DM progression. Future research should focus on longitudinal and interventional studies to explore causal relationships and clinical applications in diabetes management.

  • Open access
  • 0 Reads
The specific combinations of cytokines and tumor-derived factors determine particular immunosuppressive properties of in vitro generated murine myeloid-derived suppressor cells

MDSCs are heterogeneous populations of immature myeloid cells with potent immunosuppressive capabilities that play a significant role in tumor immunology and autoimmune diseases. These cells have emerged as critical regulators of immune responses, impacting disease progression and treatment outcomes. MDSCs use multiple mechanisms to suppress immune cells, primarily T cells, and can be divided into two subsets, polymorphonuclear (PMN-MDSCs) and monocytic (M-MDSCs) ones, which resemble mature neutrophils and monocytes, respectively.

To facilitate MDSC investigation and develop effective MDSC-mediated therapies, it is essential to establish reliable methods for their in vitro generation. In our study, we compared six protocols for the in vitro generation of functional mouse MDSCs from bone marrow progenitors. The protocols included granulocyte-macrophage colony-stimulating factor (GM-CSF) alone or in combination with interleukin-6 (IL-6) or granulocyte colony-stimulating factor (G-CSF), with or without a tumor-conditioned medium (TCM) derived from B16-F10 melanoma. Obtained MDSCs were characterized by the morphology, phenotype, and gene expression of key immunosuppressive factors, as well as the in vitro suppression of T cell proliferation.

All tested protocols yielded approximately 25% M-MDSCs and 50% PMN-MDSCs. Protocols utilizing IL-6 generated MDSCs with reduced maturation and differentiation markers, upregulated expression of Arg1 and Nos1 mRNA, increased levels of Arg-1 and TGF-β proteins and enhanced ROS production compared to the other protocols. All protocols under the study generated MDSCs that efficiently inhibited T cell proliferation in vitro, with some advantage for the GM-CSF and G-CSF+GM-CSF protocols. Interestingly, protocols combined with B16-F10-TCM reduced the immunosuppressive properties of generated MDSCs.

These results provide valuable insights into the optimal conditions for the in vitro generation of MDSCs with specific immunosuppressive properties. This knowledge will contribute to the development of more effective MDSC-mediated therapeutic approaches for various disorders, including tumor-induced immunosuppression, transplant complications, and autoimmune and inflammatory diseases.

This research was funded by the Russian Science Foundation (Grant/Award Number: 19-74-30011); and the Russian state-funded project for ICBFM SB RAS (Grant/Award Number: 125012300659-6).

  • Open access
  • 0 Reads
Pulmonary mycobacterium chimaera intracellulare: a rare case report

Abstract:

We report a rare case of pulmonary Mycobacterium chimaera intracellulare infection in a 69-year-old female with a history of pulmonary tuberculosis. The patient presented with chronic cough, weight loss, and shortness of breath and was successfully treated with a combination of antibiotics.

Introduction:

Mycobacterium chimaera intracellulare is a rare and emerging pathogen that can cause pulmonary disease, particularly in immunocompromised individuals. We present a case of pulmonary Mycobacterium chimaera intracellulare infection in a 69-year-old female with a history of pulmonary tuberculosis.

Case Presentation:

A 69-year-old female with a history of pulmonary tuberculosis presented to our hospital with a 3-month history of chronic cough, weight loss, and shortness of breath. Chest imaging revealed bilateral lung nodules and cavitations. Sputum samples were positive for Mycobacterium chimaera intracellulare, confirmed by molecular testing.

Treatment and Outcome:

The patient was treated with a combination of antibiotics, including clarithromycin, rifampicin, and ethambutol. She showed significant clinical improvement, with a resolution of symptoms and radiographic findings. The patient was treated for 12 months, with regular follow-up appointments to monitor her progress.

Discussion:

This case highlights the importance of considering Mycobacterium chimaera intracellulare in the differential diagnosis of pulmonary infections, particularly in patients with a history of pulmonary tuberculosis. Early diagnosis and treatment are crucial to prevent disease progression and improve outcomes.

Conclusion:

Pulmonary Mycobacterium chimaera intracellulare infection is a rare but important diagnosis to consider in patients with pulmonary symptoms and a history of tuberculosis. Prompt treatment with antibiotics can lead to significant clinical improvement and a resolution of symptoms.

  • Open access
  • 0 Reads
Reshaping the tumor immune microenvironment through ROS-responsive nanoparticles featuring the CRISPRi system to mediate Programmed Death-Ligand 1 attenuation and enhance cancer immunotherapy via an epigenetic inhibitor

The involvement of Programmed death-ligand 1 (PD-L1) in tumor cells, with its receptor PD-1 on immune cells, can transmit an inhibitory signal to induce immune evasion. Although the immune checkpoint inhibitor PD-L1 antibody has achieved significant antitumor efficiency in clinical treatment, its wide clinical application still involves several side effects and individual selectivity. In our research, we intend to use the CRISPR interference (CRISPRi) system to suppress PD-L1 expression in tumor cells; we combine it with the epigenetic inhibitor azacytidine (AZA) for enhanced cancer immunotherapy. We first fabricate a reactive oxygen species (ROS)-responsive poly(β-amino ester) (PBAE)-S-AZA cationic polymer prodrug. The CRISPRi plasmid and PBAE-S-AZA prodrug are complexed to form the final nanoparticles via electrostatic interaction. The nanoparticles are uptaken in tumor cells with high efficiency, followed by the escape of the plasmid via the endosome/lysosome with the cooperation of PBAE. The release of the CRISPRi plasmid can lead to a suppression of PD-L1 in tumor cells and a relief of the immune checkpoint blockade. In the meantime, the epigenetic inhibitor AZA is also released from the nanoparticles due to the high intracellular ROS level to synergistically enhance the efficacy of immunotherapy. It is demonstrated that the nanoparticles can significantly increase the proportions of CD8+ T cells, CD4+ T cells and M1 macrophages in the tumor microenvironment while decreasing the proportion of regulatory T cells (Tregs) and M2 macrophages. Thus, the nanoparticles help to achieve the combination of genome editing, immunotherapy and epigenetic regulation effectively. This study provides a novel platform for promoting antitumor treatment and precision medicine.

  • Open access
  • 0 Reads
Development and Assessment of a Candesartan-loaded Bilosomal In Situ gel for targeted Nose-to-Brain Delivery

ABSTRACT

INTRODUCTION:

Candesartan is an angiotensin II receptor blocker that has been used in the treatment of arterial hypertension. Its repurposing in migraine treatment, due to potential mechanisms involved in migraine pathophysiology, is currently being explored. Repurposing medications lowers their development costs and time to market, and the greatest benefit of repurposing is that it offers tailored therapy at a lower cost. Candesartan is a BCS class II drug with low solubility and high permeability. The oral bioavailability of Candesartan is very low, approximately 15%. Therefore, it is important to find an alternate route to deliver Candesartan to increase its bioavailability. In this study, we assess the potential of delivering Candesartan intranasally, directly to the brain, through the preparation of bilosomes incorporated into a muco-adhesive in situ gel to increase its retention time and for brain targeting to treat migraines.

METHODS:

Candesartan bilosomes will be prepared by using a thin-film hydration technique. This formulation will be characterized in terms of vesicle size, entrapment efficiency, zeta potential, morphology, and Fourier transform infrared spectroscopy. Candesartan bilosomes will be incorporated into an in situ gel and will be evaluated in terms of pH, viscosity, muco-adhesive strength, spreadability, gel strength, in vitro release, ex vivo permeation, and in vivo study.

CONCLUSION:

A bilosomal system that effectively encapsulates Candesartan will enhance its solubility, stability, and permeation across the nasal mucosa. In situ gels exhibit favorable muco-adhesive properties which contribute to prolonged contact time with the nasal mucosa, accelerating sustained drug release and enhancing brain-targeted delivery. A Candesartan-loaded bilosomal in situ gel may provide a non-invasive and efficient alternative for migraine management.

  • Open access
  • 0 Reads
Reshaping tumor immune microenvironment through ROS-responsive nanoparticles featured with CRISPRi system-mediated Programmed Death-Ligand 1 attenuation and epigenetic inhibitor-enhanced cancer immunotherapy

The engagement of programmed death-ligand 1 (PD-L1) on tumor cells with its receptor PD-1 on immune cells can transmit an inhibitory signal to induce immune evasion. Although the immune checkpoint inhibitor PD-L1 antibody has significant antitumor efficiency in clinical treatment, its widespread clinical application still faces several side effects and individual selectivity. In our research, we use the CRISPR interference (CRISPRi) system to suppress PD-L1 expression in tumor cells and combine it with epigenetic inhibitor azacytidine (AZA) for enhanced cancer immunotherapy. We first fabricate a reactive oxygen species (ROS)-responsive poly(β-amino ester) (PBAE)-S-AZA cationic polymer prodrug. CRISPRi plasmid and PBAE-S-AZA prodrug are complexed to form the final nanoparticles via electrostatic interaction. The nanoparticles exhibit high efficiency in uptake by tumor cells, followed by the endosome/lysosome escape of plasmid with the cooperation of PBAE. The release of CRISPRi plasmid can lead to the suppression of PD-L1 in tumor cells and relief of immune checkpoint blockade. Meanwhile, the epigenetic inhibitor AZA is also released from the nanoparticles due to the high intracellular ROS level to enhance the efficacy of immunotherapy synergically. It was demonstrated that the ROS-responsive nanoparticles could significantly increase the proportion of CD8+ T cells, CD4+ T cells, and M1 macrophages in a tumor microenvironment, while decreasing the proportions of regulatory T cells (Treg) and M2 macrophages. Thus, the nanoparticles help to realize the combination of genome editing, immunotherapy, and epigenetic regulation effectively. Our method provides a novel platform for promoting antitumor treatment and precision medicine.

  • Open access
  • 0 Reads
ALTERATIONS IN THE EXPRESSION OF A GROUP OF GENES ASSOCIATED WITH THE EPITHELIAL–MESENCHYMAL TRANSITION IN LUNG CANCER: A PILOT STUDY

A large number of diagnosed cases of lung cancer occur in late stages and are accompanied by metastasis, which makes research into the epithelial-to-mesenchymal transition (EMT) genes relevant. The genes responsible for the EMT include E-cadherin (CDH1), which is involved in the formation of tight junctions between epithelial cells, soluble growth factors, and some transcription factors (e.g., ZEB1, ZEB2).

The aim of this study was to evaluate the expression alterations in BMI1, CDH1, SNAI1, SNAI2, ZEB1, and ZEB2 in tumors compared to histologically normal lung tissues.

The study used 50 paired RNA samples with RIN≥7 isolated from tumors, including 37 non-metastatic and 13 metastatic tumors, and normal lung tissues. The expression levels were assessed by RT-qPCR. B2M and ACTB were used as reference genes. Statistical processing of the results was performed by ANOVA using the CFX Maestro software (Bio-Rad, USA).

The expression of six genes associated with the EMT—BMI1, CDH1, SNAI1, SNAI2, ZEB1, and ZEB2—was analyzed. CDH1 expression increased in 3.63 times in tumors compared to the norm, p ≤ 0.05; moreover, in non-metastatic tumors, it increased by 3.47 times, but in metastatic tumors, it increased by 4.21 times, p ≤ 0.05. ZEB1 and ZEB2 expression decreased by 2.32 and 2.01 times, p ≤ 0.05, and their expression in metastatic tumors decreased by 3.22 and 3.01 times, p ≤ 0.05. The expression levels of BMI1, SNAI1, and SNAI2 changed by a factor of -1.51 to 1.53; however, these data were not statistically significant.

Based on the results of this study, we can conclude that E-cadherin in lung cancer promotes either the epithelial-to-mesenchymal or mesenchymal-to-epithelial transition and, consequently, metastasis. We also showed a suppression of the expression of transcription factors encoded by the ZEB1 and ZEB2. The role of BMI1, SNAI1, and SNAI2 in lung cancer is likely limited, as their expression remains largely unchanged.

  • Open access
  • 0 Reads
Nobiletin Mitigates Vascular Calcification by Activating RORα to Protect DLC1 from EZH2-Mediated Degradation

Introduction: Vascular calcification (VC) is a common pathological feature in chronic kidney disease, atherosclerosis, and aging. Nobiletin (NOB) is a natural compound which has various biological activities. This study aimed to explore the effect of NOB on VC and its molecular mechanism. Methods: Primary vascular smooth muscle cells (VSMCs) were cultured in calcification medium for 3 days to induce calcification. For NOB-treated groups, 10µM NOB was added into medium or calcification medium. A mouse model of VC was established by intraperitoneal injection of vitamin D, and mice were orally gavaged with 50mg/kg/day NOB or vehicle. Alizarin-Red staining and calcium content quantitation were used to visualize calcium deposition. Western blot analysis, co-immunoprecipitation, quantitative proteomic analyses and methyltransferase assays were performed to detect protein expression, protein–protein interactions, and post-translational modification levels. Results: Network pharmacology analysis identified NOB as a potential therapeutic agent for VC. In vitro, Alizarin-Red staining, calcium content quantitation, qRT-PCR and Western blot showed that NOB prevented calcium accumulation in VSMCs and downregulated osteogenic markers (RUNX2, OPN, BMP2). In the vitamin D-induced mouse aortic calcification model, NOB inhibited calcium deposition without affecting serum calcium levels and decreased osteogenic marker expression in aortas. Quantitative proteomic analyses suggested that NOB-induced upregulation of deleted in liver cancer-1 (DLC1) might be a key factor in inhibiting VC. DLC1 overexpression reduced calcium deposition and osteogenic marker expression in VSMCs, while knockdown of DLC1 aggravated calcification. Mechanistically, NOB upregulated RAR-related orphan receptor alpha (RORα) expression, which competed with DLC1 for binding to enhancer of Zeste homolog 2 (EZH2). This competitive interaction inhibited EZH2-mediated methylation and degradation of DLC1, preserving its function in VSMCs. Conclusion: Our findings highlight a novel therapeutic mechanism of NOB in vascular calcification through modulation of the RORα–EZH2–DLC1 axis, providing mechanistic insights into natural-compound-mediated regulation of vascular pathology.

Top