Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.696
Filter
1.
Bull Exp Biol Med ; 177(2): 212-216, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39093471

ABSTRACT

The effect of a promising NO donor, a binuclear nitrosyl iron complex (NIC) with 3,4-dichlorothiophenolyls [Fe2(SC6H3Cl2)2(NO)4], on the adenylate cyclase and soluble guanylate cyclase enzymatic systems was studied. In in vitro experiments, this complex increased the concentration of important secondary messengers, such as cAMP and cGMP. An increase of their level by 2.4 and 4.5 times, respectively, was detected at NIC concentration of 0.1 mM. The ligand of the complex, 3,4-dichlorothiophenol, produced a less pronounced effect on adenylate cyclase. It was shown that the effect of this complex on the activity of soluble guanylate cyclase was comparable to the effect of anionic nitrosyl complex with thiosulfate ligands that exhibits vasodilating and cardioprotective properties.


Subject(s)
Cyclic AMP , Cyclic GMP , Cyclic GMP/metabolism , Cyclic AMP/metabolism , Animals , Iron/metabolism , Iron/chemistry , Adenylyl Cyclases/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Soluble Guanylyl Cyclase/metabolism , Nitrogen Oxides/pharmacology , Nitrogen Oxides/metabolism , Nitrogen Oxides/chemistry , Rats
2.
Bull Exp Biol Med ; 177(2): 266-270, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39093476

ABSTRACT

The efficiency of combinations of cytostatics cisplatin and adriamycin with antioxidant sodium 3-(3'-tert-butyl-4-hydroxyphenyl)propyl thiosulfate (TS-13), and nitric oxide (NO) donor NaNO2 was evaluated on two drug-resistant strains of leukemia P388 with changed redox-status of cells. Simultaneous use of both NO donor and TS-13 in combinations with the cytostatics did not increase the efficiency of therapy. In addition, antioxidant activity of TS-13, NaNO2, and their combinations was studied by the method of luminol-dependent chemiluminescence on the model systems with the use of the homogenized cells of sensitive strain and two drug-resistant strains of leukemia P388. It was shown that TS-13 and NO donor produced opposite effects: TS-13 decreased, while NO donor increased the content of free radicals in the model system. Combinations of antioxidant TS-13 and NO donor should be used with consideration for the redox-status of tumor treated.


Subject(s)
Antioxidants , Cisplatin , Doxorubicin , Drug Resistance, Neoplasm , Leukemia P388 , Nitric Oxide Donors , Oxidation-Reduction , Animals , Mice , Oxidation-Reduction/drug effects , Drug Resistance, Neoplasm/drug effects , Antioxidants/pharmacology , Doxorubicin/pharmacology , Leukemia P388/drug therapy , Leukemia P388/pathology , Cisplatin/pharmacology , Cisplatin/therapeutic use , Nitric Oxide Donors/pharmacology , Thiosulfates/pharmacology , Sodium Nitrite/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology
3.
Physiol Rep ; 12(15): e16147, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39097984

ABSTRACT

The cardioprotective effect of ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) in adult hearts is mediated by nitric oxide (NO). During the early developmental period, rat hearts exhibit higher resistance to ischemia-reperfusion (I/R) injury, contain higher levels of serum nitrates, and their resistance cannot be further increased by IPC or IPoC. NOS blocker (L-NAME) lowers their high resistance. Wistar rat hearts (postnatal Days 1 and 10) were perfused according to Langendorff and exposed to 40 min of global ischemia followed by reperfusion with or without IPoC. NO and reactive oxygen species donors (DEA-NONO, SIN-1) and L-NAME were administered. Tolerance to ischemia decreased between Days 1 and 10. DEA-NONO (low concentrations) significantly increased tolerance to I/R injury on both Days 1 and 10. SIN-1 increased tolerance to I/R injury on Day 10, but not on Day 1. L-NAME significantly reduced resistance to I/R injury on Day 1, but actually increased resistance to I/R injury on Day 10. Cardioprotection by IPoC on Day 10 was not affected by either NO donors or L-NAME. It can be concluded that resistance of the neonatal heart to I/R injury is NO dependent, but unlike in adult hearts, cardioprotective interventions, such as IPoC, are most likely NO independent.


Subject(s)
Animals, Newborn , Ischemic Postconditioning , Myocardial Reperfusion Injury , NG-Nitroarginine Methyl Ester , Nitric Oxide , Rats, Wistar , Animals , Nitric Oxide/metabolism , Ischemic Postconditioning/methods , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Rats , NG-Nitroarginine Methyl Ester/pharmacology , Ischemic Preconditioning, Myocardial/methods , Nitric Oxide Donors/pharmacology , Male , Heart/drug effects , Myocardium/metabolism , Molsidomine/pharmacology , Molsidomine/analogs & derivatives
4.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126104

ABSTRACT

Melatonin regulates vital physiological processes in animals, such as the circadian cycle, sleep, locomotion, body temperature, food intake, and sexual and immune responses. In plants, melatonin modulates seed germination, longevity, circadian cycle, photoperiodicity, flowering, leaf senescence, postharvest fruit storage, and resistance against biotic and abiotic stresses. In plants, the effect of melatonin is mediated by various regulatory elements of the redox network, including RNS and ROS. Similarly, the radical gas NO mediates various physiological processes, like seed germination, flowering, leaf senescence, and stress responses. The biosynthesis of both melatonin and NO takes place in mitochondria and chloroplasts. Hence, both melatonin and nitric oxide are key signaling molecules governing their biological pathways independently. However, there are instances when these pathways cross each other and the two molecules interact with each other, resulting in the formation of N-nitrosomelatonin or NOMela, which is a nitrosated form of melatonin, discovered recently and with promising roles in plant development. The interaction between NO and melatonin is highly complex, and, although a handful of studies reporting these interactions have been published, the exact molecular mechanisms governing them and the prospects of NOMela as a NO donor have just started to be unraveled. Here, we review NO and melatonin production as well as RNS-melatonin interaction under normal and stressful conditions. Furthermore, for the first time, we provide highly sensitive, ozone-chemiluminescence-based comparative measurements of the nitric oxide content, as well as NO-release kinetics between NOMela and the commonly used NO donors CySNO and GSNO.


Subject(s)
Melatonin , Nitric Oxide , Plants , Melatonin/metabolism , Nitric Oxide/metabolism , Plants/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/pharmacology , Signal Transduction , Plant Physiological Phenomena
5.
Int J Mol Sci ; 25(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39063223

ABSTRACT

Numerous research projects focused on the management of acute pulmonary hypertension as Coronavirus Disease 2019 (COVID-19) might lead to hypoxia-induced pulmonary vasoconstriction related to acute respiratory distress syndrome. For that reason, inhalative therapeutic options have been the subject of several clinical trials. In this experimental study, we aimed to examine the hemodynamic impact of the inhalation of the SIN-1A formulation (N-nitroso-N-morpholino-amino-acetonitrile, the unstable active metabolite of molsidomine, stabilized by a cyclodextrin derivative) in a porcine model of acute pulmonary hypertension. Landrace pigs were divided into the following experimental groups: iNO (inhaled nitric oxide, n = 3), SIN-1A-5 (5 mg, n = 3), and SIN-1A-10 (10 mg, n = 3). Parallel insertion of a PiCCO system and a pulmonary artery catheter (Swan-Ganz) was performed for continuous hemodynamic monitoring. The impact of iNO (15 min) and SIN-1A inhalation (30 min) was investigated under physiologic conditions and U46619-induced acute pulmonary hypertension. Mean pulmonary arterial pressure (PAP) was reduced transiently by both substances. SIN-1A-10 had a comparable impact compared to iNO after U46619-induced pulmonary hypertension. PAP and PVR decreased significantly (changes in PAP: -30.1% iNO, -22.1% SIN-1A-5, -31.2% SIN-1A-10). While iNO therapy did not alter the mean arterial pressure (MAP) and systemic vascular resistance (SVR), SIN-1A administration resulted in decreased MAP and SVR values. Consequently, the PVR/SVR ratio was markedly reduced in the iNO group, while SIN-1A did not alter this parameter. The pulmonary vasodilatory impact of inhaled SIN-1A was shown to be dose-dependent. A larger dose of SIN-1A (10 mg) resulted in decreased PAP and PVR in a similar manner to the gold standard iNO therapy. Inhalation of the nebulized solution of the new SIN-1A formulation (stabilized by a cyclodextrin derivative) might be a valuable, effective option where iNO therapy is not available due to dosing difficulties or availability.


Subject(s)
Hypertension, Pulmonary , Molsidomine , Nitric Oxide , Animals , Administration, Inhalation , Molsidomine/pharmacology , Molsidomine/analogs & derivatives , Swine , Nitric Oxide/metabolism , Hypertension, Pulmonary/drug therapy , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacology , Vasodilation/drug effects , Pulmonary Artery/drug effects , Disease Models, Animal , Hemodynamics/drug effects , Lung/metabolism , Lung/drug effects , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Male
6.
Molecules ; 29(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39064961

ABSTRACT

Herein, we report the synthesis of a new hybrid compound based on a 2'-deoxyuridine nucleoside conjugated with a NO photo-donor moiety (dU-t-NO) via CuAAC click chemistry. Hybrid dU-t-NO, as well as two previously reported 2'-deoxyadenosine based hybrids (dAdo-S-NO and dAdo-t-NO), were evaluated for their cytotoxic and cytostatic activities in selected cancer cell lines. dAdo-S-NO and dAdo-t-NO hybrids displayed higher activity with respect to dU-t-NO. All hybrids showed effective release of NO in the micromolar range. The photochemical behavior of the newly reported hybrid, dU-t-NO, was studied in the RKO colon carcinoma cell line, whereas the dAdo-t-NO hybrid was tested in both colon carcinoma RKO and hepatocarcinoma Hep 3B2.1-7 cell lines to evaluate the potential effect of NO released upon irradiation on cell viability. A customized irradiation apparatus for in vitro experiments was also designed.


Subject(s)
Antineoplastic Agents , Nitric Oxide Donors , Nitric Oxide , Nucleosides , Humans , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Nitric Oxide/metabolism , Nitric Oxide/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Nucleosides/chemistry , Nucleosides/pharmacology , Cell Survival/drug effects , Click Chemistry , Cell Proliferation/drug effects , Molecular Structure , Deoxyuridine/chemistry , Deoxyuridine/pharmacology , Deoxyuridine/analogs & derivatives
7.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000090

ABSTRACT

The acidic byproducts of bacteria in plaque around orthodontic brackets contribute to white spot lesion (WSL) formation. Nitric oxide (NO) has antibacterial properties, hindering biofilm formation and inhibiting the growth of oral microbes. Materials that mimic NO release could prevent oral bacteria-related pathologies. This study aims to integrate S-nitroso-acetylpenicillamine (SNAP), a promising NO donor, into orthodontic elastomeric ligatures, apply an additional polymer coating, and evaluate the NO-release kinetics and antimicrobial activity against Streptococus mutans. SNAP was added to clear elastomeric chains (8 loops, 23 mm long) at three concentrations (50, 75, 100 mg/mL, and a control). Chains were then coated, via electrospinning, with additional polymer (Elastollan®) to aid in extending the NO release. NO flux was measured daily for 30 days. Samples with 75 mg/mL SNAP + Elastollan® were tested against S. mutans for inhibition of biofilm formation on and around the chain. SNAP was successfully integrated into ligatures at each concentration. Only the 75 mg/mL SNAP chains maintained their elasticity. After polymer coating, samples exhibited a significant burst of NO on the first day, exceeding the machine's reading capacity, which gradually decreased over 29 days. Ligatures also inhibited S. mutans growth and biofilm formation. Future research will assess their mechanical properties and cytotoxicity. This study presents a novel strategy to address white spot lesion (WSL) formation and bacterial-related pathologies by utilizing nitric oxide-releasing materials. Manufactured chains with antimicrobial properties provide a promising solution for orthodontic challenges, showing significant potential for academic-industrial collaboration and commercial viability.


Subject(s)
Biofilms , Elastomers , Nitric Oxide , Streptococcus mutans , Streptococcus mutans/drug effects , Streptococcus mutans/growth & development , Elastomers/chemistry , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Biofilms/drug effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Orthodontic Brackets/microbiology , Microbial Sensitivity Tests , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/chemical synthesis , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/chemical synthesis , Humans
8.
BMC Plant Biol ; 24(1): 678, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39014343

ABSTRACT

Cut flowers deteriorate rapidly after harvest, lasting mere days. To extend their vase life, various postharvest techniques are employed. Due to limited knowledge about the postharvest physiology of Alstroemeria cut flowers and the specific role of secondary compounds and antioxidant systems in their protection, this study investigated the optimal dosage of sodium nitroprusside (SNP) as a nitric oxide (NO) donor to enhance quality and antioxidant defenses. Preharvest foliar application of SNP at 0, 50, 100, and 200 µM followed by short-term pulsing treatments upon harvest at the same concentrations were applied in a factorial design. Results revealed that a preharvest 100 µM SNP treatment combined with a 50 µM postharvest pulse significantly increased the total amount of phenols (over 20%), antioxidant capacity (more than doubled), and the activity of two antioxidant enzymes (ascorbate peroxidase by over 35% and guaiacol peroxidase by about 20%). Notably, this combination also diminished ion leakage (by about 20%), ultimately extending the vase life by more than 40% compared to untreated plants. Therefore, SNP application at these specific dosages proves effective in bolstering Alstroemeria cut flower quality and vase life through enhanced total phenols and a strengthened antioxidant system.


Subject(s)
Antioxidants , Flowers , Nitroprusside , Nitroprusside/pharmacology , Flowers/drug effects , Flowers/physiology , Antioxidants/metabolism , Phenols/metabolism , Nitric Oxide Donors/pharmacology , Peroxidase/metabolism , Ascorbate Peroxidases/metabolism
9.
J Med Chem ; 67(15): 13089-13105, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39044437

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly lethal malignancy, and its clinical management encounters severe challenges due to its high metastatic propensity and the absence of effective therapeutic targets. To improve druggability of aurovertin B (AVB), a natural polyketide with a significant antiproliferative effect on TNBC, a series of NO donor/AVB hybrids were synthesized and tested for bioactivities. Among them, compound 4d significantly inhibited the proliferation and metastasis of TNBC in vitro and in vivo with better safety than that of AVB. The structure-activity relationship analysis suggested that the types of NO donor and the linkers had considerable effects on the activities. Mechanistic investigations unveiled that 4d induced apoptosis and ferroptosis by the reduction of mitochondrial membrane potential and the down-regulation of GPX4, respectively. The antimetastatic effect of 4d was associated with the upregulation of DUSP1. Overall, these compelling results underscore the tremendous potential of 4d for treating TNBC.


Subject(s)
Antineoplastic Agents , Apoptosis , Ferroptosis , Nitric Oxide Donors , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , Ferroptosis/drug effects , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/therapeutic use , Nitric Oxide Donors/chemical synthesis , Structure-Activity Relationship , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Pyrans/chemistry , Pyrans/pharmacology
10.
J Vasc Res ; 61(4): 179-196, 2024.
Article in English | MEDLINE | ID: mdl-38952123

ABSTRACT

INTRODUCTION: The comorbidities of ischemic heart disease (IHD) and diabetes mellitus (DM) compromise the protection of the diabetic heart from ischemia/reperfusion (I/R) injury. We hypothesized that manipulation of reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) pathways might protect the diabetic heart, and intervention of these pathways could be a new avenue for potentially protecting the diabetic heart. METHODS: All hearts were subjected to 30-min ischemia and 30-min reperfusion. During reperfusion, hearts were exposed to molecules proven to protect the heart from I/R injury. The hemodynamic data were collected using suitable software. The infarct size, troponin T levels, and protein levels in hearts were evaluated. RESULTS: Both cyclosporine-A and nitric oxide donor (SNAP) infusion at reperfusion protected 4-week diabetic hearts from I/R injury. However, 6-week diabetic hearts were protected only by SNAP, but not cyclosporin-A. These treatments significantly (p < 0.05) improved cardiac hemodynamics and decreased infarct size. CONCLUSIONS: The administration of SNAP to diabetic hearts protected both 4- and 6-week diabetic hearts; however, cyclosporine-A protected only the 4-week diabetic hearts. The eNOS/GLUT-4 pathway executed the SNAP-mediated cardioprotection.


Subject(s)
Cyclosporine , Diabetes Mellitus, Experimental , Myocardial Reperfusion Injury , Myocardium , Nitric Oxide Donors , Nitric Oxide , Signal Transduction , Animals , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide/metabolism , Diabetes Mellitus, Experimental/complications , Male , Cyclosporine/pharmacology , Nitric Oxide Donors/pharmacology , Myocardium/metabolism , Myocardium/pathology , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Blood Glucose/metabolism , Blood Glucose/drug effects , Time Factors , Rats, Sprague-Dawley , Troponin T/metabolism , Hyperglycemia/metabolism , Hyperglycemia/complications , Glucose Transporter Type 4
11.
Meat Sci ; 216: 109580, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38941777

ABSTRACT

This study aimed to quantitively profile the S-nitrosylation in beef semimembranosus (SM) with different treatments (nitric oxide donor or nitric oxide synthase inhibitor) by applying iodoTMT-based nitrosoproteomics. Results showed that 2096 S-nitrosylated cysteine sites in 368 proteins were detected in beef SM. Besides, differential SNO-modified proteins were screened, some of which were involved in crucial biochemical pathways, including calcium-releasing-related proteins, energy metabolic enzymes, myofibrils, and cytoskeletal proteins. GO analysis indicated that differential proteins were localized in a wide range of cellular compartments, such as cytoplasm, organelle, and mitochondrion, providing a prerequisite for S-nitrosylation exerting broad roles in post-mortem muscles. Furthermore, KEGG analysis validated that these proteins participated in the regulation of diverse post-mortem metabolic processes, especially glycolysis. To conclude, changes of S-nitrosylation levels in post-mortem muscles could impact the structure and function of crucial muscle proteins, which lead to different levels of muscle metabolism and ultimately affect beef quality.


Subject(s)
Muscle Proteins , Muscle, Skeletal , Proteomics , Red Meat , Cattle , Animals , Red Meat/analysis , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscle Proteins/metabolism , Cysteine/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology
12.
Biochim Biophys Acta Gen Subj ; 1868(9): 130652, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38857773

ABSTRACT

Mitochondria play a crucial role in maintaining Ca2+ homeostasis in cells. Due to the critical regulatory role of the products of oxidative and non-oxidative metabolism of L-arginine, it is essential to clarify their effect on Ca2+ transport in smooth muscle mitochondria. Experiments were performed on the uterine myocytes of rats and isolated mitochondria. The possibility of NO synthesis by mitochondria was demonstrated by confocal microscopy and spectrofluorimetry methods using the NO-sensitive fluorescent probe DAF-FM and Mitotracker Orange CM-H2TMRos. It was shown that 50 µM L-arginine stimulates the energy-dependent accumulation of Ca2+ in mitochondria using the fluorescent probe Fluo-4 AM. A similar effect occurred when using nitric oxide donors 100 µM SNP, SNAP, and sodium nitrite (SN) directly. The stimulating effect was eliminated in the presence of the NO scavenger C-PTIO. Nitric oxide reduces the electrical potential in mitochondria without causing them to swell. The stimulatory effect of spermine on the accumulation of Ca2+ by mitochondria is attributed to the enhancement of NO synthesis, which was demonstrated with the use of C-PTIO, NO-synthase inhibitors (100 µM NA and L-NAME), as well as by direct monitoring of NO synthesis fluorescent probe DAF-FM. A conclusion was drawn about the potential regulatory effect of the product of the oxidative metabolism of L-arginine - NO on the transport of Ca2+ in the mitochondria of the myometrium, as well as the corresponding effect of the product of non-oxidative metabolism -spermine by increasing the synthesis of NO in these subcellular structures.


Subject(s)
Arginine , Calcium , Nitric Oxide , Female , Animals , Arginine/metabolism , Calcium/metabolism , Rats , Nitric Oxide/metabolism , Oxidation-Reduction , Myometrium/metabolism , Myometrium/drug effects , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/drug effects , Rats, Wistar , Mitochondria/metabolism , Mitochondria/drug effects , Uterus/metabolism , Uterus/drug effects , Spermine/metabolism , Spermine/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/drug effects , Biological Transport/drug effects
13.
Fitoterapia ; 177: 106091, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38908760

ABSTRACT

Nitric oxide (NO) is an important gas messenger molecule with a wide range of biological functions. High concentration of NO exerts promising antitumor effects and is regarded as one of the hot spots in cancer research, that have limitations in their direct application due to its gaseous state, short half-life (seconds) and high reactivity. Lysyl oxidase (LOX) is a copper-dependent amine oxidase that is responsible for the covalent bonding between collagen and elastin and promotes tumor cell invasion and metastasis. The overexpression of LOX in triple-negative breast cancer (TNBC) makes it an attractive target for TNBC therapy. Herein, novel NO donor prodrug molecules were designed and synthesized based on the naturally derived piperlongumine (PL) skeleton, which can be selectively activated by LOX to release high concentrations of NO and PL derivatives, both of them play a synergistic role in TNBC therapy. Among them, the compound TM-1 selectively released NO in highly invasive TNBC cells (MDA-MB-231), and TM-1 was also confirmed as a potential TNBC cell line inhibitor with an inhibitory concentration of 2.274 µM. Molecular docking results showed that TM-1 had a strong and selective binding affinity with LOX protein.


Subject(s)
Dioxolanes , Drug Design , Molecular Docking Simulation , Nitric Oxide , Protein-Lysine 6-Oxidase , Triple Negative Breast Neoplasms , Protein-Lysine 6-Oxidase/metabolism , Humans , Triple Negative Breast Neoplasms/drug therapy , Dioxolanes/pharmacology , Dioxolanes/chemistry , Cell Line, Tumor , Nitric Oxide/metabolism , Molecular Structure , Nitric Oxide Donors/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Piperidones
14.
J Biomed Mater Res B Appl Biomater ; 112(7): e35442, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923117

ABSTRACT

The development of drug-resistant microorganisms is taking a heavy toll on the biomedical world. Clinical infections are costly and becoming increasingly dangerous as bacteria that once responded to standard antibiotic treatment are developing resistance mechanisms that require innovative treatment strategies. Nitric oxide (NO) is a gaseous molecule produced endogenously that has shown potent antibacterial capabilities in numerous research studies. Its multimechanistic antibacterial methods prevent the development of resistance and have shown potential as an alternative to antibiotics. However, there has yet to be a direct comparison study evaluating the antibacterial properties of NO against antibiotic susceptible and antibiotic-resistant clinically isolated bacterial strains. Herein, standardized lab and clinically isolated drug-resistant bacterial strains are compared side-by-side for growth and viability following treatment with NO released from S-nitrosoglutathione (GSNO), an NO donor molecule. Evaluation of growth kinetics revealed complete killing of E. coli lab and clinical strains at 17.5 mM GSNO, though 15 mM displayed >50% killing and significantly reduced metabolic activity, with greater dose dependence for membrane permeability. Clinical P. aeruginosa showed greater susceptibility to GSNO during growth curve studies, but metabolic activity and membrane permeability demonstrated similar effects for 12.5 mM GSNO treatment of lab and clinical strains. MRSA lab and clinical strains exhibited total killing at 17.5 mM treatment, though metabolic activity was decreased, and membrane permeation began at 12.5 mM for both strains. Lastly, both S. epidermidis strains were killed by 15 mM GSNO, with sensitivities in metabolic activity and membrane permeability at 12.5 mM GSNO. The mirrored antibacterial effects seen by the lab and clinical strains of two Gram-negative and two Gram-positive bacteria reveal the translational success of NO as an antibacterial therapy and potential alternative to standard antibiotic treatment.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Nitric Oxide , Nitric Oxide/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemistry , Drug Resistance, Bacterial/drug effects , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
15.
ACS Appl Mater Interfaces ; 16(19): 24248-24260, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38693878

ABSTRACT

Biomedical devices are vulnerable to infections and biofilm formation, leading to extended hospital stays, high expenditure, and increased mortality. Infections are clinically treated via the administration of systemic antibiotics, leading to the development of antibiotic resistance. A multimechanistic strategy is needed to design an effective biomaterial with broad-spectrum antibacterial potential. Recent approaches have investigated the fabrication of innately antimicrobial biomedical device surfaces in the hope of making the antibiotic treatment obsolete. Herein, we report a novel fabrication strategy combining antibacterial nitric oxide (NO) with an antibiofilm agent N-acetyl cysteine (NAC) on a polyvinyl chloride surface using polycationic polyethylenimine (PEI) as a linker. The designed biomaterial could release NO for at least 7 days with minimal NO donor leaching under physiological conditions. The proposed surface technology significantly reduced the viability of Gram-negative Escherichia coli (>97%) and Gram-positive Staphylococcus aureus (>99%) bacteria in both adhered and planktonic forms in a 24 h antibacterial assay. The composites also exhibited a significant reduction in biomass and extra polymeric substance accumulation in a dynamic environment over 72 h. Overall, these results indicate that the proposed combination of the NO donor with mucolytic NAC on a polymer surface efficiently resists microbial adhesion and can be used to prevent device-associated biofilm formation.


Subject(s)
Acetylcysteine , Anti-Bacterial Agents , Biofilms , Escherichia coli , Nitric Oxide , Staphylococcus aureus , Acetylcysteine/chemistry , Acetylcysteine/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Microbial Sensitivity Tests , Polyvinyl Chloride/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology
16.
Placenta ; 151: 59-66, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718734

ABSTRACT

INTRODUCTION: To evaluate the maternal and fetal hemodynamic effects of treatment with a nitric oxide donor and oral fluid in pregnancies complicated by fetal growth restriction. METHODS: 30 normotensive participants with early fetal growth restriction were enrolled. 15 participants were treated until delivery with transdermal glyceryl trinitrate and oral fluid intake (Treated group), and 15 comprised the untreated group. All women underwent non-invasive assessment of fetal and maternal hemodynamics and repeat evaluation 2 weeks later. RESULTS: In the treated group, maternal hemodynamics improved significantly after two weeks of therapy compared to untreated participants. Fetal hemodynamics in the treated group showed an increase in umbilical vein diameter by 18.87 % (p < 0.01), in umbilical vein blood flow by 48.16 % (p < 0.01) and in umbilical vein blood flow corrected for estimated fetal weight by 30.03 % (p < 0.01). In the untreated group, the characteristics of the umbilical vein were unchanged compared to baseline. At the same time, the cerebro-placental ratio increased in the treated group, while it was reduced in the untreated group, compared to baseline values. The treated group showed a higher birthweight centile (p = 0.03) and a lower preeclampsia rate (p = 0.04) compared to the untreated group. DISCUSSION: The combined therapeutic approach with nitric oxide donor and oral fluid intake in fetal growth restriction improves maternal hemodynamics, which becomes more hyperdynamic (volume-dominant). At the same time, in the fetal circuit, umbilical vein flow increased and fetal brain sparing improved. Although a modest sample size, there was less preeclampsia and a higher birthweight suggesting beneficial maternal and fetal characteristics of treatment.


Subject(s)
Fetal Growth Retardation , Nitric Oxide Donors , Umbilical Veins , Humans , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/physiopathology , Pregnancy , Pilot Projects , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/administration & dosage , Adult , Nitroglycerin/pharmacology , Nitroglycerin/administration & dosage , Hemodynamics/drug effects , Fetus/blood supply , Fetus/metabolism , Young Adult , Oxygen/metabolism , Oxygen/blood
17.
Acta Biomater ; 182: 28-41, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38761961

ABSTRACT

The regenerative microenvironment after peripheral nerve injury is imbalanced and difficult to rebalance, which is mainly affected by inflammation, oxidative stress, and inadequate blood supply. The difficulty in remodeling the nerve regeneration microenvironment is the main reason for slow nerve regeneration. Traditional drug treatments have certain limitations, such as difficulty in penetrating the blood-nerve barrier and lack of pleiotropic effects. Therefore, there is an urgent need to build multifunctional nerve grafts that can effectively regulate the regenerative microenvironment and promote nerve regeneration. Nitric oxide (NO), a highly effective gas transmitter with diatomic radicals, is an important regulator of axonal growth and migration, synaptic plasticity, proliferation of neural precursor cells, and neuronal survival. Moreover, NO provides potential anti-inflammation, anti-oxidation, and blood vessel promotion applications. However, excess NO may cause cell death and neuroinflammatory cell damage. The prerequisite for NO treatment of peripheral nerve injury is that it is gradually released over time. In this study, we constructed an injectable NO slow-release system with two main components, including macromolecular NO donor nanoparticles (mPEG-P(MSNO-EG) nanoparticles, NO-NPs) and a carrier for the nanoparticles, mPEG-PA-PP injectable temperature-sensitive hydrogel. Due to the multiple physiological regulation of NO and better physiological barrier penetration, the conduit effectively regulates the inflammatory response and oxidative stress of damaged peripheral nerves, promotes nerve vascularization, and nerve regeneration and docking, accelerating the nerve regeneration process. STATEMENT OF SIGNIFICANCE: The slow regeneration speed of peripheral nerves is mainly due to the destruction of the regeneration microenvironment. Neural conduits with drug delivery capabilities have the potential to improve the microenvironment of nerve regeneration. However, traditional drugs are hindered by the blood nerve barrier and cannot effectively target the injured area. NO, an endogenous gas signaling molecule, can freely cross the blood nerve barrier and act on target cells. However, excessive NO can lead to cell apoptosis. In this study, a NO sustained-release system was constructed to regulate the microenvironment of nerve regeneration through various pathways and promote nerve regeneration.


Subject(s)
Delayed-Action Preparations , Nerve Regeneration , Nitric Oxide , Animals , Nitric Oxide/metabolism , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/therapy , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Rats, Sprague-Dawley , Rats , Peripheral Nerves/drug effects , Peripheral Nerves/pathology , Nanoparticles/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Male , Hydrogels/chemistry , Sciatic Nerve/drug effects
18.
J Control Release ; 371: 16-28, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763388

ABSTRACT

Metastasis leads to high mortality among cancer patients. It is a complex, multi-step biological process that involves the dissemination of cancer cells from the primary tumor and their systemic spread throughout the body, primarily through the epithelial-mesenchymal transition (EMT) program and immune evasion mechanisms. It presents a challenge in how to comprehensively treat metastatic cancer cells throughout the entire stage of the metastatic cascade using a simple system. Here, we fabricate a nanogel (HNO-NG) by covalently crosslinking a macromolecular nitric oxide (NO) donor with a photothermal IR780 iodide-containing hyaluronic acid derivative via a click reaction. This enables stable storage and tumor-targeted, photothermia-triggered release of NO to combat tumor metastasis throughout all stages. Upon laser irradiation (HNO-NG+L), the surge in NO production within tumor cells impairs the NF-κB/Snail/RKIP signaling loop that promotes the EMT program through S-nitrosylation, thus inhibiting cell dissemination from the primary tumor. On the other hand, it induces immunogenic cell death (ICD) and thereby augments anti-tumor immunity, which is crucial for killing both the primary tumor and systemically distributed tumor cells. Therefore, HNO-NG+L, by fully leveraging EMT reversal, ICD induction, and the lethal effect of NO, achieved impressive eradication of the primary tumor and significant prevention of lung metastasis in a mouse model of orthotropic 4T1 breast tumor that spontaneously metastasizes to the lungs, extending the NO-based therapeutic approach against tumor metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , Mice, Inbred BALB C , Nanogels , Nitric Oxide , Animals , Epithelial-Mesenchymal Transition/drug effects , Nanogels/chemistry , Nanogels/administration & dosage , Female , Cell Line, Tumor , Neoplasm Metastasis/prevention & control , Humans , Mice , Hyaluronic Acid/chemistry , Hyaluronic Acid/administration & dosage , Polyethyleneimine/chemistry , Polyethyleneimine/administration & dosage , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacology , Photothermal Therapy/methods , Polyethylene Glycols
19.
Fitoterapia ; 176: 105964, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38663561

ABSTRACT

Berberine was used as the lead compound in the present study to design and synthesize novel berberine derivatives by splicing bromine bridges of different berberine carbon chain lengths coupled nitric oxide donors, and their lipid lowering activities were assessed in a variety of ways. This experiment synthesized 17 new berberine nitric oxide donor derivatives. Compared with berberine hydrochloride, most of the compounds exhibited certain glycerate inhibitory activity, and compounds 6a, 6b, 6d, 12b and 12d showed higher inhibitory activity than berberine, with 6a, 6b and 6d having significant inhibitory activity. In addition, compound 6a linked to furazolidone nitric oxide donor showed better NO release in experiments; In further mechanistic studies, we screened and got two proteins, PCSK9 and ACLY, and docked two proteins with 17 compounds, and found that most of the compounds bound better with ATP citrate lyase (ACLY), among which there may be a strong interaction between compound 6a and ACLY, and the interaction force was better than the target drug Bempedoic Acid, which meaning that 6a may exert hypolipidemic effects by inhibiting ACLY; moreover, we also found that 6a may had the better performance in gastrointestinal absorption, blood-brain barrier permeability, Egan, Muegge class drug principle model calculation and bioavailability.


Subject(s)
Berberine , Hypolipidemic Agents , Nitric Oxide Donors , Berberine/pharmacology , Berberine/analogs & derivatives , Berberine/chemical synthesis , Berberine/chemistry , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/chemical synthesis , Hypolipidemic Agents/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/chemical synthesis , Nitric Oxide Donors/chemistry , Humans , Molecular Structure , ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , ATP Citrate (pro-S)-Lyase/metabolism , Proprotein Convertase 9/metabolism , Molecular Docking Simulation , Animals , Blood-Brain Barrier/drug effects , Nitric Oxide/metabolism , PCSK9 Inhibitors
20.
Biomed Pharmacother ; 174: 116540, 2024 May.
Article in English | MEDLINE | ID: mdl-38579400

ABSTRACT

Rheumatoid arthritis (RA) is characterized by high level of reactive oxygen species (ROS) and proinflammatory cytokines, which facilitate the activation of the inflammatory signaling such as NF-κB pathway and exacerbate the development of inflammation. Herein, we designed a nanodrug by encapsulating the NO donor S-nitrosoglutathione (GSNO) into an emulsion and coating the surface with a polydopamine (PDA) layer to yield GSNO@PDA, which simultaneously scavenged the extra ROS and suppressed NF-κB signaling for potent RA treatment. To enhance the cellular uptake and NO generation efficiency, dextran sulfate (DS) and Cu2+ were anchored on the surface of GSNO@PDA to obtain the final formulation GSNO@PDA@DS. Our results demonstrated that GSNO@PDA@DS were successfully prepared and the modification of DS effectively boosted the cellular uptake of GSNO@PDA@DS. Moreover, GSNO@PDA@DS lowered cellular ROS and elevated intracellular NO, resulting in a decrease of M1 phenotype, inhibition of NF-κB pathway and down-regulation of proinflammatory cytokine tumor necrosis factor-α (TNF-α). Further in vivo studies confirmed that GSNO@PDA@DS significantly relieved symptoms and bone erosion by regulating the microenvironment of RA, highlighting the potential of GSNO@PDA@DS for RA therapy through ROS scavenging and NO-mediated suppression of inflammatory signaling.


Subject(s)
Arthritis, Rheumatoid , NF-kappa B , Nitric Oxide Donors , Polymers , Reactive Oxygen Species , S-Nitrosoglutathione , Reactive Oxygen Species/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Animals , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/administration & dosage , Mice , NF-kappa B/metabolism , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/administration & dosage , RAW 264.7 Cells , Polymers/chemistry , Indoles/pharmacology , Indoles/administration & dosage , Free Radical Scavengers/pharmacology , Free Radical Scavengers/administration & dosage , Drug Synergism , Male , Signal Transduction/drug effects , Dextran Sulfate , Tumor Necrosis Factor-alpha/metabolism , Nitric Oxide/metabolism , Drug Delivery Systems/methods
SELECTION OF CITATIONS
SEARCH DETAIL