Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 386
Filtrar
1.
BMC Vet Res ; 20(1): 196, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741109

RESUMEN

BACKGROUND: Hypoxia is a detrimental factor in solid tumors, leading to aggressiveness and therapy resistance. OMX, a tunable oxygen carrier from the heme nitric oxide/oxygen-binding (H-NOX) protein family, has the potential to reduce tumor hypoxia. [18F]Fluoromisonidazole ([18F]FMISO) positron emission tomography (PET) is the most widely used and investigated method for non-invasive imaging of tumor hypoxia. In this study, we used [18F]FMISO PET/CT (computed tomography) to assess the effect of OMX on tumor hypoxia in spontaneous canine tumors. RESULTS: Thirteen canine patients with various tumors (n = 14) were randomly divided into blocks of two, with the treatment groups alternating between receiving intratumoral (IT) OMX injection (OMX IT group) and intravenous (IV) OMX injection (OMX IV group). Tumors were regarded as hypoxic if maximum tumor-to-muscle ratio (TMRmax) was greater than 1.4. In addition, hypoxic volume (HV) was defined as the region with tumor-to-muscle ratio greater than 1.4 on [18F]FMISO PET images. Hypoxia was detected in 6/7 tumors in the OMX IT group and 5/7 tumors in the OMX IV injection group. Although there was no significant difference in baseline hypoxia between the OMX IT and IV groups, the two groups showed different responses to OMX. In the OMX IV group, hypoxic tumors (n = 5) exhibited significant reductions in tumor hypoxia, as indicated by decreased TMRmax and HV in [18F]FMISO PET imaging after treatment. In contrast, hypoxic tumors in the OMX IT group (n = 6) displayed a significant increase in [18F]FMISO uptake and variable changes in TMRmax and HV. CONCLUSIONS: [18F]FMISO PET/CT imaging presents a promising non-invasive procedure for monitoring tumor hypoxia and assessing the efficacy of hypoxia-modulating therapies in canine patients. OMX has shown promising outcomes in reducing tumor hypoxia, especially when administered intravenously, as evident from reductions in both TMRmax and HV in [18F]FMISO PET imaging.


Asunto(s)
Enfermedades de los Perros , Misonidazol , Neoplasias , Tomografía Computarizada por Tomografía de Emisión de Positrones , Hipoxia Tumoral , Animales , Perros , Misonidazol/análogos & derivados , Tomografía Computarizada por Tomografía de Emisión de Positrones/veterinaria , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Enfermedades de los Perros/diagnóstico por imagen , Enfermedades de los Perros/tratamiento farmacológico , Femenino , Hipoxia Tumoral/efectos de los fármacos , Masculino , Neoplasias/veterinaria , Neoplasias/tratamiento farmacológico , Neoplasias/diagnóstico por imagen , Tiosemicarbazonas/uso terapéutico , Tiosemicarbazonas/farmacología , Complejos de Coordinación
2.
J Med Chem ; 67(7): 5744-5757, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38553427

RESUMEN

To develop a next-generation metal agent and dual-agent multitargeted combination therapy, we developed a copper (Cu) compound based on the properties of the human serum albumin (HSA)-indomethacin (IND) complex to remodel the tumor microenvironment (TME). We optimized a series of Cu(II) isopropyl 2-pyridyl ketone thiosemicarbazone compounds to obtain a Cu(II) compound (C4) with significant cytotoxicity and then constructed an HSA-IND-C4 complex (HSA-IND-C4) delivery system. IND and C4 bind to the hydrophobic cavities of the IB and IIA domains of HSA, respectively. In vivo, the HSA-IND-C4 not only showed enhanced antitumor efficacy relative to C4 and C4 + IND but also improved their targeting ability and decreased their side effects. The antitumor mechanism of C4 + IND involved acting on the different components of the TME. IND inhibited tumor-related inflammation, while C4 not only induced apoptosis and autophagy of cancer cells but also inhibited tumor angiogenesis.


Asunto(s)
Antineoplásicos , Neoplasias , Profármacos , Tiosemicarbazonas , Humanos , Albúmina Sérica Humana/química , Cobre/química , Albúmina Sérica/química , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Indometacina/uso terapéutico , Microambiente Tumoral , Profármacos/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Neoplasias/tratamiento farmacológico
3.
ACS Chem Neurosci ; 15(7): 1432-1455, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38477556

RESUMEN

Alzheimer's disease (AD) is the most prevalent cause of dementia characterized by a progressive cognitive decline. Addressing neuroinflammation represents a promising therapeutic avenue to treat AD; however, the development of effective antineuroinflammatory compounds is often hindered by their limited blood-brain barrier (BBB) permeability. Consequently, there is an urgent need for accurate, preclinical AD patient-specific BBB models to facilitate the early identification of immunomodulatory drugs capable of efficiently crossing the human AD BBB. This study presents a unique approach to BBB drug permeability screening as it utilizes the familial AD patient-derived induced brain endothelial-like cell (iBEC)-based model, which exhibits increased disease relevance and serves as an improved BBB drug permeability assessment tool when compared to traditionally employed in vitro models. To demonstrate its utility as a small molecule drug candidate screening platform, we investigated the effects of diacetylbis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(atsm)) and a library of metal bis(thiosemicarbazone) complexes─a class of compounds exhibiting antineuroinflammatory therapeutic potential in neurodegenerative disorders. By evaluating the toxicity, cellular accumulation, and permeability of those compounds in the AD patient-derived iBEC, we have identified 3,4-hexanedione bis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(dtsm)) as a candidate with good transport across the AD BBB. Furthermore, we have developed a multiplex approach where AD patient-derived iBEC were combined with immune modulators TNFα and IFNγ to establish an in vitro model representing the characteristic neuroinflammatory phenotype at the patient's BBB. Here, we observed that treatment with CuII(dtsm) not only reduced the expression of proinflammatory cytokine genes but also reversed the detrimental effects of TNFα and IFNγ on the integrity and function of the AD iBEC monolayer. This suggests a novel pathway through which copper bis(thiosemicarbazone) complexes may exert neurotherapeutic effects on AD by mitigating BBB neuroinflammation and related BBB integrity impairment. Together, the presented model provides an effective and easily scalable in vitro BBB platform for screening AD drug candidates. Its improved translational potential makes it a valuable tool for advancing the development of metal-based compounds aimed at modulating neuroinflammation in AD.


Asunto(s)
Enfermedad de Alzheimer , Tiosemicarbazonas , Humanos , Barrera Hematoencefálica/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Cobre/metabolismo , Enfermedades Neuroinflamatorias , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/metabolismo , Tiosemicarbazonas/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo
4.
Int Immunopharmacol ; 126: 111259, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37992446

RESUMEN

Multiple studies in the literature have demonstrated that synthetic compounds containing heterocyclic rings possess a reparative potential against acute and chronic inflammation. In the present study, two novel thiosemicarbazone derivatives based on l-ethyl-6-(thiophen-2-yl)indoline-2,3-dione with different phenyl substituted thiosemicarbazides were synthesized by condensation reaction and the structures of proposed target compounds (KP-2 and KP-5) were confirmed by UV-VIS, FTIR, 1H-NMR and 13C-NMR. In-vitro anti-inflammatory behavior of KP-2 and KP-5 was confirmed by bovine serum albumin (BSA) and ovine serum albumin (OSA) analysis. Acute and chronic anti-inflammatory potential of synthesized compounds were evaluated by using carrageenan and complete Freund's adjuvant (CFA) as inflammation-inducing agents, respectively. Inhibition of pro-inflammatory mediators and prevention of protein denaturation owing to synchronization of more electronegative flouro-groups substituted on phenyl rings along with heterocyclic indoline ring provides anti-inflammatory effects and are corroborated by radiological, histopathological analysis. Additional support was provided through density functional theory (DFT) and molecular docking. KP-5 exhibited excellent lead-likeness based on its physicochemical parameters, making it a viable drug candidate. The synthesized compounds also showed promising ADMET properties, enhancing their potential as therapeutic agents. These findings emphasize the pivotal role of new compounds for drug design and development.


Asunto(s)
Tiosemicarbazonas , Animales , Ovinos , Humanos , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Antiinflamatorios/uso terapéutico , Inflamación/tratamiento farmacológico , Carragenina , Estructura Molecular , Edema/inducido químicamente , Edema/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa 2/farmacología
5.
J Mol Recognit ; 36(12): e3059, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37723924

RESUMEN

Donepezil is one of the most used drugs in the treatment of Alzheimer's disease. Its activity as an AChE inhibitor makes new studies with these enzyme inhibitors attractive. For this purpose, in this study, 12 compounds including thiosemicarbazone pharmacophore, have been synthesized for the treatment of the Alzheimer's disease. 3,4-Dimethoxybenzene or 1,3-benzodioxolone rings were used for the PAS region. The substituted piperazine benzene structure is preferred for the CAS region. At the same time, the thiosemicarbazone pharmacophore structure with known ChE enzyme inhibition potential was used as a bridge connecting the CAS and PAS regions. Structure determination of compounds 3a-3l were revealed using 13 C-NMR, 1 H-NMR, and HRMS spectroscopic methods. The inhibition profile of obtained compounds (3a-3l) against ChE was evaluated using in vitro modified Ellman method. Compounds 3a, 3b, 3f, 3g and 3i exhibited inhibitory activity against the AChE enzyme. Compound 3a showed the highest inhibitory potential with an IC50 = 0.030 ± 0.001 µM. As a result of molecular docking studies, compound 3a displayed important interactions compared to other active derivatives. Molecular dynamics studies are important to see the stability of the complex formed by ligand and protein. RMSD, RMSF ang Rg parameters were calculated via dynamic studies. In conclusion, compound 3a may be a potential AChE enzyme inhibitor with its strong inhibitory potential and behavior in silico.


Asunto(s)
Enfermedad de Alzheimer , Tiosemicarbazonas , Humanos , Simulación del Acoplamiento Molecular , Enfermedad de Alzheimer/tratamiento farmacológico , Simulación de Dinámica Molecular , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/química , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Inhibidores Enzimáticos/farmacología , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Relación Estructura-Actividad , Estructura Molecular
6.
Pharmacol Res ; 193: 106806, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37244387

RESUMEN

The estrogen receptor-α (ER-α) is a key driver of breast cancer (BC) and the ER-antagonist, tamoxifen, is a central pillar of BC treatment. However, cross-talk between ER-α, other hormone and growth factor receptors enables development of de novo resistance to tamoxifen. Herein, we mechanistically dissect the activity of a new class of anti-cancer agents that inhibit multiple growth factor receptors and down-stream signaling for the treatment of ER-positive BC. Using RNA sequencing and comprehensive protein expression analysis, we examined the activity of di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), on the expression and activation of hormone and growth factor receptors, co-factors, and key resistance pathways in ER-α-positive BC. DpC differentially regulated 106 estrogen-response genes, and this was linked to decreased mRNA levels of 4 central hormone receptors involved in BC pathogenesis, namely ER, progesterone receptor (PR), androgen receptor (AR), and prolactin receptor (PRL-R). Mechanistic investigation demonstrated that due to DpC and Dp44mT binding metal ions, these agents caused a pronounced decrease in ER-α, AR, PR, and PRL-R protein expression. DpC and Dp44mT also inhibited activation and down-stream signaling of the epidermal growth factor (EGF) family receptors, and expression of co-factors that promote ER-α transcriptional activity, including SRC3, NF-κB p65, and SP1. In vivo, DpC was highly tolerable and effectively inhibited ER-α-positive BC growth. Through bespoke, non-hormonal, multi-modal mechanisms, Dp44mT and DpC decrease the expression of PR, AR, PRL-R, and tyrosine kinases that act with ER-α to promote BC, constituting an innovative therapeutic approach.


Asunto(s)
Neoplasias de la Mama , Tiosemicarbazonas , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Progesterona/uso terapéutico , Andrógenos/uso terapéutico , Receptores de Prolactina , Prolactina/uso terapéutico , Tamoxifeno/farmacología , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Receptores ErbB , Estrógenos/uso terapéutico
7.
Neuroscience ; 509: 125-131, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36436699

RESUMEN

CuII(atsm) is a blood-brain barrier permeant copper(II) compound that is under investigation in human clinical trials for the treatment of neurodegenerative diseases of the central nervous system (CNS). Imaging in humans by positron emission tomography shows the compound accumulates in affected regions of the CNS in patients. Most therapeutic studies to date have utilised oral administration of CuII(atsm) in an insoluble form, as either solid tablets or a liquid suspension. However, two pre-clinical studies have demonstrated disease-modifying outcomes following transdermal application of soluble CuII(atsm) prepared in dimethyl sulphoxide. Whether differences in the method of administration lead to different degrees of tissue accumulation of the compound has never been examined. Here, we compare the two methods of administration in wild-type mice by assessing changes in tissue concentrations of copper. Both administration methods resulted in elevated copper concentrations in numerous tissues, with the largest increases evident in the liver, brain and spinal cord. In all instances where treatment with CuII(atsm) resulted in elevated tissue copper, transdermal application of soluble CuII(atsm) led to higher concentrations of copper. In contrast to CuII(atsm), an equivalent dose of copper(II) chloride resulted in minimal changes to tissue copper concentrations, regardless of the administration method. Data presented herein provide quantitative insight to transdermal application of soluble CuII(atsm) as a potential alternative to oral administration of the compound in an insoluble formulation.


Asunto(s)
Compuestos Organometálicos , Tiosemicarbazonas , Ratones , Humanos , Animales , Compuestos Organometálicos/uso terapéutico , Cobre , Tiosemicarbazonas/uso terapéutico , Médula Espinal/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Tomografía de Emisión de Positrones
8.
Mol Biochem Parasitol ; 252: 111520, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36122704

RESUMEN

Schistosomiasis is a neglected disease that affects about 258 million people worldwide. Caused by Schistosoma mansoni, helminth which, in Brazil, it is present on 19 states and capital. Praziquantel (PZQ) treatment presents low efficacy and adverse effects in parasites juvenile stages. Thiosemicarbazones and thiazolidinones are rising as potent chemical groups that have biological activity wide spectrum, and with radical modifications, they may become more effective and selective. Aiming to evaluate the action of these molecules against S. mansoni, JF series thiosemicarbazones and thiazolidinones (LqIT/UFPE) were synthesized: JF30, JF31, JF33, JF34, JF35, JF36, JF38, JF39, JF42 and JF43. Several parameters were evaluated, such as: their cytotoxicity in VERO cells, in vitro schistosomicidal activity for juvenile and adult worms and their action on worms through ultrastructural changes. Cytotoxicity indices ranged from 272 µM to 725 µM. When evaluating mortality rate, adult and juvenile worms showed 100 % mortality rate within 24 h and 48 h, respectively, when exposed to the compounds JF31 and JF43 at a dose of 200 µM. Also, motility, mortality and oviposition parameters were evaluated: JF31 and JF43 presented a score of 0 in 24 h, meaning total absence of movement, whereas no eggs and soft tissue damage were observed under optical microscopy. Through scanning electron microscopy, integumentary alterations caused by the compounds JF31 and JF43 were observed, such as: exposure of the musculature, formation of integumentary bubbles, integuments with abnormal morphology and destruction of tubercles and spikes. The results shoerd that the compound JF31 was 2.39 times more selective for adult worms and JF43 was 3.74 times more selective for juvenile worms. Thus, the compounds JF43 and JF31 are the most promising for presenting schistosomicidal activity of S. mansoni.


Asunto(s)
Esquistosomiasis mansoni , Esquistosomicidas , Tiosemicarbazonas , Chlorocebus aethiops , Animales , Femenino , Esquistosomicidas/farmacología , Esquistosomicidas/uso terapéutico , Schistosoma mansoni , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Células Vero , Praziquantel/farmacología , Esquistosomiasis mansoni/tratamiento farmacológico
9.
Microbiol Spectr ; 10(3): e0195121, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35412374

RESUMEN

Metal chelation is generally considered as a promising antifungal approach but its specific mechanisms are unclear. Here, we identify 13 thiosemicarbazone derivatives that exert broad-spectrum antifungal activity with potency comparable or superior to that of fluconazole in vitro by screening a small compound library comprising 89 thiosemicarbazone derivatives as iron chelators. Among the hits, 19ak exhibits minimal cytotoxicity and potent activity against either azole-sensitive or azole-resistant fungal pathogens. Mechanism investigations reveal that 19ak inhibits mitochondrial respiration mainly by retarding mitochondrial respiratory chain complex I activity through iron chelation, and further reduces mitochondrial membrane potential and ATP synthesis in Candida albicans. In addition, 19ak inhibits fungal ribosome biogenesis mainly by disrupting intracellular zinc homeostasis. 19ak also stimulates the activities of antioxidant enzymes and decreases reactive oxygen species formation in C. albicans, resulting in an increase in detrimental intracellular reductive stress. However, 19ak has minor effects on mammalian cells in depleting intracellular iron and zinc. Moreover, 19ak exhibits low capacity to induce drug resistance and in vivo efficacy in a Galleria mellonella infection model. These findings uncover retarded fungal mitochondrial respiration and ribosome biogenesis as downstream effects of disruption of iron and zinc homeostasis in C. albicans and provide a basis for the thiosemicarbazone 19ak in antifungal application. IMPORTANCE The increasing incidence of fungal infections and resistance to existing antifungals call for the development of broad-spectrum antifungals with novel mechanisms of action. In this study, we demonstrate that a thiosemicarbazone derivative 19ak selectively inhibits mitochondrial respiration mainly by retarding mitochondrial respiratory chain complex I activity through iron chelation and inhibits ribosome biogenesis mainly by disrupting intracellular zinc homeostasis in C. albicans. In addition, 19ak exhibits low capacity to induce fungal resistance, minimal cytotoxicity, and in vivo antifungal efficacy. This study provides the basis of thiosemicarbazone derivative 19ak as a metal chelator for the treatment of fungal infections.


Asunto(s)
Micosis , Tiosemicarbazonas , Animales , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Azoles/farmacología , Azoles/uso terapéutico , Candida albicans , Farmacorresistencia Fúngica , Hierro/farmacología , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Mamíferos , Pruebas de Sensibilidad Microbiana , Micosis/tratamiento farmacológico , Respiración , Ribosomas , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Zinc
10.
Semin Cancer Biol ; 79: 58-67, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-32741700

RESUMEN

Dysfunction of the TP53 (p53) gene occurs in most if not all human malignancies. Two principal mechanisms are responsible for this dysfunction; mutation and downregulation of wild-type p53 mediated by MDM2/MDM4. Because of its almost universal inactivation in malignancy, p53 is a highly attractive target for the development of new anticancer drugs. Although multiple strategies have been investigated for targeting dysfunctional p53 for cancer treatment, only 2 of these have so far yielded compounds for testing in clinical trials. These strategies include the identification of compounds for reactivating the mutant form of p53 back to its wild-type form and compounds for inhibiting the interaction between wild-type p53 and MDM2/MDM4. Currently, multiple p53-MDM2/MDM4 antagonists are undergoing clinical trials, the most advanced being idasanutlin which is currently undergoing testing in a phase III clinical trial in patients with relapsed or refractory acute myeloid leukemia. Two mutant p53-reactivating compounds have progressed to clinical trials, i.e., APR-246 and COTI-2. Although promising data has emerged from the testing of both MDM2/MDM4 inhibitors and mutant p53 reactivating compounds in preclinical models, it is still unclear if these agents have clinical efficacy. However, should any of the compounds currently being evaluated in clinical trials be shown to have efficacy, it is likely to usher in a new era in cancer treatment, especially as p53 dysfunction is so prevalent in human cancers.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Aminoquinolinas/uso terapéutico , Proteínas de Ciclo Celular/metabolismo , Humanos , Neoplasias/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Pirrolidinas/uso terapéutico , Quinuclidinas/uso terapéutico , Tiosemicarbazonas/uso terapéutico , Proteína p53 Supresora de Tumor/genética , para-Aminobenzoatos/uso terapéutico
11.
Molecules ; 28(1)2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36615218

RESUMEN

Triazole-based thiosemicarbazone derivatives (6a-u) were synthesized then characterized by spectroscopic techniques, such as 1HNMR and 13CNMR and HRMS (ESI). Newly synthesized derivatives were screened in vitro for inhibitory activity against acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes. All derivatives (except 6c and 6d, which were found to be completely inactive) demonstrated moderate to good inhibitory effects ranging from 0.10 ± 0.050 to 12.20 ± 0.30 µM (for AChE) and 0.20 ± 0.10 to 14.10 ± 0.40 µM (for BuChE). The analogue 6i (IC50 = 0.10 ± 0.050 for AChE and IC50 = 0.20 ± 0.050 µM for BuChE), which had di-substitutions (2-nitro, 3-hydroxy groups) at ring B and tri-substitutions (2-nitro, 4,5-dichloro groups) at ring C, and analogue 6b (IC50 = 0.20 ± 0.10 µM for AChE and IC50 = 0.30 ± 0.10 µM for BuChE), which had di-Cl at 4,5, -NO2 groups at 2-position of phenyl ring B and hydroxy group at ortho-position of phenyl ring C, emerged as the most potent inhibitors of both targeted enzymes (AChE and BuChE) among the current series. A structure-activity relationship (SAR) was developed based on nature, position, number, electron donating/withdrawing effects of substitution/s on phenyl rings. Molecular docking studies were used to describe binding interactions of the most active inhibitors with active sites of AChE and BuChE.


Asunto(s)
Enfermedad de Alzheimer , Inhibidores de la Colinesterasa , Tiosemicarbazonas , Humanos , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad , Tiosemicarbazonas/síntesis química , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico
12.
Cell Oncol (Dordr) ; 44(6): 1307-1323, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34586588

RESUMEN

PURPOSE: Association of the metal chelating portion of thiosemicarbazone with the cytotoxic activity of sigma-2 receptors appears a promising strategy for the treatment of pancreatic tumors. Here, we developed a novel sigma-2 receptor targeting thiosemicarbazone (FA4) that incorporates a moiety associated with lysosome destabilization and ROS increase in order to design more efficient antitumor agents. METHODS: The density of sigma receptors in pancreatic cancer cells was evaluated by flow cytometry. In these cells, cytotoxicity (MTT assay) and activation of ER- and mitochondria-dependent cell death pathways (mRNA expression of GRP78, ATF6, IRE1, PERK; ROS levels by MitoSOX and DCFDA-AM; JC-1 staining) induced by the thiosemicarbazones FA4, MLP44, PS3 and ACthio-1, were evaluated. The expression of autophagic proteins (ATG5, ATG7, ATG12, beclin, p62 and LC3-I) was also studied. In addition, the in vivo effect of FA4 in xenograft models with and without gemcitabine challenge was investigated. RESULTS: We found that FA4 exerted a more potent cytotoxicity than previously studied thiosemicarbazones (MLP44, PS3 and ACthio-1), which were found to display variable effects on the ER or the mitochondria-dependent pro-apoptotic axis. By contrast, FA4 activated pro-apoptotic pathways and decreased autophagy, except in MiaPaCa2 cells, in which autophagic proteins were expressed at lower levels and remained unmodified by FA4. FA4 treatment of PANC-1 xenografted mouse models, poorly responsive to conventional chemotherapy, significantly reduced tumor volumes and increased intratumor apoptosis compared to gemcitabine, with no signs of toxicity. CONCLUSIONS: Our data indicate that FA4 exhibits encouraging activity in pancreatic cancer cells unresponsive to gemcitabine. These results warrant further investigation in patient-derived pancreatic cancers, and hold promise for the development of therapies that can more efficiently target the specific characteristics of individual tumor types.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Receptores sigma/metabolismo , Tiosemicarbazonas/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Neoplasias Pancreáticas/genética , Especies Reactivas de Oxígeno/metabolismo , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacología
13.
Toxicol Mech Methods ; 31(8): 581-588, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34240667

RESUMEN

Diabetes mellitus (DM) is chronic and metabolic disorder, which is mainly attributed by hyperglycemia. Vanadium salts and their oxo-complexes have been shown to possess insulin-mimetic and anti-diabetic activities in animal models and diabetic patients. The main goal of this study was to investigate the protective effect of oxovanadium(IV) complex based on thiosemicarbazone (VOL) [L: (N(1)-2,4-dihydroxybenzylidene-N-(4)-2-hydroxybenzylidene-S-methyl-isothiosemicarbazidato-oxovanadium(IV)] on glycoprotein components levels and oxidative lung injury of streptozotocin (STZ)-induced diabetic rats. Male Swiss albino rats were separated into four groups. Group I (n = 5): Control (normal) animals, Group II (n = 5): Control animals administered with VOL, Group III (n = 6): STZ-induced diabetic animals, and Group IV (n = 5): STZ-induced diabetic rats treated with VOL. VOL was given to the experimental animals by gavage at a dose of 0.2 mM/kg body weight every day for 12 days. Diabetes was induced by single intraperitoneal injection of STZ (65 mg/kg body weight). On the 12th day, lung tissue samples were taken. Glycoprotein components, advanced oxidation protein products, protein carbonyl, hydroxyproline levels, and prolidase, arginase, xanthine oxidase, catalase, superoxide dismutase, glutathione peroxidase, glutathione reductase, glutathione-S-transferase and adenosine deaminase activities significantly increased whereas aryl esterase, paraoxonase-1, carbonic anhydrase, Na+/K+-ATPase activities remarkably decreased in lung tissue of diabetic rats. Treatment with VOL reversed these effects showing a beneficial effect. The present study shows that VOL has a protective effect against diabetes-induced lung damage as well as on abnormal glycoprotein component levels.


Asunto(s)
Diabetes Mellitus Experimental , Lesión Pulmonar , Tiosemicarbazonas , Animales , Antioxidantes , Glucemia , Diabetes Mellitus Experimental/tratamiento farmacológico , Glicoproteínas , Humanos , Pulmón , Masculino , Estrés Oxidativo , Ratas , Tiosemicarbazonas/uso terapéutico
14.
Int Immunopharmacol ; 96: 107778, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34162145

RESUMEN

Following induction of inflammation, the nuclear factor kappa B (NF-κB) in activated macrophages induces the transcription of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and cyclooxygenase (COX), an inflammatory enzyme implicated in the synthesis of prostaglandins (PGs). The latter are involved in the transition and the maintenance of chronic inflammation underling various chronic disorders that require treatment. Concerning this, many anti-inflammatory drugs are available to treat the inflammatory disorders, but their therapeutic use is associated with a variety of side effects. Therefore, the discovery of new safer and potential anti-inflammatory drugs is necessary. In this regard, thiosemicarbazones (TSC) compounds and their metals complexes attracted high interest due to their wide range of biological activities, interestingly, the anti-inflammatory activity. They are formed by the action of thiosemicarbazide on an aldehyde or ketone, and contain a sulfur atom in place of the oxygen atom. Their ability to form a stable complex with transition metal is known to enhances the biological activity and reduces the side effects of the parent compound. Thus, this review article describes the inflammatory response mediated by NF-κB-COX-PGs and summarizes the anti-inflammatory activity of different thiosemicarbazones derivatives synthesized in research area.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacología , Animales , Antiinflamatorios/uso terapéutico , FN-kappa B/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/metabolismo , Tiosemicarbazonas/uso terapéutico
15.
Bioorg Chem ; 113: 105018, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34098396

RESUMEN

Chagas disease (ChD), caused by Trypanosoma cruzi, remains a challenge for the medical and scientific fields due to the inefficiency of the therapeutic approaches available for its treatment. Thiosemicarbazones and hydrazones present a wide spectrum of bioactivities and are considered a platform for the design of new anti-T. cruzi drug candidates. Herein, the potential antichagasic activities of [(E)-2-(1-(4-chlorophenylthio)propan-2-ylidene)-hydrazinecarbothioamides] (C1, C3), [(E)-N'-(1-((4-chlorophenyl)thio)propan-2-ylidene)benzohydrazide] (C2), [(E)-2-(1-(4-, and [(E)-2-(1-((4-chlorophenyl)thio)propan-2-ylidene)hydrazinecarboxamide] (C4) were investigated. Macrophages (MOs) from C57BL/6 mice stimulated with C1 and C3, but not with C2 and C4, reduced amastigote replication and trypomastigote release, independent of nitric oxide (NO) and reactive oxygen species production and indoleamine 2,3-dioxygenase activity. C3, but not C1, reduced parasite uptake by MOs and potentiated TNF production. In cardiomyocytes, C3 reduced trypomastigote release independently of NO, TNF, and IL-6 production. C1 and C3 were non-toxic to the host cells. A reduction of parasite release was found during infection of MOs with trypomastigotes pre-incubated with C1 or C3 and MOs pre-stimulated with compounds before infection. Moreover, C1 and C3 acted directly on trypomastigotes, killing them faster than Benznidazole, and inhibited T. cruzi proliferation at various stages of its intracellular cycle. Mechanistically, C1 and C3 inhibit parasite duplication, and this process cannot be reversed by inhibiting the DNA damage response. In vivo, C1 and C3 attenuated parasitemia in T. cruzi-infected mice. Moreover, C3 loaded in a lipid nanocarrier system (nanoemulsion) maintained anti-T. cruzi activity in vivo. Collectively, these data suggest that C1 and C3 are candidates for the treatment of ChD and present activity in both the host and parasite cells.


Asunto(s)
Tiosemicarbazonas/química , Tripanocidas/química , Animales , Supervivencia Celular/efectos de los fármacos , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/patología , Cisteína Endopeptidasas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Diseño de Fármacos , Femenino , Estadios del Ciclo de Vida/efectos de los fármacos , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Conformación Molecular , Óxido Nítrico/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/metabolismo , Ratas , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/fisiología
16.
Mini Rev Med Chem ; 21(14): 1813-1829, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33583370

RESUMEN

Limonene and perillyl alcohol are natural monoterpenes that have attracted the attention of medicinal chemists due to their promising anticancer activities. Considering this, both compounds were explored as scaffolds to obtain various derivatives with anticancer activity. In this review, the data are organized for the first time, with a focus on the synthetic methods and strategies to obtain the derivatives throughout the period from 2000 to 2020. A brief discussion regarding the structure and activity relationships of the most active derivatives, stereoisomers, and their mechanisms of action is presented. Among the active compounds, a series of limonenes with thiosemicarbazone groups and perillyl alcohol hybrids with glycosides or drugs are illustrated. Taking all of this into account, this review may help researchers develop new promising anticancer candidates based on the structures of limonene and perillyl alcohol.


Asunto(s)
Antineoplásicos/química , Limoneno/química , Monoterpenos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carbohidratos/química , Supervivencia Celular/efectos de los fármacos , Humanos , Limoneno/farmacología , Limoneno/uso terapéutico , Monoterpenos/farmacología , Monoterpenos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Relación Estructura-Actividad , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico
17.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32948029

RESUMEN

The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.


Asunto(s)
Antineoplásicos/uso terapéutico , Quelantes/uso terapéutico , Hierro , Proteínas de Neoplasias/fisiología , Péptido Hidrolasas/fisiología , Inhibidores de Proteasas/uso terapéutico , Zinc , Antineoplásicos/farmacología , Línea Celular Tumoral , Transformación Celular Neoplásica , Quelantes/farmacología , Progresión de la Enfermedad , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Líquido Extracelular/enzimología , Vesículas Extracelulares/enzimología , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Calicreínas/antagonistas & inhibidores , Calicreínas/fisiología , Metaloproteinasas de la Matriz/fisiología , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Oxaprozina/farmacología , Oxaprozina/uso terapéutico , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Fenilalanina/uso terapéutico , Inhibidores de Proteasas/farmacología , Proteínas Quinasas/fisiología , Piridinas/farmacología , Piridinas/uso terapéutico , Tiofenos/farmacología , Tiofenos/uso terapéutico , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico
18.
Cancer Chemother Pharmacol ; 86(5): 633-640, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32989483

RESUMEN

PURPOSE: To investigate the metabolic pathways of triapine in primary cultures of human hepatocytes and human hepatic subcellular fractions; to investigate interactions of triapine with tenofovir and emtricitabine; and to evaluate triapine as a perpetrator of drug interactions. The results will better inform future clinical studies of triapine, a radiation sensitizer currently being studied in a phase III study. METHODS: Triapine was incubated with human hepatocytes and subcellular fractions in the presence of a number of inhibitors of drug metabolizing enzymes. Triapine depletion was monitored by LC-MS/MS. Tenofovir and emtricitabine were co-incubated with triapine in primary cultures of human hepatocytes. Triapine was incubated with a CYP probe cocktail and human liver microsomes, followed by LC-MS/MS monitoring of CYP specific metabolite formation. RESULTS: Triapine was not metabolized by FMO, AO/XO, MAO-A/B, or NAT-1/2, but was metabolized by CYP450s. CYP1A2 accounted for most of the depletion of triapine. Tenofovir and emtricitabine did not alter triapine depletion. Triapine reduced CYP1A2 activity and increased CYP2C19 activity. CONCLUSION: CYP1A2 metabolism is the major metabolic pathway for triapine. Triapine may be evaluated in cancer patients in the setting of HIV with emtricitabine or tenofovir treatment. Confirmatory clinical trials may further define the in vivo triapine metabolic fate and quantify any drug-drug interactions.


Asunto(s)
Inhibidores del Citocromo P-450 CYP1A2/farmacocinética , Inductores del Citocromo P-450 CYP2C19/farmacocinética , Neoplasias/terapia , Piridinas/farmacocinética , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Tiosemicarbazonas/farmacocinética , Células Cultivadas , Quimioradioterapia/métodos , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP1A2/metabolismo , Inhibidores del Citocromo P-450 CYP1A2/uso terapéutico , Citocromo P-450 CYP2C19/metabolismo , Inductores del Citocromo P-450 CYP2C19/uso terapéutico , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Emtricitabina/farmacocinética , Hepatocitos , Humanos , Inactivación Metabólica , Microsomas Hepáticos , Cultivo Primario de Células , Piridinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Espectrometría de Masas en Tándem , Tenofovir/farmacocinética , Tiosemicarbazonas/uso terapéutico
19.
J Inorg Biochem ; 210: 111134, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32673842

RESUMEN

Being a structural and catalytic cofactor in a number of biological pathways, copper accumulates in tumors owing to selective permeability of the cancer cell membranes. Copper(II) ion forms the active centers in a large number of metalloproteins. The coordination of Schiff's base ligands to the metal ion results in the high extent of increase in anticancer activity. The copper(II) complexes can cleave DNA through oxidative and hydrolytic pathways, cell apoptosis via intrinsic reactive oxygen species (ROS) mediated mitochondrial pathway due to excessive production of ROS and hence, are found more active than Ni and Pt complexes. Flexible Cu(I/II) redox behavior helps the copper complexes to form more potent, clinically effective and less toxic copper based antiproliferative drugs of lower IC50 value and higher growth inhibitory activity. Copper(II) complexes of thiosemicarbazones of Isatin, Pyridine, Benzoyl pyridine, Diacetyl/Dimethyl glyoxal, Acetophenone/Acetoacetanalide, Thiazole/Pyrazole, Quinoline, Carboxybenzaldehyde, Cinnamaldehyde/Cuminaldehyde, Citronellal, Chromone, Pyridoxal, 8-Ethyl-2-hydroxytricyclo (7.3.1.02,7) tridecan-13-one, Acyl Diazines, Naphthalene, Proline, 5-Formyluracil, 2-Hydroxy-8-propyltricyclo (7.3.1.02,7) tridecan-13-one, 9-cis-Retinal, Curcumin, Helicin (Salicylaldehyde-ß-D-glucoside), Thiophene carboxaldehyde, Salicylaldehyde, Iminodiacetate, and 3-Formyl-4-hydroxy benzenesulfonic acid have been found to exhibit more anticancer activity toward HCT116, MCF7, A549, U937, HeLa, HepG2, SGC-7901, A2780 cell lines than that of their corresponding thiosemicarbazones and standard topoisomerase-II inhibitors.


Asunto(s)
Antineoplásicos/uso terapéutico , Complejos de Coordinación/uso terapéutico , Neoplasias/tratamiento farmacológico , Tiosemicarbazonas/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Cobre/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Tiosemicarbazonas/química , Tiosemicarbazonas/farmacología , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología , Inhibidores de Topoisomerasa II/uso terapéutico
20.
Bioorg Chem ; 102: 104081, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32711086

RESUMEN

A new series of thiosemicarbazones were designed and synthesized. Their structures were confirmed by spectral characterization and single crystal XRD studies. Compounds MTSC-2 and ETSC-3 crystallized in the orthorhombic crystal system with space group Pbc21 andPca21respectively. Density functional theory computational studies were performed on MTSC-2 and ETSC-3 along with natural bond orbital analysis and Mulliken population analysis to study the structural and electronic properties of the thiosemicarbazones. The HOMOs of the two thiosemicarbazones are -5.2943 and -5.1133 eV respectively while the LUMOs are -1.6879 and -1.6398 eV respectively. The energy gap is 3.6064 and 3.4736 eV respectively. Molecular docking studies were performed to determine the binding mode of the thiosemicarbazones against ß-tubulin. The theoretical studies were further supplemented with tubulin polymerization inhibition assay. All the four thiosemicarbazones proved effective in inhibiting the polymerization of α- and ß-tubulin heterodimers into microtubules. The anticancer activity of these compounds showed their extreme potency against A549 and HepG2 cancer cell lines with IC50 values of 0.051 - 0.189 µm and 0.042 - 0.136 µm respectively. Compound PTSC-4 showed the highest activity both against tubulin and the two cancer cell lines. This was in correlation with the theoretical studies. Hence, these four compounds, specifically PTSC-4, can be considered to be potential leads in the development of non-metallic anticancer agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Simulación del Acoplamiento Molecular/métodos , Tiosemicarbazonas/uso terapéutico , Tubulina (Proteína)/uso terapéutico , Antineoplásicos/farmacología , Diseño de Fármacos , Humanos , Estructura Molecular , Relación Estructura-Actividad , Tiosemicarbazonas/farmacología , Tubulina (Proteína)/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...