Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(49): 31177-31188, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33219123

RESUMO

A transplanted stem cell's engagement with a pathologic niche is the first step in its restoring homeostasis to that site. Inflammatory chemokines are constitutively produced in such a niche; their binding to receptors on the stem cell helps direct that cell's "pathotropism." Neural stem cells (NSCs), which express CXCR4, migrate to sites of CNS injury or degeneration in part because astrocytes and vasculature produce the inflammatory chemokine CXCL12. Binding of CXCL12 to CXCR4 (a G protein-coupled receptor, GPCR) triggers repair processes within the NSC. Although a tool directing NSCs to where needed has been long-sought, one would not inject this chemokine in vivo because undesirable inflammation also follows CXCL12-CXCR4 coupling. Alternatively, we chemically "mutated" CXCL12, creating a CXCR4 agonist that contained a strong pure binding motif linked to a signaling motif devoid of sequences responsible for synthetic functions. This synthetic dual-moity CXCR4 agonist not only elicited more extensive and persistent human NSC migration and distribution than did native CXCL 12, but induced no host inflammation (or other adverse effects); rather, there was predominantly reparative gene expression. When co-administered with transplanted human induced pluripotent stem cell-derived hNSCs in a mouse model of a prototypical neurodegenerative disease, the agonist enhanced migration, dissemination, and integration of donor-derived cells into the diseased cerebral cortex (including as electrophysiologically-active cortical neurons) where their secreted cross-corrective enzyme mediated a therapeutic impact unachieved by cells alone. Such a "designer" cytokine receptor-agonist peptide illustrates that treatments can be controlled and optimized by exploiting fundamental stem cell properties (e.g., "inflammo-attraction").


Assuntos
Quimiocina CXCL12/genética , Neurônios/metabolismo , Ligação Proteica/genética , Receptores CXCR4/genética , Astrócitos/metabolismo , Astrócitos/patologia , Movimento Celular/genética , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Humanos , Células-Tronco Pluripotentes Induzidas , Inflamação/genética , Ligantes , Mutagênese/genética , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Neurônios/patologia
3.
Cell Death Dis ; 13(12): 1022, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36473850

RESUMO

Dysregulated gene expression programs and redox and metabolic adaptations allow cancer cells to survive under high oxidative burden. These mechanisms also represent therapeutic vulnerabilities. Using triple-negative breast cancer (TNBC) as a model, we show that compared to normal human breast epithelial cells, the TNBC cells, MDA-MB-231 and MDA-MB-468 that harbor constitutively active STAT3 also express higher glucose-6-phosphate dehydrogenase (G6PD), thioredoxin reductase (TrxR)1, NADPH, and GSH levels for survival. Present studies discover that the natural product, R001, targets these adaptation mechanisms. Treatment of TNBC cells with R001 inhibited constitutively active STAT3, STAT3-regulated gene expression, and the functions of G6PD and TrxR1. Consequently, in the TNBC, but not normal cells, R001 suppressed GSH levels, but raised NADPH levels, reflective of a loss of mitochondrial respiration and which led to reactive oxygen species (ROS) induction, all of which led to loss of viable cells and inhibition of anchorage-dependent and independent growth. R001 treatment further led to early pyroptosis and late DNA damage, cell cycle arrest, and apoptosis only in the TNBC cells. Oral administration of 5 mg/kg R001 inhibited MDA-MB-468 xenografts growth in mice, with reduced pY705-STAT3, G6PD, TrxR1, and GSH levels. R001 serves as a therapeutic entity that targets the vulnerabilities of TNBC cells to inhibit tumor growth in vivo.


Assuntos
Produtos Biológicos , Neoplasias da Mama , Humanos , Camundongos , Animais , Feminino , NADP , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Fator de Transcrição STAT3
4.
Cancer Lett ; 534: 215613, 2022 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-35276290

RESUMO

Signal transducer and activator of transcription (Stat)3 is a valid anticancer therapeutic target. We have discovered a highly potent chemotype that amplifies the Stat3-inhibitory activity of lead compounds to levels previously unseen. The azetidine-based compounds, including H172 (9f) and H182, irreversibly bind to Stat3 and selectively inhibit Stat3 activity (IC50 0.38-0.98 µM) over Stat1 or Stat5 (IC50 > 15.8 µM) in vitro. Mass spectrometry detected the Stat3 cysteine peptides covalently bound to the azetidine compounds, and the key residues, Cys426 and Cys468, essential for the high potency inhibition, were confirmed by site-directed mutagenesis. In triple-negative breast cancer (TNBC) models, treatment with the azetidine compounds inhibited constitutive and ligand-induced Stat3 signaling, and induced loss of viable cells and tumor cell death, compared to no effect on the induction of Janus kinase (JAK)2, Src, epidermal growth factor receptor (EGFR), and other proteins, or weak effects on cells that do not harbor aberrantly-active Stat3. H120 (8e) and H182 as a single agent inhibited growth of TNBC xenografts, and H278 (hydrochloric acid salt of H182) in combination with radiation completely blocked mouse TNBC growth and improved survival in syngeneic models. We identify potent azetidine-based, selective, irreversible Stat3 inhibitors that inhibit TNBC growth in vivo.


Assuntos
Azetidinas , Neoplasias de Mama Triplo Negativas , Animais , Apoptose , Azetidinas/farmacologia , Linhagem Celular Tumoral , Humanos , Camundongos , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
5.
J Med Chem ; 64(1): 695-710, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33352047

RESUMO

We optimized our previously reported proline-based STAT3 inhibitors into an exciting new series of (R)-azetidine-2-carboxamide analogues that have sub-micromolar potencies. 5a, 5o, and 8i have STAT3-inhibitory potencies (IC50) of 0.55, 0.38, and 0.34 µM, respectively, compared to potencies greater than 18 µM against STAT1 or STAT5 activity. Further modifications derived analogues, including 7e, 7f, 7g, and 9k, that addressed cell membrane permeability and other physicochemical issues. Isothermal titration calorimetry analysis confirmed high-affinity binding to STAT3, with KD of 880 nM (7g) and 960 nM (9k). 7g and 9k inhibited constitutive STAT3 phosphorylation and DNA-binding activity in human breast cancer, MDA-MB-231 or MDA-MB-468 cells. Furthermore, treatment of breast cancer cells with 7e, 7f, 7g, or 9k inhibited viable cells, with an EC50 of 0.9-1.9 µM, cell growth, and colony survival, and induced apoptosis while having relatively weaker effects on normal breast epithelial, MCF-10A or breast cancer, MCF-7 cells that do not harbor constitutively active STAT3.


Assuntos
Azetidinas/química , Fator de Transcrição STAT3/antagonistas & inibidores , Amidas/química , Apoptose/efeitos dos fármacos , Azetidinas/metabolismo , Azetidinas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/química , DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Concentração Inibidora 50 , Fosforilação/efeitos dos fármacos , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade
6.
Chem Biol Interact ; 308: 235-243, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31145889

RESUMO

mHA11, a 2-amino-4-phenyl-4H-chromene-3-carboxylate analog, is a microtubule-targeting agent discovered by our group through the modification of the Bcl-2 inhibitor HA14-1. mHA11 exhibits cytotoxicities against tumor cells with nM IC50 values, whereas it has only a minimal effect on normal cells. We explored the plasma pharmacokinetics, tissue distribution, and excretion of mHA11 in rats using a liquid chromatography/tandem mass spectrometry (LC-MS/MS) method. Next, we identified the metabolites of mHA11 and assessed the influence of cytochrome P450 (CYP) isozymes on mHA11 metabolism. We also examined the in vitro stability in rat plasma and rat liver microsomes (RLMs), the blood-to plasma (B/P) ratio, and the inhibitory effect on CYP isozyme activities. After oral administration at 5, 15, and 45 mg/kg, mHA11 was absorbed and eliminated rapidly. There was a linear correlation between the area under the concentration-time curve (AUC0-∞) and the dose (R2 = 0.983). The bioavailability of mHA11 was 4.1% at the oral dose of 15 mg/kg mHA11 was extensively distributed in various tissues and exhibited a high penetration into the brain. No significant parent drug was detected in urine or bile, and only 0.74% was recovered in feces, whereas two demethylated metabolites, M1 and M2, were found in the urine and feces, and further studies showed that CYP2C19 primarily contributed to metabolites formation. mHA11 was stable in rat plasma but degraded significantly in RLMs; its B/P ratio was 1.05 in rat blood. In addition, mHA11 dose-dependently inhibited the activities of rat CYP isozymes, including CYP1A2, CYP2C6, CYP2C11, CYP2D2, CYP2E1 and CYP3A2. The present study is the first report on the disposition of mHA11 in rats and provides important data for further research and development of this inhibitor.


Assuntos
Benzopiranos/farmacocinética , Moduladores de Tubulina/farmacocinética , Administração Oral , Animais , Benzopiranos/sangue , Benzopiranos/química , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Estabilidade de Medicamentos , Humanos , Masculino , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Espectrometria de Massas em Tandem , Distribuição Tecidual , Moduladores de Tubulina/sangue , Moduladores de Tubulina/química
7.
J Pharm Biomed Anal ; 161: 159-167, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30165332

RESUMO

DV1 is a potent and selective D-peptide antagonist of CXCR4 and being developed as a novel drug candidate molecule. For preclinical pharmacokinetic study of DV1, we established an efficient and reliable liquid chromatography coupled to tandem mass spectrometric (LC-MS/MS) method for the assay of DV1 in rat plasma. Plasma samples were acidified by formic acid and then their protein content precipitated by acetonitrile. Sample separation was processed with a C18 column (4.6 mm × 100 mm, 5 µm) and washed by a water-acetonitrile gradient mobile phase containing 0.1% (v/v) formic acid at a flow rate of 0.4 mL/min. The mass spectrometer was operated in the multiple reaction monitoring mode and positive electrospray ionization. The assay had a good linearity over the range of 10-10000 ng/mL (r>0.998) for DV1. The adsorption of the peptide was diminished by organic additives during the quantitative procedure. The intra- and inter-day precision was 1.9-9.8% and the accuracy was 91.2-110.0%. No significant variation was observed under the optimized conditions. The recovery was above 52% with low matrix effects. The method was successfully applied to a pharmacokinetic study of DV1 after subcutaneous injection at dose of 10 mg/kg in rats. The half-life and AUCinf of DV1 were calculated as 8.7 h and 35,553 ng/mL·h, respectively. It is the first report on the quantitative analysis and pharmacokinetic characterization of a D-peptide targeted CXCR4, which should be useful for further preclinical studies and development of this and other peptide therapeutics.


Assuntos
Peptídeos/sangue , Peptídeos/farmacocinética , Receptores CXCR4/antagonistas & inibidores , Espectrometria de Massas em Tandem/métodos , Animais , Cromatografia Líquida de Alta Pressão/métodos , Estabilidade de Medicamentos , Masculino , Ratos , Temperatura
8.
Chem Biol Interact ; 247: 55-63, 2016 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-26827781

RESUMO

DDPH (1-(2, 6-dimethylphenoxy)-2-(3, 4-dimethoxyphenylethylamino) propane hydrochloride) is a promising novel antihypertensive agent, with potent antihypertensive, neuroprotective and cardioprotective effects. This study aimed to investigate the effects of DDPH on the expression and activity of hepatic cytochrome P450 (CYP) isoforms and evaluate the metabolic drug-drug interactions of DDPH with propafenone. Our results showed that orally administered DDPH (12.5-50 mg/kg/d) for 7 days significantly inhibited CYP2D1 and CYP3A1 activity and mRNA and protein expression but weakly increased CYP1A2 activity and expression in rats. Enzyme kinetics studies showed that DDPH was a competitive inhibitor of CYP2D1 and mixed inhibitor of CYP3A1 in rat liver microsomes with Ki values of 3.70 ± 0.42 µM and 4.79 ± 1.10 µM respectively. With human liver microsomes, DDPH was a noncompetitive inhibitor of CYP2D6 (Ki = 0.85 ± 0.06 µM) and mixed inhibitor of CYP3A (Ki = 2.15 ± 0.41 µM). Further in vivo study showed that oral administration of DDPH (12.5-50 mg/kg/d) for 7 days in rats significantly increased the area under the plasma concentration-time curve (AUC) of propafenone by 25.4%-63.9%, with a concomitant decrease in the plasma clearance. In conclusion, the results indicated that DDPH inhibited CYP2D and CYP3A activities and down-regulated their protein expression and mRNA transcription. DDPH might cause metabolic drug-drug interactions through modulation of the activity and expression of CYP2D and CYP3A. This information could be important in the prediction of possible drug-drug interactions as well as for the effective therapy and the limitation of toxicity of DDPH in clinical practice.


Assuntos
Anti-Hipertensivos/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fenetilaminas/farmacologia , Animais , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Propafenona/farmacocinética , Ratos , Ratos Sprague-Dawley
9.
Sci Rep ; 5: 17730, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26635117

RESUMO

Gefitinib (Gef) provides clinical benefits to non-small cell lung cancer (NSCLC) patients with activating EGFR mutations. However, acquired resistance (AR) is a major obstacle to effective Gef therapy. This study demonstrated that resveratrol (Res) could synergize with Gef to inhibit the proliferation of Gef-resistant NSCLC cells. The underlying mechanisms of synergism were investigated, and the results showed that cotreatment with Gef and Res could inhibit EGFR phosphorylation by increasing intracellular Gef accumulation through the impairment of Gef elimination from PC9/G cells. Consistently, CYP1A1 and ABCG2 expression were inhibited. Meanwhile, the cotreatment significantly induced cell apoptosis, autophagy, cell cycle arrest and senescence accompanied by increased expression of cleaved caspase-3, LC3B-II, p53 and p21. Further studies revealed that autophagy inhibition enhanced apoptosis and abrogated senescence while apoptosis inhibition had no notable effect on cell autophagy and senescence during cotreatment with Gef and Res. These results indicated that in addition to apoptosis, senescence promoted by autophagy contributes to the antiproliferation effect of combined Gef and Res on PC9/G cells. In conclusion, combined treatment with Gef and Res may represent a rational strategy to overcome AR in NSCLC cells.


Assuntos
Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Estilbenos/administração & dosagem , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/biossíntese , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Neoplasias/biossíntese , Quinazolinas/administração & dosagem , Resveratrol
10.
J Chromatogr B Analyt Technol Biomed Life Sci ; 879(25): 2554-60, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21840272

RESUMO

Letosteine has been found to be effective in treating patients with chronic bronchopneumopathies in clinical practice. To provide robust support for its pharmacokinetic and clinical studies, a rapid and sensitive method based on liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was developed and validated for the analysis of letosteine in plasma samples. After protein precipitation, the plasma samples were separated on a reversed-phase C(18) column in less than 1.5 min. The LC-MS/MS system was performed in the positive ion multiple-reaction-monitoring (MRM) mode to produce intensive product ions of m/z 280.1→160.0 for letosteine and m/z 248.1→121.1 for the internal standard, tinidazole. The method was found to have excellent linearity (r ≥ 0.9974), precision (RSD ≤ 5.83%), extraction recovery (71.8-73.0%) and stability (RE, -8.45% to 9.03%) over a concentration range of 0.1140-152.0 µgL(-1). Compared to the previous published radioactive method, LC-MS/MS method showed many advantages including shorter analysis time, simpler preparation procedure, increased sensitivity as well as lower safety risks. In addition, this method was successfully applied to study the pharmacokinetics of letosteine following a single and multiple dose oral administration in Chinese healthy volunteers.


Assuntos
Cromatografia de Fase Reversa/métodos , Espectrometria de Massas em Tandem/métodos , Tiazolidinas/sangue , Tiazolidinas/farmacocinética , Análise de Variância , Estabilidade de Medicamentos , Feminino , Humanos , Análise dos Mínimos Quadrados , Masculino , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA