Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Cells ; 12(5)2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36899898

RESUMO

The concept of chelation therapy as a valuable therapeutic approach in neurological disorders led us to develop multi-target, non-toxic, lipophilic, brain-permeable compounds with iron chelation and anti-apoptotic properties for neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), age-related dementia and amyotrophic lateral sclerosis (ALS). Herein, we reviewed our two most effective such compounds, M30 and HLA20, based on a multimodal drug design paradigm. The compounds have been tested for their mechanisms of action using animal and cellular models such as APP/PS1 AD transgenic (Tg) mice, G93A-SOD1 mutant ALS Tg mice, C57BL/6 mice, Neuroblastoma × Spinal Cord-34 (NSC-34) hybrid cells, a battery of behavior tests, and various immunohistochemical and biochemical techniques. These novel iron chelators exhibit neuroprotective activities by attenuating relevant neurodegenerative pathology, promoting positive behavior changes, and up-regulating neuroprotective signaling pathways. Taken together, these results suggest that our multifunctional iron-chelating compounds can upregulate several neuroprotective-adaptive mechanisms and pro-survival signaling pathways in the brain and might function as ideal drugs for neurodegenerative disorders, such as PD, AD, ALS, and aging-related cognitive decline, in which oxidative stress and iron-mediated toxicity and dysregulation of iron homeostasis have been implicated.


Assuntos
Doença de Alzheimer , Esclerose Lateral Amiotrófica , Hidroxiquinolinas , Doença de Parkinson , Camundongos , Animais , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Camundongos Endogâmicos C57BL , Quelantes de Ferro/uso terapêutico , Camundongos Transgênicos , Doença de Parkinson/patologia , Envelhecimento , Ferro/metabolismo
2.
Life Sci ; 293: 120272, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35065164

RESUMO

Carbostyrils are quinolone derivatives, with possible growth inhibition properties on cancer cells. Unlike many tumors, 15-Lipoxygenase-1 (15-LOX-1) is highly expressed in prostate cancer (PCa) cells and has oncogenic properties. Here, with the hypothesis that 6-, 7- and 8-geranyloxycarbostyril (GQ) have inhibitory properties on 15-LOX-1, their effects were assessed on PCa cells. Their cytotoxic effects were evaluated by MTT assay and mechanism of cell death was investigated using annexin V/PI staining. Finally, the anti-tumor properties of 8-GQ were assessed in immunocompromised C57BL/6 mice bearing human PCa cells. Accordingly, these compounds could effectively inhibit 15-LOX activity in PCa cells. MTT and flow cytometry tests confirmed their toxic effects on PCa cells, with no significant toxicity on normal cells, and apoptosis was the main mechanism of cell death. In vivo results indicated that use of 8-GQ at 50 mg/kg had stronger anti-tumor effects than 5 mg/kg cisplatin, with fewer side effects on normal tissues. Therefore, 8-GQ can be introduced as a potential drug candidate with 15-LOX-1 inhibitory potency, which can be effective in treatment of prostate cancer, and should be considered for further drug screening investigations.


Assuntos
Antineoplásicos/uso terapêutico , Araquidonato 15-Lipoxigenase/metabolismo , Hidroxiquinolinas/uso terapêutico , Inibidores de Lipoxigenase/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Quinolonas/uso terapêutico , Animais , Antineoplásicos/química , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Hidroxiquinolinas/química , Inibidores de Lipoxigenase/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias da Próstata/patologia , Quinolonas/química , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Eur J Pharmacol ; 910: 174505, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34534532

RESUMO

B-cell non-Hodgkin lymphoma (NHL) is among the ten most common malignancies. Survival rates range from very poor to over 90% and highly depend on the stage and subtype. Characteristic features of NHL are recurrent translocations juxtaposing an oncogene (e.g. MYC, BCL2) to the enhancers in the immunoglobulin heavy chain (IGH) locus. Survival and proliferation of many B-cell lymphomas depend on the expression of the translocated oncogene. Thus, targeting IGH enhancers as an anti-lymphoma treatment seems a promising strategy. Recently, a small molecule - 7-[[(4-methyl-2-pyridinyl)amino](2-pyridinyl)methyl]-8-quinolinol (compound 30666) was identified to decrease activity of the Eµ enhancer and reduce the expression of translocated oncogenes in multiple myeloma and some NHL cell lines (Dolloff, 2019). Here, we aimed to test the effect of compound 30666 in Burkitt lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL) and shed light on its mechanism of action. We report that both IGH-translocation positive NHL cells as well as IGH-translocation negative B cells and non-B cell controls treated with compound 30666 exhibited consistent growth inhibition. A statistically significant increase in cell percentage in sub-G1 phase of cell cycle was observed, suggesting induction of apoptosis. Compound 30666 downregulated MYC levels in BL cell lines and altered IGH enhancer RNA expression. Moreover, a global decrease of H3K27ac and an increase of H3K4me1 was observed upon 30666 treatment, which suggests switching enhancers to a poised or primed state. Altogether, our findings indicate that 30666 inhibitor affects enhancer activity but might not be as specific for IGH enhancers as previously reported.


Assuntos
Linfoma de Burkitt/tratamento farmacológico , Elementos Facilitadores Genéticos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hidroxiquinolinas/farmacologia , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Piridinas/farmacologia , Translocação Genética/efeitos dos fármacos , Linfoma de Burkitt/genética , Linfoma de Burkitt/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ensaios de Seleção de Medicamentos Antitumorais , Código das Histonas/efeitos dos fármacos , Humanos , Hidroxiquinolinas/uso terapêutico , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Piridinas/uso terapêutico
4.
Cancer Res ; 81(7): 1883-1895, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33531374

RESUMO

GRP78 (glucose-regulated protein, 78 kDa) is a key regulator of endoplasmic reticulum (ER) stress signaling. Cancer cells are highly proliferative and have high demand for protein synthesis and folding, which results in significant stress on the ER. To respond to ER stress and maintain cellular homeostasis, cells activate the unfolded protein response (UPR) that promotes either survival or apoptotic death. Cancer cells utilize the UPR to promote survival and growth. In this study, we describe the discovery of a series of novel hydroxyquinoline GRP78 inhibitors. A representative analogue, YUM70, inhibited pancreatic cancer cell growth in vitro and showed in vivo efficacy in a pancreatic cancer xenograft model with no toxicity to normal tissues. YUM70 directly bound GRP78 and inactivated its function, resulting in ER stress-mediated apoptosis. A YUM70 analogue conjugated with BODIPY showed colocalization of the compound with GRP78 in the ER. Moreover, a YUM70-PROTAC (proteolysis targeting chimera) was synthesized to force degradation of GRP78 in pancreatic cancer cells. YUM70 showed a strong synergistic cytotoxicity with topotecan and vorinostat. Together, our study demonstrates that YUM70 is a novel inducer of ER stress, with preclinical efficacy as a monotherapy or in combination with topoisomerase and HDAC inhibitors in pancreatic cancer. SIGNIFICANCE: This study identifies a novel ER stress inducer that binds GRP78 and inhibits pancreatic cancer cell growth in vitro and in vivo, demonstrating its potential as a therapeutic agent for pancreatic cancer.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Choque Térmico/antagonistas & inibidores , Hidroxiquinolinas/farmacologia , Neoplasias Pancreáticas/patologia , Células A549 , Animais , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Feminino , Células HCT116 , Células HT29 , Humanos , Hidroxiquinolinas/uso terapêutico , Células MCF-7 , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
5.
Proc Natl Acad Sci U S A ; 117(11): 6103-6113, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123069

RESUMO

Clinical observation of the association between cancer aggressiveness and embryonic development stage implies the importance of developmental signals in cancer initiation and therapeutic resistance. However, the dynamic gene expression during organogenesis and the master oncofetal drivers are still unclear, which impeded the efficient elimination of poor prognostic tumors, including human hepatocellular carcinoma (HCC). In this study, human embryonic stem cells were induced to differentiate into adult hepatocytes along hepatic lineages to mimic liver development in vitro. Combining transcriptomic data from liver cancer patients with the hepatocyte differentiation model, the active genes derived from different hepatic developmental stages and the tumor tissues were selected. Bioinformatic analysis followed by experimental assays was used to validate the tumor subtype-specific oncofetal signatures and potential therapeutic values. Hierarchical clustering analysis revealed the existence of two subtypes of liver cancer with different oncofetal properties. The gene signatures and their clinical significance were further validated in an independent clinical cohort and The Cancer Genome Atlas database. Upstream activator analysis and functional screening further identified E2F1 and SMAD3 as master transcriptional regulators. Small-molecule inhibitors specifically targeting the oncofetal drivers extensively down-regulated subtype-specific developmental signaling and inhibited tumorigenicity. Liver cancer cells and primary HCC tumors with different oncofetal properties also showed selective vulnerability to their specific inhibitors. Further precise targeting of the tumor initiating steps and driving events according to subtype-specific biomarkers might eliminate tumor progression and provide novel therapeutic strategy.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Hepatócitos/patologia , Neoplasias Hepáticas/genética , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/cirurgia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Estudos de Coortes , Intervalo Livre de Doença , Fator de Transcrição E2F1/antagonistas & inibidores , Fator de Transcrição E2F1/metabolismo , Feminino , Perfilação da Expressão Gênica , Hepatectomia , Células-Tronco Embrionárias Humanas , Humanos , Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Isoquinolinas/farmacologia , Isoquinolinas/uso terapêutico , Estimativa de Kaplan-Meier , Fígado/crescimento & desenvolvimento , Fígado/patologia , Fígado/cirurgia , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/cirurgia , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico , Piridinas/farmacologia , Piridinas/uso terapêutico , Pirróis/farmacologia , Pirróis/uso terapêutico , Transdução de Sinais/genética , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Invest Dermatol ; 140(6): 1295-1299.e1, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31705876
7.
Curr Drug Discov Technol ; 17(2): 203-212, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30306874

RESUMO

BACKGROUND: According to WHO, in 2017, about 90.5 million people suffered from cancer and about 8.8 million deaths occurred due to disease. Although the chemotherapeutic agents have decreased the mortality among the cancer patients but high toxicity and non-specific targets are still major drawbacks. Many researchers have identified linomide, a 4-hydroxy-2-quinolone derivative, as a lead molecule for the development of anticancer agents. With this background, we thought of the following objective. OBJECTIVE: The objective of this research work involves the synthesis of a series of N-(2-(4- hydroxy-2-oxo-1-phenyl-1,2-dihydroquinolin-3-yl)-2-oxoethyl)-N-alkyl substituted benzene sulfonamides IVa-d (1-3) by replacing the anilide moiety at the third position of linomide with sulfamoylacyl and also N-methyl by N-phenyl functionality. To perform in silico anticancer activity by using Molegro Virtual Docker (MVD-2013, 6.0) software and in vitro anticancer activity by MTT assay. METHODS: The starting material 4-hydroxy-1-phenylquinolin-2(1H)-one was treated with N-bromosuccinamide to yield compound II. Condensation of compound II with primary amines resulted in compounds IIIa-d, which, on coupling with substituted aromatic sulfonyl chlorides yield the title compounds IVa-d (1-3). RESULTS: All the synthesized compounds were satisfactorily characterized by spectral data. The results of docking revealed that the synthesized compounds exhibited well-conserved hydrogen bonds with one or more amino acid residues in the active pocket of EGFRK tyrosine kinase domain (PDB ID: 1m17). The MolDock Score of compound IVd-1 (-115.503) was the highest amongst those tested. The in vitro anticancer activity results showed that compound IVc-1 (R= - (CH2) 2-CH3 ; R'= -H) and IV d-1 (R= -CH2-C6H5; R'= -H) were found to be most potent against K562 cell line with an IC50 of 0.451 µM/ml and 0.455 µM/ml respectively. Compound IVd-1 also showed better potency against A549 cell line with IC50 value of 0.704 µM/ml. CONCLUSION: The results of in silico and in vitro anticancer activity are in agreement with each other. Compound IV d-1 was found to be most active of the series.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Hidroxiquinolinas/farmacologia , Neoplasias/tratamento farmacológico , Células A549 , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Domínio Catalítico/efeitos dos fármacos , Proliferação de Células , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Receptores ErbB/ultraestrutura , Humanos , Ligação de Hidrogênio , Hidroxiquinolinas/química , Hidroxiquinolinas/uso terapêutico , Concentração Inibidora 50 , Células K562 , Simulação de Acoplamento Molecular , Relação Estrutura-Atividade
8.
J Neuroinflammation ; 16(1): 83, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-30975172

RESUMO

BACKGROUND: Neuropathic pain is a serious clinical problem that needs to be solved urgently. ASK1 is an upstream protein of p38 and JNK which plays important roles in neuroinflammation during the induction and maintenance of chronic pain. Therefore, inhibition of ASK1 may be a novel therapeutic approach for neuropathic pain. Here, we aim to investigate the effects of paeoniflorin on ASK1 and neuropathic pain. METHODS: The mechanical and thermal thresholds of rats were measured using the Von Frey test. Cell signaling was assayed using western blotting and immunohistochemistry. RESULTS: Chronic constrictive injury (CCI) surgery successfully decreased the mechanical and thermal thresholds of rats and decreased the phosphorylation of ASK1 in the rat spinal cord. ASK1 inhibitor NQDI1 attenuated neuropathic pain and decreased the expression of p-p38 and p-JNK. Paeoniflorin mimicked ASK1 inhibitor NQDI1 and inhibited ASK1 phosphorylation. Paeoniflorin decreased the expression of p-p38 and p-JNK, delayed the progress of neuropathic pain, and attenuated neuropathic pain. Paeoniflorin reduced the response of astrocytes and microglia to injury, decreased the expression of IL-1ß and TNF-α, and downregulated the expression of CGRP induced by CCI. CONCLUSIONS: Paeoniflorin is an effective drug for the treatment of neuropathic pain in rats via inhibiting the phosphorylation of ASK1, suggesting it may be effective in patients with neuropathic pain.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Apoptose/efeitos dos fármacos , Encefalite/tratamento farmacológico , Glucosídeos/uso terapêutico , MAP Quinase Quinase Quinase 5/metabolismo , Monoterpenos/uso terapêutico , Neuropatia Ciática/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Encefalite/complicações , Hidroxiquinolinas/uso terapêutico , Hiperalgesia/fisiopatologia , Interleucina-1beta/metabolismo , Masculino , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/complicações
9.
BMC Cancer ; 19(1): 149, 2019 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760223

RESUMO

BACKGROUND: Chemotherapy-induced alopecia has been well documented as a cause of distress to patients undergoing cancer treatment. Almost all traditional chemotherapeutic agents cause severe alopecia. Despite advances in the treatment of chemotherapy-induced alopecia, there is no effective treatment for preventing chemotherapy-induced alopecia. METHODS: In the present study, we investigated the potential role of a multi-target iron chelator, M30 in protecting against cyclophosphamide-induced alopecia in C57BL/6 mice implanted with an osmotic pump. M30 enhanced hair growth and prevented cyclophosphamide-induced abnormal hair in the mice. Furthermore, we examined the gene expression profiles derived from skin biopsy specimens of normal mice, cyclophosphamide-treated mice, and cyclophosphamide treated mice with M30 supplement. RESULTS: The top genes namely Tnfrsf19, Ercc2, Lama5, Ctsl, and Per1 were identified by microarray analysis. These genes were found to be involved in the biological processes of hair cycle, hair cycle phase, hair cycle process, hair follicle development, hair follicle maturation, hair follicle morphogenesis, regulation of hair cycle. CONCLUSION: Our study demonstrates that M30 treatment is a promising therapy for cyclophosphamide-induced alopecia and suggests that the top five genes have unique preventive effects in cyclophosphamide-induced transformation.


Assuntos
Alopecia/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Antioxidantes/uso terapêutico , Ciclofosfamida/efeitos adversos , Hidroxiquinolinas/uso terapêutico , Quimioterapia de Indução/efeitos adversos , Neoplasias/tratamento farmacológico , Alopecia/induzido quimicamente , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclofosfamida/uso terapêutico , Modelos Animais de Doenças , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Receptores do Fator de Necrose Tumoral/genética
10.
Invest Ophthalmol Vis Sci ; 58(12): 5287-5297, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29049732

RESUMO

Purpose: Retinitis pigmentosa (RP) is a group of hereditary retinal degeneration in which mutations commonly result in the initial phase of rod cell death followed by gradual cone cell death. The mechanisms by which the mutations lead to photoreceptor cell death in RP have not been clearly elucidated. There is currently no effective treatment for RP. The purpose of this work was to explore iron chelation therapy for improving cone survival and function in the rd10 mouse model of RP. Methods: Two iron-chelating drugs, 5-(4-(2-hydroxyethyl) piperazin-1-yl (methyl)-8-hydroxyquinoline (VK28) and its chimeric derivative 5-(N-methyl-N-propargyaminomethyl)-quinoline-8-oldihydrochloride (VAR10303), were injected intraperitoneally to rd10 mice every other day starting from postnatal day 14. We investigate the effects of the two compounds on cone rescue at three time points, using a combination of immunocytochemistry, RT-PCR, Western blot analysis, and a series of visual function tests. Results: VK28 and VAR10303 treatments partially rescued cones, and significantly improved visual function in rd10 mice. Moreover, we showed that the neuroprotective effects of VK28 and VAR10303 were correlated to inhibition of neuroinflammation, oxidative stress, and apoptosis. Furthermore, we demonstrated that downregulation of NF-kB and p53 is likely to be the mechanisms by which proinflammatory mediators and apoptosis are reduced in the rd10 retina, respectively. Conclusions: VK28 and VAR10303 provided partial histologic and functional rescue of cones in RD10 mice. Our study demonstrated that iron chelation therapy might represent an effective therapeutic strategy for RP patients.


Assuntos
Modelos Animais de Doenças , Quelantes de Ferro/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Células Fotorreceptoras Retinianas Cones/fisiologia , Retinose Pigmentar/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/fisiologia , Eletrorretinografia , Hidroxiquinolinas/uso terapêutico , Imuno-Histoquímica , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Piperazinas/uso terapêutico , Quinolinas/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Proteína Supressora de Tumor p53/metabolismo
11.
Neurotox Res ; 31(2): 230-244, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27826939

RESUMO

The aim of the present study was to evaluate the therapeutic effect of the novel neuroprotective multitarget brain permeable monoamine oxidase inhibitor/iron chelating-radical scavenging drug, VAR10303 (VAR), co-administered with high-calorie/energy-supplemented diet (ced) in SOD1G93A transgenic amyotrophic lateral sclerosis (ALS) mice. Administration of VAR-ced was initiated after the appearance of disease symptoms (at day 88), as this regimen is comparable with the earliest time at which drug therapy could start in ALS patients. Using this rescue protocol, we demonstrated in the current study that VAR-ced treatment provided several beneficial effects in SOD1G93A mice, including improvement in motor performance, elevation of survival time, and attenuation of iron accumulation and motoneuron loss in the spinal cord. Moreover, VAR-ced treatment attenuated neuromuscular junction denervation and exerted a significant preservation of myofibril regular morphology, associated with a reduction in the expression levels of genes related to denervation and atrophy in the gastrocnemius (GNS) muscle in SOD1G93A mice. These effects were accompanied by upregulation of mitochondrial DNA and elevated activities of complexes I and II in the GNS muscle. We have also demonstrated that VAR-ced treatment upregulated the mitochondrial biogenesis master regulator, peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) and increased PGC-1α-targeted metabolic genes and proteins, such as, PPARγ, UCP1/3, NRF1/2, Tfam, and ERRα in GNS muscle. These results provide evidence of therapeutic potential of VAR-ced in SOD1G93A mice with underlying molecular mechanisms, further supporting the importance role of multitarget iron chelators in ALS treatment.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , DNA Mitocondrial/metabolismo , Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Destreza Motora/efeitos dos fármacos , Taxa de Sobrevida , Esclerose Lateral Amiotrófica/dietoterapia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Terapia Combinada , Denervação , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Ferro/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios Motores/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miofibrilas/efeitos dos fármacos , Junção Neuromuscular/patologia , Estresse Oxidativo/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Medula Espinal/metabolismo , Medula Espinal/fisiologia , Superóxido Dismutase-1/genética , Regulação para Cima/efeitos dos fármacos
12.
Biomed Pharmacother ; 80: 145-150, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27133051

RESUMO

Tumour growth is closely related to the development of new blood vessels to supply oxygen and nutrients to cancer cells. Without the neovascular formation, tumour volumes cannot increase and undergo metastasis. Antiangiogenesis is one of the most promising approaches for antitumour therapy. The exploration of new antiangiogenic agents would be helpful in antitumour therapy. Quinoline is an aromatic nitrogen compound characterized by a double-ring structure which exhibits a benzene ring fused to pyridine at two adjacent carbon atoms. The high stability of quinoline makes it preferable in a variety of therapeutic and pharmaceutical applications, including antitumour treatment. This work is to examine the potential antiangiogenic activity of the synthetic compound 2-Formyl-8-hydroxy-quinolinium chloride. We found that 2-Formyl-8-hydroxy-quinolinium chloride could inhibit the growth of human umbilical vein endothelial cells in vitro. Using the diethylnitrosamine-induced hepatocarcinogenesis model, 2-Formyl-8-hydroxy-quinolinium chloride showed strong antiangiogenic activity. Furthermore, 2-Formyl-8-hydroxy-quinolinium chloride could inhibit the growth of large Hep3B xenografted tumour from the nude mice. We assume that 2-Formyl-8-hydroxy-quinolinium chloride could be a potential antiangiogenic and antitumour agent and it is worthwhile to further study its underlying working mechanism.


Assuntos
Inibidores da Angiogênese/farmacologia , Hidroxiquinolinas/farmacologia , Compostos de Quinolínio/farmacologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinogênese/patologia , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dietilnitrosamina , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Hidroxiquinolinas/química , Hidroxiquinolinas/uso terapêutico , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Camundongos Endogâmicos C57BL , Camundongos Nus , Compostos de Quinolínio/química , Compostos de Quinolínio/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Life Sci ; 136: 108-19, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26159898

RESUMO

AIM: Novel effective treatment is urgently needed for sporadic Alzheimer's disease (sAD). M30 ([5-(N-methyl-N-propargylaminomethyl)-8-hydroxyquinoline]) and HLA-20 (5-{4-propargylpiperazin-1-ylmethyl}-8-hydroxyquinoline) are brain permeable, iron chelating compounds with antioxidant activity, showing also neuroprotective activity in animal models of neurodegeneration.Weaimed to explore their therapeutic potential in non-transgenic (non-Tg) rat model of sAD developed by intracerebroventricular administration of streptozotocin (STZ-icv). MAIN METHODS: Therapeutic effects of chronic oral M30 (2 and 10 mg/kg) and HLA20 (5 and 10 mg/kg) treatment on cognitive impairment in STZ-icv rat model were explored by Morris Water Maze (MWM) and Passive Avoidance (PA) tests in neuropreventive and neurorescue paradigms. Data were analysed by Kruskal­Wallis and Mann­Whitney U test (p b 0.05). KEY FINDINGS: Five-day oral pre-treatment with M30 and HLA20 dose-dependently prevented development of spatial memory impairment (MWM probe trial-time +116%/M30; +60%/HLA20) in STZ-icv rat model (p b 0.05). Eleven-week oral treatment with M30 (3×/week), initiated 8 days after STZ-icv administration dosedependently ameliorated already developed cognitive deficits in MWM test (reduced number of mistakes 3 months after the STZ-icv treatment ­ 59%; p b 0.05) and fully restored them in PA test (+314%; p b 0.05). Chronic M30 treatment fully restored (−47%/PHF1;−65%/AT8; p b 0.05) STZ-induced hyperphosphorylation of tau protein and normalized decreased expression of insulin degrading enzyme (+37%; p b 0.05) in hippocampus. SIGNIFICANCE: The results provide first evidence of therapeutic potential of M30 and HLA20 in STZ-icv rat model of sAD with underlying molecular mechanism, further supporting the important role of multi-target ironchelators in sAD treatment.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Hidroxiquinolinas/farmacologia , Quelantes de Ferro/farmacologia , Transtornos da Memória/tratamento farmacológico , Piperazinas/farmacologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/psicologia , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hidroxiquinolinas/uso terapêutico , Quelantes de Ferro/uso terapêutico , Masculino , Memória de Longo Prazo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Piperazinas/uso terapêutico , Ratos Wistar , Estreptozocina
14.
Neurobiol Aging ; 36(3): 1529-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25499799

RESUMO

Recently, we have designed and synthesized a novel multipotent, brain-permeable iron-chelating drug, VAR10303 (VAR), possessing both propargyl and monoamine oxidase (MAO) inhibitory moieties. The present study was undertaken to determine the multiple pharmacological activities of VAR in neurodegenerative preclinical models. We demonstrate that VAR affords iron chelating/iron-induced lipid-peroxidation inhibitory potency and brain selective MAO-A and MAO-B inhibitory effects, with only limited tyramine-cardiovascular potentiation of blood pressure. The results show that in 6-hydroxydopamine rat (neuroprotection) and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse (neurorescue) Parkinson's disease models, VAR significantly attenuated the loss of striatal dopamine levels, markedly reduced dopamine turnover, and increased tyrosine-hydroxylase levels. Furthermore, chronic systemic treatment of aged rats with VAR improved cognitive behavior deficits and enhanced the expression levels of neurotrophic factors (e.g., brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and nerve growth factor), Bcl-2 family members and synaptic plasticity in the hippocampus. Our study indicates that the multitarget compound VAR exerted neuroprotective and neurorestorative effects in animal models of Parkinson's disease and aging, further suggesting that a drug that can regulate multiple brain targets could be an ideal treatment-strategy for age-associated neurodegenerative disorders.


Assuntos
Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Quelantes de Ferro/farmacologia , Quelantes de Ferro/uso terapêutico , Inibidores da Monoaminoxidase/farmacologia , Inibidores da Monoaminoxidase/uso terapêutico , Fármacos Neuroprotetores , Doença de Parkinson/tratamento farmacológico , Envelhecimento , Animais , Cognição , Modelos Animais de Doenças , Dopamina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Monoaminoxidase , Fatores de Crescimento Neural/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/psicologia , Ratos Sprague-Dawley
15.
Chin J Nat Med ; 12(3): 222-4, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24702810

RESUMO

AIM: To investigate the quinoline alkaloids from the roots of Dictamnus angustifolius G.Don ex Sweet (Rutaceae). METHOD: The quinoline alkaloids were isolated by various column chromatographic methods and their structures were elucidated on the basis of spectral analysis. RESULTS: A new quinoline alkaloid, 5-methoxylrobustine (1), along with five known quinoline alkaloids were obtained, and their structures were identified as dictamnine (2), robustine (3), isopteleine (4), γ-fagarine (5), and skimmianine (6). Cytotoxicity testing of these alkaloids showed that all of them had weak cytotoxic activities against human breast cancer cells (MCF7). CONCLUSION: Compound 1 is a new quinoline alkaloid. Alkaloid 3 showed stronger anti-proliferation effect than the other alkaloids.


Assuntos
Antineoplásicos Fitogênicos/isolamento & purificação , Neoplasias da Mama/tratamento farmacológico , Dictamnus/química , Hidroxiquinolinas/isolamento & purificação , Fitoterapia , Extratos Vegetais/química , Raízes de Plantas/química , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Humanos , Hidroxiquinolinas/química , Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Estrutura Molecular , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Quinolinas/química , Quinolinas/isolamento & purificação , Quinolinas/farmacologia , Quinolinas/uso terapêutico
16.
Hematol Oncol Clin North Am ; 28(2): 323-39, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24589269

RESUMO

P-selectin on endothelial cell surfaces is central to impaired microvascular blood flow in sickle cell disease (SCD). Restoration of blood flow is expected to provide therapeutic benefit for SCD patients, whatever the mechanism of action of the treatment. Long-term oral administration of a P-selectin-blocking agent potentially improves blood flow and averts acute painful vaso-occlusive crises in patients with SCD. This review focuses on the pathophysiology of the impairment of microvascular blood flow in SCD with an emphasis on the role of P-selectin and summarizes the status of development of antiselectin therapies as a means of improving microvascular flow.


Assuntos
Anemia Falciforme/fisiopatologia , Endotélio Vascular/fisiopatologia , Selectina-P/fisiologia , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/metabolismo , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Glicolipídeos/farmacologia , Glicolipídeos/uso terapêutico , Humanos , Hidroxiquinolinas/farmacologia , Hidroxiquinolinas/uso terapêutico , Selectina-P/antagonistas & inibidores , Selectina-P/metabolismo , Poliéster Sulfúrico de Pentosana/farmacologia , Poliéster Sulfúrico de Pentosana/uso terapêutico
17.
Mol Neurobiol ; 46(1): 217-20, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22847630

RESUMO

Based on a multimodal drug design strategy for age-related neurodegenerative diseases, we have synthesized a multifunctional nontoxic, brain-permeable iron-chelating compound, M30, possessing the neuroprotective N-propargyl moiety of the anti-Parkinsonian drug, monoamine oxidase-B inhibitor, rasagiline and the antioxidant-iron chelator moiety of the 8-hydroxyquinoline derivative of the iron chelator, VK28. In the present short overview, we describe the neuroprotective and the neurorestorative activity of M30, acting against multiple brain targets, including regulation on amyloid ß, neurogenesis, and activation of hypoxia inducible factor signaling pathways. The diverse pharmacological properties and several pathological targets of M30 make this drug potential valuable for therapeutic strategy of Alzheimer's-like neuropathology and aging.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/patologia , Hidroxiquinolinas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Envelhecimento/efeitos dos fármacos , Amiloidose/tratamento farmacológico , Amiloidose/patologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Hidroxiquinolinas/química , Hidroxiquinolinas/farmacologia , Camundongos , Fármacos Neuroprotetores/farmacologia
18.
Chem Biol Drug Des ; 80(2): 291-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22553933

RESUMO

3-Aryl-2-quinolone derivates were extensively investigated for their inhibition of farnesyl transferase. Taking this as a cue, we studied the other possible mechanism of antitumor activity of 2-quinolone derivates. A series of new 2-quinolone derivatives have been synthesized and screened for their cytotoxicity by trypan blue assay on Ehrlich ascites carcinoma cells and MTT assay on MCF-7 cells. Compound 1a (nJST) was found to be more effective in both studies with the lowest CTC(50) value among all nine synthesized compounds. This compound was further screened on four different cell lines, viz. human breast adenocarcinoma (MCF-7, MDA-MB-231), colon cancer (HCT-15), murine melanoma (B16F10) cell lines for 24 and 48 h. The CTC(50) value of the compound was found to be <10 µm. Compound 1a induced DNA damage which was revealed by DNA fragmentation studies and further confirmed by nuclear staining. The compound also showed significant elevation in Bax and reduction Bcl-2 gene expression levels. Acute toxicity study in mice indicated that the compound is safe till 2000 mg/kg. Two different doses 50 and 100 mg/kg were selected and studied in Ehrlich ascites carcinoma model of cancer and have shown significant improvement in survival time and hematological parameters.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Hidroxiquinolinas/química , Hidroxiquinolinas/uso terapêutico , Neoplasias/tratamento farmacológico , Quinolonas/química , Quinolonas/uso terapêutico , Proteína X Associada a bcl-2/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma de Ehrlich/tratamento farmacológico , Carcinoma de Ehrlich/genética , Carcinoma de Ehrlich/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiquinolinas/farmacologia , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Quinolonas/farmacologia , Proteína X Associada a bcl-2/genética
19.
J Alzheimers Dis ; 30(1): 1-16, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22387411

RESUMO

Alzheimer's disease (AD) is a multifactorial syndrome involving a complex array of different, while related, factors in its progression. Accordingly, novel approaches that can simultaneously modulate several disease-related targets hold great promise for the effective treatment of AD. This review describes the development of novel hybrid molecules with multimodal activity, including: i) M30, the brain permeable selective monoamine oxidase (MAO)-A and -B inhibitor with chelating and neuroprotective activity; ii) HLA20, a brain permeable metal chelator with neuroprotective activity; iii) HLA20A, an acetylcholinesterase (AChE) inhibitor with site-activated chelating and neuroprotective activity; iv) M30D, an AChE and MAO-A and -B inhibitor with site-activated chelating and neuroprotective activity; and v) analogs of the neuroprotective aminoacid peptide, NAPVSIPQ. HLA20A and M30D act as pro-chelators and can be activated to liberate their respective active chelators HLA20 and M30 through pseudo inhibition of AChE. We first discuss the knowledge and structure-based strategy for the rational design of these novel compounds. Then, we review our recent studies on these drug candidates, regarding their wide range in vitro and in vivo activities, with emphasis on antioxidant-chelating potency and AchE and MAO-A and -B inhibitory activity, as well as neuroprotective/neurorescue effects. Finally, we discuss the diverse molecular mechanisms of action of these compounds with relevance to AD, including modulation of amyloid-ß and amyloid-ß protein precursor expression/processing; induction of cell cycle arrest; inhibition of neuronal death markers; and upregulation of neurotrophic factors, as well as activation of protein kinase signaling pathways.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Inibidores da Colinesterase/uso terapêutico , Indanos/uso terapêutico , Inibidores da Monoaminoxidase/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Quelantes/uso terapêutico , Inibidores da Colinesterase/farmacologia , Humanos , Hidroxiquinolinas/uso terapêutico , Indanos/farmacologia , Inibidores da Monoaminoxidase/farmacologia , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos/uso terapêutico , Piperazinas/uso terapêutico
20.
Antioxid Redox Signal ; 17(6): 860-77, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22360429

RESUMO

AIMS: The aim of the present study was to evaluate the therapeutic effect of the novel neuroprotective multi-target nontoxic, lipophilic, brain permeable monoamine oxidase inhibitor and iron chelating-radical scavenging drug, M30, on the neuropathology and deficits of spatial learning and memory in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (Tg) Alzheimer's disease (AD) mice. RESULTS: Here, we report that systemic treatment of APP/PS1 Tg mice with M30 for 9 months, significantly attenuated cognitive impairments in a variety of tasks of spatial learning and memory retention, working memory, learning abilities, anxiety levels, and memory for novel food and nesting behavior. Furthermore, we found that M30 reduced cerebral iron accumulation accompanied by a marked decrease in several AD-like phenotypes, including cerebral APP levels, amyloid ß (Aß) levels and plaques, phospho-APP and phospho-tau. Signaling studies revealed that M30 markedly downregulated the levels of phosphorylated cyclin-dependent kinase 5 and increased protein kinase B and glycogen synthase kinase 3ß phosphorylation. INNOVATION: Accumulation and deposition of brain iron is central to various neuropathological processes in AD, including oxidative stress, amyloid deposition, and tau phosphorylation. Thus, the concept of iron chelation holds considerable promise as a therapeutic strategy for AD pathogenesis. Here, for the first time, we demonstrated that, when systemically administered to APP/PS1 Tg mice, our novel multifunctional iron chelating/radical scavenging compound, M30, effectively reduced Aß accumulation and tau phosphorylation, and attenuated memory deficits. CONCLUSIONS: These findings suggest that M30 is a potential therapeutic agent for the prevention and treatment of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Hidroxiquinolinas/uso terapêutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Comportamento Animal , Western Blotting , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fosforilação , Presenilina-1/genética , Presenilina-1/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA