Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.638
Filter
1.
Biochem Pharmacol ; 222: 116075, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38395266

ABSTRACT

Cancer is recognized as the major cause of death worldwide and the most challenging public health issues. Tumor cells exhibit molecular adaptations and metabolic reprograming to sustain their high proliferative rate and autophagy plays a pivotal role to supply the high demand for metabolic substrates and for recycling cellular components, which has attracted the attention of the researchers. The modulation of the autophagic process sensitizes tumor cells to chemotherapy-induced cell death and reverts drug resistance. In this regard, many in vitro and in vivo studies having shown the anticancer activity of phenothiazine (PTZ) derivatives due to their potent cytotoxicity in tumor cells. Interestingly, PTZ have been used as antiemetics in antitumor chemotherapy-induced vomiting, maybe exerting a combined antitumor effect. Among the mechanisms of cytotoxicity, the modulation of autophagy by these drugs has been highlighted. Therefore, the use of PTZ derivatives can be considered as a repurposing strategy in antitumor chemotherapy. Here, we provided an overview of the effects of antipsychotic PTZ on autophagy in tumor cells, evidencing the molecular targets and discussing the underlying mechanisms. The modulation of autophagy by PTZ in tumor cells have been consistently related to their cytotoxic action. These effects depend on the derivative, their concentration, and also the type of cancer. Most data have shown the impairment of autophagic flux by PTZ, probably due to the blockade of lysosome-autophagosome fusion, but some studies have also suggested the induction of autophagy. These data highlight the therapeutic potential of targeting autophagy by PTZ in cancer chemotherapy.


Subject(s)
Antineoplastic Agents , Antipsychotic Agents , Neoplasms , Humans , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Drug Repositioning , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Autophagy , Neoplasms/drug therapy , Cell Line, Tumor , Apoptosis
3.
Microbiol Spectr ; 11(4): e0102323, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37341603

ABSTRACT

Infections by pathogenic Acinetobacter species represent a significant burden on the health care system, despite their relative rarity, due to the difficulty of treating infections through oral antibiotics. Multidrug resistance is commonly observed in clinical Acinetobacter infections and multiple molecular mechanisms have been identified for this resistance, including multidrug efflux pumps, carbapenemase enzymes, and the formation of bacterial biofilm in persistent infections. Phenothiazine compounds have been identified as a potential inhibitor of type IV pilus production in multiple Gram-negative bacterial species. Here, we report the ability of two phenothiazines to inhibit type IV pilus-dependent surface (twitching) motility and biofilm formation in multiple Acinetobacter species. Biofilm formation was inhibited in both static and continuous flow models at micromolar concentrations without significant cytotoxicity, suggesting that type IV pilus biogenesis was the primary molecular target for these compounds. These results suggest that phenothiazines may be useful lead compounds for the development of biofilm dispersal agents against Gram-negative bacterial infections. IMPORTANCE Acinetobacter infections are a growing burden on health care systems worldwide due to increasing antimicrobial resistance through multiple mechanisms. Biofilm formation is an established mechanism of antimicrobial resistance, and its inhibition has the potential to potentiate the use of existing drugs against pathogenic Acinetobacter. Additionally, as discussed in the manuscript, anti-biofilm activity by phenothiazines has the potential to help to explain their known activity against other bacteria, including Staphylococcus aureus and Mycobacterium tuberculosis.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Humans , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Bacteria , Drug Resistance, Multiple, Bacterial
4.
Eur J Med Chem ; 254: 115337, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37060756

ABSTRACT

Cancer is a leading cause of death worldwide and there are still limited options for cure. Chemotherapy is the most significant treatment for cancer which increased survival rates, despite this, it is associated with numerous side effects, as well as cancer relapsing due to drug resistance insurgence; consequently, it is still a challenging task to develop new potent and less toxic anti-cancer agents for patients' care. Phenothiazine moiety, which leads a class of well-known antipsychotic drugs, possesses a wide range of biological activities and has been also introduced in cancer chemotherapy. This review aims in disclosing the use of phenothiazines during the last five years for the development of different anti-cancer drug candidates. The design and the synthetic strategies adopted, the SAR investigations and the role of reviewed phenothiazine derivatives as anti-cancer agents and multi-drug resistance (MDR) reversals are here fully described and discussed.


Subject(s)
Antineoplastic Agents , Antipsychotic Agents , Humans , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Drug Resistance, Multiple
5.
Neurosci Biobehav Rev ; 141: 104809, 2022 10.
Article in English | MEDLINE | ID: mdl-35970416

ABSTRACT

Over a century ago, the phenothiazine dye, methylene blue, was discovered to have both antipsychotic and anti-cancer effects. In the 20th-century, the first phenothiazine antipsychotic, chlorpromazine, was found to inhibit cancer. During the years of elucidating the pharmacology of the phenothiazines, reserpine, an antipsychotic with a long historical background, was likewise discovered to have anti-cancer properties. Research on the effects of antipsychotics on cancer continued slowly until the 21st century when efforts to repurpose antipsychotics for cancer treatment accelerated. This review examines the history of these developments, and identifies which antipsychotics might treat cancer, and which cancers might be treated by antipsychotics. The review also describes the molecular mechanisms through which antipsychotics may inhibit cancer. Although the overlap of molecular pathways between schizophrenia and cancer have been known or suspected for many years, no comprehensive review of the subject has appeared in the psychiatric literature to assess the significance of these similarities. This review fills that gap and discusses what, if any, significance the similarities have regarding the etiology of schizophrenia.


Subject(s)
Antipsychotic Agents , Neoplasms , Schizophrenia , Antipsychotic Agents/therapeutic use , Chlorpromazine/therapeutic use , Humans , Methylene Blue/therapeutic use , Neoplasms/drug therapy , Phenothiazines/therapeutic use , Reserpine/therapeutic use , Schizophrenia/drug therapy
6.
Arch Virol ; 167(11): 2203-2212, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35920983

ABSTRACT

Dengue virus (DENV), a member of the genus Flavivirus, family Flaviviridae, is the most widespread viral pathogen transmitted to humans by mosquitoes. Despite the increased incidence of DENV infection, there are no antiviral drugs available for treatment or prevention. Phenothiazines are heterocyclic compounds with various pharmacological properties that are very adaptable for drug repurposing. In the present report, we analyzed the antiviral activity against DENV and the related Zika virus (ZIKV) of trifluoperazine (TFP), a phenothiazine derivative in clinical use as an antipsychotic and antiemetic agent. TFP exhibited dose-dependent inhibitory activity against the four DENV serotypes and ZIKV in monkey Vero cells at non-cytotoxic concentrations with 50% effective concentration values in the range 1.6-6.4 µM. A similar level of antiviral efficacy was exhibited by TFP against flavivirus infection in the human cell lines A549 and HepG2. Mechanistic studies, performed using time-dependent infectivity assays, real-time RT-PCR, Western blot, and immunofluorescence techniques, indicated that uncoating of the virus during penetration into the cell was the main target for TFP in infected cells, but the compound also exerted a minor effect on a late stage of the virus multiplication cycle. This study demonstrates that TFP, a pharmacologically active phenothiazine, is a selective inhibitor of DENV multiplication in cell culture. Our findings open perspectives for the repositioning of phenothiazines like TFP with a wide spectrum of antiviral efficacy as potential agents for the control of pathogenic flaviviruses.


Subject(s)
Antiemetics , Antipsychotic Agents , Dengue Virus , Dengue , Zika Virus Infection , Zika Virus , Animals , Antiemetics/pharmacology , Antiemetics/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Dengue/drug therapy , Humans , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Trifluoperazine/pharmacology , Trifluoperazine/therapeutic use , Vero Cells , Virus Replication
7.
Bioorg Med Chem Lett ; 74: 128911, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35907606

ABSTRACT

Ferroptosis was an iron-dependent, nonapoptotic form of regulated cell death. In our previous study, we discovered a potent ferroptosis inhibitor with phenothiazine scaffold (1), but subsequent investigation showed that this compound had potent hERG binding affinity. Herein, we report the discovery of a series of 2-vinyl-10H-phenothiazine derivatives as new class of ferroptosis inhibitors. Structure-activity relationship (SAR) analyses led to the identification of compound 7j, which exhibited significantly reduced hERG inhibition (IC50 > 30 µM) while maintaining high ferroptosis inhibitory activity (EC50 = 0.001 µM on the erastin-induced HT1080 cell ferroptosis model). Further studies confirmed 7j acted as a ROS scavenger and could relieve DOX-induced cardiomyopathy. 7j also displayed favorable pharmacokinetic properties and exhibited no obvious toxicity in vivo and vitro. Overall, this study provides a promising lead compound for drug discovery targeting ferroptosis.


Subject(s)
Cardiomyopathies , Ferroptosis , Ethers , Humans , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Structure-Activity Relationship
8.
Acad Emerg Med ; 29(9): 1118-1131, 2022 09.
Article in English | MEDLINE | ID: mdl-35138658

ABSTRACT

OBJECTIVES: This review was designated to evaluate the efficacy of parenteral ketorolac in treating acute migraine headache. METHODS: We searched databases Cochrane Central Register of Controlled Trials (CENTRAL), Medline, and Google Scholar up to January 2021 and identified randomized controlled trials comparing ketorolac to any other medications in treating patients presenting with migraine headache. RESULTS: Thirteen trials were included in our review, comprising 944 participants. We derived seven comparisons: ketorolac versus phenothiazines, metoclopramide, sumatriptan, dexamethasone, sodium valproate, caffeine, and diclofenac. There were no significant differences in the reduction of pain intensity at 1 h under the comparisons between ketorolac and phenothiazines (standard mean difference [SMD] = 0.09, p = 0.74) or metoclopramide (SMD = 0.02, p = 0.95). We also found no difference in the outcome recurrence of headache (ketorolac vs. phenothiazines (risk ratio [RR] =0.98, p = 0.97)], ability to return to work or usual activity (ketorolac vs. metoclopramide [RR = 0.64, p = 0.13]), need for rescue medication (ketorolac vs. phenothiazines [RR = 1.72, p = 0.27], ketorolac vs. metoclopramide [RR 2.20, p = 0.18]), and frequency of adverse effects (ketorolac vs. metoclopramide [RR = 1.07, p = 0.82]). Limited trials suggested that ketorolac offered better pain relief at 1 h compared to sumatriptan and dexamethasone; had lesser frequency of adverse effects than phenothiazines; and was superior to sodium valproate in terms of reduction of pain intensity at 1 h, need for rescue medication, and sustained headache freedom within 24 h. CONCLUSIONS: Ketorolac may have similar efficacy to phenothiazines and metoclopramide in treating acute migraine headache. Ketorolac may also offer better pain control than sumatriptan, dexamethasone, and sodium valproate. However, given the lack of evidence due to inadequate number of trials available, future studies are warranted.


Subject(s)
Ketorolac , Migraine Disorders , Caffeine/therapeutic use , Dexamethasone/therapeutic use , Diclofenac/therapeutic use , Humans , Ketorolac/therapeutic use , Metoclopramide/adverse effects , Migraine Disorders/drug therapy , Pain/drug therapy , Phenothiazines/therapeutic use , Sumatriptan/therapeutic use , Valproic Acid/therapeutic use
9.
Molecules ; 27(1)2022 Jan 03.
Article in English | MEDLINE | ID: mdl-35011508

ABSTRACT

The molecular hybridization approach has been used to develop compounds with improved efficacy by combining two or more pharmacophores of bioactive scaffolds. In this context, hybridization of various relevant pharmacophores with phenothiazine derivatives has resulted in pertinent compounds with diverse biological activities, interacting with specific or multiple targets. In fact, the development of new drugs or drug candidates based on phenothiazine system has been a promising approach due to the diverse activities associated with this tricyclic system, traditionally present in compounds with antipsychotic, antihistaminic and antimuscarinic effects. Actually, the pharmacological actions of phenothiazine hybrids include promising antibacterial, antifungal, anticancer, anti-inflammatory, antimalarial, analgesic and multi-drug resistance reversal properties. The present review summarizes the progress in the development of phenothiazine hybrids and their biological activity.


Subject(s)
Drug Development , Phenothiazines , Animals , Humans , Phenothiazines/chemical synthesis , Phenothiazines/chemistry , Phenothiazines/therapeutic use , Structure-Activity Relationship
10.
Rev. psiquiatr. Urug ; 85(1): 28-42, oct. 2021. graf, tab
Article in Spanish | LILACS, UY-BNMED, BNUY | ID: biblio-1343130

ABSTRACT

El tratamiento farmacológico de demostrada eficacia en la esquizofrenia es el antipsicótico. Sin embargo, en muchas ocasiones se requiere medicación concomitante que depende de comorbilidades y efectos adversos. Se realizó un estudio cuantitativo, longitudinal, retrospectivo, considerando el año 2006 y 2016, en una población de usuarios con esquizofrenia de la Policlínica del Hospital Vilardebó, analizando los tratamientos con psicofármacos. Se diferenciaron los tratamientos según monoterapia antipsicótica y polifarmacia con 2 antipsicóticos, y polifarmacia con más de 2 antipsicóticos, antidepresivos, estabilizantes del humor, benzodiacepinas y anticolinérgicos. La población inicial en 2006 fue de 621 pacientes y 398 pacientes continuaban en tratamiento en 2016. Mantuvieron el trata-miento con antipsicóticos 377 pacientes; 184 mantuvieron benzodiacepinas; 59 se mantuvieron con anticolinérgicos; 49, con estabilizantes del humor y 47, con antidepresivos. La monoterapia antipsicótica se presentó en torno al 50 % de la población estudiada. Se deberían revisar aquellas prácticas que se infieren a partir de este estudio, como el uso prolongado de anticolinérgicos, benzodiacepinas, y polifarmacia con más de 2 antipsicóticos, que está extendida en los usuarios con esquizofrenia. El tratamiento con clozapina fue el más estable y no parece aumentar la mortalidad en estos pacientes


Antipsychotics are the proved effective therapy for schizophrenia. However, on many occasions, associated drugs are required depending on comorbidities and side effects. A retrospective longitudinal quantitative study of drug prescription for 2006 and 2016 in patients with schizophrenia diagnosis was carried out in an outpatient clinic at Hospital Vilardebó. Treatments were classified as antipsychotic monotherapy, two antipsychotic drugs polypharmacy and polypharmacy with two antipsychotic drugs, antidepressants, mood stabilizers, benzodiazepines and anticholinergic drugs. Initial population in 2006 included 621 patients, 398 were still being treated in 2016. Antipsychotic drugs were still being received in 377 patients, benzodiazepines in 184, anticholinergic drugs in 59, mood stabilizers in 49, and anti-depressants in 47. Antipsychotic monotherapy was 50% of the population. Those practices that can be inferred from this study, with lengthy use of anticholinergic drugs, benzodiazepines, and the use of more than 2 antipsychotic drugs in patients with schizophrenia diagnosis should be revised. Clozapine therapy was the most stable and does not seem to increase mortality.


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Schizophrenia/drug therapy , Antipsychotic Agents/therapeutic use , Drug Therapy/statistics & numerical data , Phenothiazines/therapeutic use , Chlorpromazine/therapeutic use , Epidemiology, Descriptive , Retrospective Studies , Cohort Studies , Clozapine/therapeutic use , Risperidone/therapeutic use , Polypharmacy , Age and Sex Distribution , Tiapride Hydrochloride/therapeutic use , Quetiapine Fumarate/therapeutic use , Aripiprazole/therapeutic use , Olanzapine/therapeutic use , Haloperidol/therapeutic use , Methotrimeprazine/therapeutic use
11.
Mol Neurobiol ; 58(12): 6136-6152, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34455546

ABSTRACT

A depressive or hibernation-like effect of chlorpromazine and promethazine (C + P) on brain activity was reported to induce neuroprotection, with or without induced-hypothermia. However, the underlying mechanisms remain unclear. The current study evaluated the pharmacological function of C + P on the inhibition of neuroinflammatory response and inflammasome activation after ischemia/reperfusion. A total of 72 adult male Sprague-Dawley rats were subjected to 2 h middle cerebral artery occlusion (MCAO) followed by 6 or 24 h reperfusion. At the onset of reperfusion, rats received C + P (8 mg/kg) with temperature control. Brain cell death was detected by measuring CD68 and myeloperoxidase (MPO) levels. Inflammasome activation was measured by mRNA levels of NLRP3, IL-1ß, and TXNIP, and protein quantities of NLRP3, IL-1ß, TXNIP, cleaved-Caspase-1, and IL-18. Activation of JAK2/STAT3 pathway was detected by the phosphorylation of STAT3 (p-STAT3) and JAK2 (p-JAK2), and the co-localization of p-STAT3 and NLRP3. Activation of the p38 pathway was assessed with the protein levels of p-p38/p38. The mRNA and protein levels of HIF-1α, FoxO1, and p-FoxO1, and the co-localization of p-STAT3 with HIF-1α or FoxO1 were quantitated. As expected, C + P significantly reduced cell death and attenuated the neuroinflammatory response as determined by reduced CD68 and MPO. C + P decreased ischemia-induced inflammasome activation, shown by reduced mRNA and protein expressions of NLRP3, IL-1ß, TXNIP, cleaved-Caspase-1, and IL-18. Phosphorylation of JAK2/STAT3 and p38 pathways and the co-localization of p-STAT3 with NLRP3 were also inhibited by C + P. Furthermore, mRNA levels of HIF-1α and FoxO1 were decreased in the C + P group. While C + P inhibited HIF-1α protein expression, it increased FoxO1 phosphorylation, which promoted the exclusion of FoxO1 from the nucleus and inhibited FoxO1 activity. At the same time, C + P reduced the co-localization of p-STAT3 with HIF-1α or FoxO1. In conclusion, C + P treatment conferred neuroprotection in stroke by suppressing neuroinflammation and NLRP3 inflammasome activation. The present study suggests that JAK2/STAT3/p38/HIF-1α/FoxO1 are vital regulators and potential targets for efficacious therapy following ischemic stroke.


Subject(s)
Inflammasomes/drug effects , Ischemic Stroke/drug therapy , Neuroinflammatory Diseases/drug therapy , Phenothiazines/therapeutic use , Animals , Cell Death/drug effects , Cytokines/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemic Stroke/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/metabolism , Phenothiazines/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
Theranostics ; 11(14): 7005-7017, 2021.
Article in English | MEDLINE | ID: mdl-34093867

ABSTRACT

The tumor suppressor protein p53 remains in a wild type but inactive form in ~50% of all human cancers. Thus, activating it becomes an attractive approach for targeted cancer therapies. In this regard, our lab has previously discovered a small molecule, Inauhzin (INZ), as a potent p53 activator with no genotoxicity. Method: To improve its efficacy and bioavailability, here we employed nanoparticle encapsulation, making INZ-C, an analog of INZ, to nanoparticle-encapsulated INZ-C (n-INZ-C). Results: This approach significantly improved p53 activation and inhibition of lung and colorectal cancer cell growth by n-INZ-C in vitro and in vivo while it displayed a minimal effect on normal human Wi38 and mouse MEF cells. The improved activity was further corroborated with the enhanced cellular uptake observed in cancer cells and minimal cellular uptake observed in normal cells. In vivo pharmacokinetic evaluation of these nanoparticles showed that the nanoparticle encapsulation prolongates the half-life of INZ-C from 2.5 h to 5 h in mice. Conclusions: These results demonstrate that we have established a nanoparticle system that could enhance the bioavailability and efficacy of INZ-C as a potential anti-cancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Indoles/pharmacology , Lung Neoplasms/drug therapy , Nanoparticles/chemistry , Phenothiazines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/therapeutic use , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Phenothiazines/chemistry , Phenothiazines/pharmacokinetics , Phenothiazines/therapeutic use , Spectroscopy, Fourier Transform Infrared , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
13.
Eur J Med Chem ; 219: 113419, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33845233

ABSTRACT

The pathogenesis of Alzheimer's disease (AD) has been associated with dysregulation of histone deacetylases (HDACs). Previously, acridine-based HDAC inhibitors have shown potential in ameliorating HDAC activity and enhancing neurite outgrowth. In this study, the acridine ring was modified using various phenothiazine derivatives. Several resulting compounds exhibited potent enzyme-inhibiting activity towards class II HDACs when compared to the clinically approved HDAC inhibitor SAHA. Compound 4f demonstrated the highest class II HDAC inhibition (IC50 = 4.6-600 nM), as well as promotion of neurite outgrowth. Importantly, compound 4f displayed no cytotoxicity against neuron cells. Compound 4f was further evaluated for cellular effects. Altogether, these findings show a potential strategy in HDAC inhibition for treatment of the neurological disease.


Subject(s)
Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylases/chemistry , Hydroxamic Acids/chemistry , Phenothiazines/chemistry , Acetylation/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Binding Sites , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Molecular Docking Simulation , Neurites/drug effects , Neurites/physiology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Phenothiazines/metabolism , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
14.
ACS Appl Mater Interfaces ; 13(10): 11588-11596, 2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33656316

ABSTRACT

Increasing threats from both pathogenic infections and antibiotic resistance highlight the pressing demand for nonantibiotic agents and alternative therapies. Herein, we report several new phenothiazinium-based derivatives, which could be readily synthesized via fragment-based assembly, which exhibited remarkable bactericidal activities both in vitro and in vivo. Importantly, in contrast to numerous clinically and preclinically used antibacterial photosensitizers, these compounds were able to eliminate various types of microorganisms, including Gram-(+) Staphylococcus aureus (S. aureus), Gram-(-) Escherichia coli, multidrug-resistant S. aureus, and their associated biofilms, at low drug and light dosages (e.g., 0.21 ng/mL in vitro and 1.63 ng/cm2 in vivo to eradicate S. aureus at 30 J/cm2). This study thus unveils the potential of these novel phenothiaziniums as potent antimicrobial agents for highly efficient photodynamic antibacterial chemotherapy.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Escherichia coli/drug effects , Escherichia coli/physiology , Escherichia coli Infections/drug therapy , Humans , Mice , Phenothiazines/chemistry , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology
15.
Photodiagnosis Photodyn Ther ; 34: 102198, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33578028

ABSTRACT

AIM: The aim of the present study was to compare the effects of the phenothiazine photosensitizers methylene blue (MB), toluidine blue-O (TBO) and butyl toluidine blue (BuTB) in antimicrobial photodynamic therapy (aPDT), as adjuvant therapy to scaling and root planing (SRP) in the treatment of experimental periodontitis (EP) in rats. MATERIAL AND METHODS: 120 Wistar rats underwent ligation around the lower left molar. After seven days, the ligature was removed. The animals were separated into the following groups (n = 15): EP, no treatment; SRP, SRP and irrigation with saline solution; MB, SRP and deposition of MB; TBO, SRP and deposition of TBO; BuTB, SRP and deposition of BuTB; MB-aPDT, SRP and aPDT with MB; TBO-aPDT, SRP and aPDT with TBO and; BuTB-aPDT, SRP and aPDT with BuTB. The aPDT session was performed after SRP, with deposition of the photosensitizer and irradiation with a diode laser (DL; InGaAlP, 660 nm, 40 mW, 60 s, 2.4 J). Histological and histometric analysis was performed. RESULTS: BuTB-aPDT group had a lesser extent of the inflammatory process compared to the EP, SRP, MB and TBO at all experimental periods (p < 0.05). At 15 days, the aPDT treated groups had a greater bone tissue structure than groups EP and SRP (p < 0.05) The BuTB showed lower Alveolar Bone Loss (ABL) compared to the TBO-aPDT group at 30 days (p < 0.05). CONCLUSION: aPDT using the photosensitizer BuTB proved to be the adjuvant therapy that most favored the reduction of inflammatory infiltrate in the furcation area and ABL.


Subject(s)
Periodontitis , Photochemotherapy , Animals , Dental Scaling , Periodontitis/drug therapy , Phenothiazines/pharmacology , Phenothiazines/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Rats , Rats, Wistar , Root Planing
16.
Eur J Med Chem ; 209: 112842, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33065375

ABSTRACT

Ferroptosis is a new type of programmed cell death discovered recently and has been demonstrated to be involved in a number of human diseases such as ischemic stroke. Ferroptosis inhibitors are expected to have potential to treat these diseases. Herein, we report the identification of promethazine derivatives as a new type of ferroptosis inhibitors. Structure-activity relationship (SAR) analyses led to the discovery of the most potent compound 2-(1-(4-(4-methylpiperazin-1-yl)phenyl)ethyl)-10H-phenothiazine (51), which showed an EC50 (half maximal effective concentration) value of 0.0005 µM in the erastin-induced HT1080 cell ferroptosis model. In the MCAO (middle cerebral artery occlusion) ischemic stroke model, 51 presented an excellent therapeutic effect. This compound also displayed favorable pharmacokinetic properties, in particular, a good ability to permeate the blood-brain barrier. Overall, 51 could be a promising lead compound for the treatment of ferroptosis related diseases and deserves further investigations.


Subject(s)
Ferroptosis/drug effects , Ischemic Stroke/drug therapy , Phenothiazines/chemistry , Phenothiazines/pharmacology , Animals , Cell Line , Humans , Ischemic Stroke/pathology , Male , Phenothiazines/pharmacokinetics , Phenothiazines/therapeutic use , Rats, Sprague-Dawley
18.
Eur J Pharmacol ; 887: 173553, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32949606

ABSTRACT

In 2020 the whole world focused on antivirus drugs towards SARS-CoV-2. Most of the researchers focused on drugs used in other viral infections or malaria. We have not seen such mobilization towards one topic in this century. The whole situation makes clear that progress needs to be made in antiviral drug development. The first step to do it is to characterize the potential antiviral activity of new or already existed drugs on the market. Phenothiazines are antipsychotic agents used previously as antiseptics, anthelminthics, and antimalarials. Up to date, they are tested for a number of other disorders including the broad spectrum of viruses. The goal of this paper was to summarize the current literature on activity toward RNA-viruses of such drugs like chlorpromazine, fluphenazine, perphenazine, prochlorperazine, and thioridazine. We identified 49 papers, where the use of the phenothiazines for 23 viruses from different families were tested. Chlorpromazine, fluphenazine, perphenazine, prochlorperazine, and thioridazine possess anti-viral activity towards different types of viruses. These drugs inhibit clathrin-dependent endocytosis, cell-cell fusion, infection, replication of the virus, decrease viral invasion as well as suppress entry into the host cells. Additionally, since the drugs display activity at nontoxic concentrations they have therapeutic potential for some viruses, still, further research on animal and human subjects are needed in this field to verify cell base research.


Subject(s)
Antipsychotic Agents/pharmacology , Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Phenothiazines/pharmacology , Pneumonia, Viral/drug therapy , RNA Viruses/drug effects , Animals , Antipsychotic Agents/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 , Chlorpromazine/pharmacology , Chlorpromazine/therapeutic use , Fluphenazine/pharmacology , Fluphenazine/therapeutic use , Humans , Pandemics , Perphenazine/pharmacology , Perphenazine/therapeutic use , Phenothiazines/therapeutic use , Prochlorperazine/pharmacology , Prochlorperazine/therapeutic use , SARS-CoV-2 , Thioridazine/pharmacology , Thioridazine/therapeutic use , COVID-19 Drug Treatment
19.
FASEB J ; 34(10): 13959-13977, 2020 10.
Article in English | MEDLINE | ID: mdl-32851720

ABSTRACT

Growing evidence supports a central role of NADPH oxidases (NOXs) in the regulation of platelets, which are circulating cells involved in both hemostasis and thrombosis. Here, the use of Nox1-/- and Nox1+/+ mice as experimental models of human responses demonstrated a critical role of NOX1 in collagen-dependent platelet activation and pathological arterial thrombosis, as tested in vivo by carotid occlusion assays. In contrast, NOX1 does not affect platelet responses to thrombin and normal hemostasis, as assayed in tail bleeding experiments. Therefore, as NOX1 inhibitors are likely to have antiplatelet effects without associated bleeding risks, the NOX1-selective inhibitor 2-acetylphenothiazine (2APT) and a series of its derivatives generated to increase inhibitory potency and drug bioavailability were tested. Among the 2APT derivatives, 1-(10H-phenothiazin-2-yl)vinyl tert-butyl carbonate (2APT-D6) was selected for its high potency. Both 2APT and 2APT-D6 inhibited collagen-dependent platelet aggregation, adhesion, thrombus formation, superoxide anion generation, and surface activation marker expression, while responses to thrombin or adhesion to fibrinogen were not affected. In vivo administration of 2APT or 2APT-D6 led to the inhibition of mouse platelet aggregation, oxygen radical output, and thrombus formation, and carotid occlusion, while tail hemostasis was unaffected. Differently to in vitro experiments, 2APT-D6 and 2APT displayed similar potency in vivo. In summary, NOX1 inhibition with 2APT or its derivative 2APT-D6 is a viable strategy to control collagen-induced platelet activation and reduce thrombosis without deleterious effects on hemostasis. These compounds should, therefore, be considered for the development of novel antiplatelet drugs to fight cardiovascular diseases in humans.


Subject(s)
Carotid Artery Thrombosis/drug therapy , Enzyme Inhibitors/pharmacology , NADPH Oxidase 1/antagonists & inhibitors , Phenothiazines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Animals , Carotid Artery Thrombosis/prevention & control , Cells, Cultured , Collagen/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Fibrinogen/metabolism , Hemorrhage/etiology , Humans , Mice , Mice, Inbred C57BL , Phenothiazines/adverse effects , Phenothiazines/therapeutic use , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , Thrombin/metabolism
20.
J Med Chem ; 63(9): 4896-4907, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32267685

ABSTRACT

Development of new photosensitizers (PSs) with high photodynamic efficacy and minimal side effects is of great interest in photodynamic therapy (PDT). In this work, we reported several pyridine-embedded phenothiazinium (pyridophenothiazinium) dyes, which could be conveniently synthesized in a few short steps and acted as highly efficient and potent PSs to selectively localize to lysosomes and photosensitively kill cancer cells. Among them, compound 5, which possessed the ability of promoting intracellular reactive oxygen species (ROS) upon light irradiation by almost 40-fold higher than that of methylene blue (MB, a general phenothiazinium-based PS), exhibited a remarkable phototherapeutic index (PI = 53.8) against HT29 cancer cells, leading to eradication of large solid tumors (∼300 mm3) in a xenograft mouse model without apparent side effects. These results suggest that the pyridophenothiazinium dyes developed herein, especially compound 5, may serve as promising lysosome-targeted PSs for efficient photodynamic antitumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Coloring Agents/therapeutic use , Lysosomes/metabolism , Neoplasms/drug therapy , Phenothiazines/therapeutic use , Photosensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Coloring Agents/chemical synthesis , Coloring Agents/pharmacokinetics , Coloring Agents/radiation effects , Female , Humans , Light , Male , Mice, Inbred BALB C , Phenothiazines/chemical synthesis , Phenothiazines/pharmacokinetics , Phenothiazines/radiation effects , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/radiation effects , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Pyridines/radiation effects , Pyridines/therapeutic use , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...