Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Med Res Rev ; 44(4): 1404-1445, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38279990

RESUMO

Neurodegenerative diseases (NDs) cause progressive loss of neuron structure and ultimately lead to neuronal cell death. Since the available drugs show only limited symptomatic relief, NDs are currently considered as incurable. This review will illustrate the principal roles of the signaling systems of cyclic adenosine and guanosine 3',5'-monophosphates (cAMP and cGMP) in the neuronal functions, and summarize expression/activity changes of the associated enzymes in the ND patients, including cyclases, protein kinases, and phosphodiesterases (PDEs). As the sole enzymes hydrolyzing cAMP and cGMP, PDEs are logical targets for modification of neurodegeneration. We will focus on PDE inhibitors and their potentials as disease-modifying therapeutics for the treatment of Alzheimer's disease, Parkinson's disease, and Huntington's disease. For the overlapped but distinct contributions of cAMP and cGMP to NDs, we hypothesize that dual PDE inhibitors, which simultaneously regulate both cAMP and cGMP signaling pathways, may have complementary and synergistic effects on modifying neurodegeneration and thus represent a new direction on the discovery of ND drugs.


Assuntos
Doenças Neurodegenerativas , Inibidores de Fosfodiesterase , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Inibidores de Fosfodiesterase/uso terapêutico , Inibidores de Fosfodiesterase/farmacologia , Animais , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Cancer Cell Int ; 24(1): 69, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-38341584

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common brain tumor with the worst prognosis. Temozolomide is the only first-line drug for GBM. Unfortunately, the resistance issue is a classic problem. Therefore, it is essential to develop new drugs to treat GBM. As an oncogene, Skp2 is involved in the pathogenesis of various cancers including GBM. In this study, we investigated the anticancer effect of AAA237 on human glioblastoma cells and its underlying mechanism. METHODS: CCK-8 assay was conducted to evaluate IC50 values of AAA237 at 48, and 72 h, respectively. The Cellular Thermal Shift Assay (CETSA) was employed to ascertain the status of Skp2 as an intrinsic target of AAA237 inside the cellular milieu. The EdU-DNA synthesis test, Soft-Agar assay and Matrigel assay were performed to check the suppressive effects of AAA237 on cell growth. To identify the migration and invasion ability of GBM cells, transwell assay was conducted. RT-qPCR and Western Blot were employed to verify the level of BNIP3. The mRFP-GFP-LC3 indicator system was utilized to assess alterations in autophagy flux and investigate the impact of AAA237 on the dynamic fusion process between autophagosomes and lysosomes. To investigate the effect of compound AAA237 on tumor growth in vivo, LN229 cells were injected into the brains of mice in an orthotopic model. RESULTS: AAA237 could inhibit the growth of GBM cells in vitro. AAA237 could bind to Skp2 and inhibit Skp2 expression and the degradation of p21 and p27. In a dose-dependent manner, AAA237 demonstrated the ability to inhibit colony formation, migration, and invasion of GBM cells. AAA237 treatment could upregulate BNIP3 as the hub gene and therefore induce BNIP3-dependent autophagy through the mTOR pathway whereas 3-MA can somewhat reverse this process. In vivo, the administration of AAA237 effectively suppressed the development of glioma tumors with no side effects. CONCLUSION: Compound AAA237, a novel Skp2 inhibitor, inhibited colony formation, migration and invasion of GBM cells in a dose-dependent manner and time-dependent manner through upregulating BNIP3 as the hub gene and induced BNIP3-dependent autophagy through the mTOR pathway therefore it might be a viable therapeutic drug for the management of GBM.

3.
Mol Cell Biochem ; 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38985252

RESUMO

Cardiovascular disease (CVD) stands as a predominant global cause of morbidity and mortality, necessitating effective and cost-efficient therapies for cardiovascular risk reduction. Mitochondrial coupling factor 6 (CF6), identified as a novel proatherogenic peptide, emerges as a significant risk factor in endothelial dysfunction development, correlating with CVD severity. CF6 expression can be heightened by CVD risk factors like mechanical force, hypoxia, or high glucose stimuli through the NF-κB pathway. Many studies have explored the CF6-CVD relationship, revealing elevated plasma CF6 levels in essential hypertension, atherosclerotic cardiovascular disease (ASCVD), stroke, and preeclampsia patients. CF6 acts as a vasoactive and proatherogenic peptide in CVD, inducing intracellular acidosis in vascular endothelial cells, inhibiting nitric oxide (NO) and prostacyclin generation, increasing blood pressure, and producing proatherogenic molecules, significantly contributing to CVD development. CF6 induces an imbalance in endothelium-dependent factors, including NO, prostacyclin, and asymmetric dimethylarginine (ADMA), promoting vasoconstriction, vascular remodeling, thrombosis, and insulin resistance, possibly via C-src Ca2+ and PRMT-1/DDAH-2-ADMA-NO pathways. This review offers a comprehensive exploration of CF6 in the context of CVD, providing mechanistic insights into its role in processes impacting CVD, with a focus on CF6 functions, intracellular signaling, and regulatory mechanisms in vascular endothelial cells.

4.
Pharmacol Res ; 206: 107286, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38936522

RESUMO

The Frizzleds (FZDs) receptors on the cell surface belong to the class F of G protein-coupled receptors (GPCRs) which are the major receptors of WNT protein that mediates the classical WNT signaling pathway and other non-classical pathways. Besides, the FZDs also play a core role in tissue regeneration and tumor occurrence. With the structure and mechanism of FZDs activation becoming clearer, a series of FZDs modulators (inhibitors and agonists) have been developed, with the hope of bringing benefits to the treatment of cancer and degenerative diseases. Most of the FZDs inhibitors (small molecules, antibodies or designed protein inhibitors) block WNT signaling through binding to the cysteine-rich domain (CRD) of FZDs. Several small molecules impede FZDs activation by targeting to the third intracellular domain or the transmembrane domain of FZDs. However, three small molecules (FZM1.8, SAG1.3 and purmorphamine) activate the FZDs through direct interaction with the transmembrane domain. Another type of FZDs agonists are bivalent or tetravalent antibodies which activate the WNT signaling via inducing FZD-LRP5/6 heterodimerization. In this article, we reviewed the FZDs modulators reported in recent years, summarized the critical molecules' discovery processes and the elucidated relevant structural and pharmacological mechanisms. We believe the summaried molecular mechanisms of the relevant modulators could provide important guidance and reference for the future development of FZD modulators.


Assuntos
Receptores Frizzled , Humanos , Receptores Frizzled/metabolismo , Receptores Frizzled/antagonistas & inibidores , Animais , Via de Sinalização Wnt/efeitos dos fármacos , Desenvolvimento de Medicamentos
5.
Bioorg Chem ; 149: 107507, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38850778

RESUMO

Opioids are currently the most effective and widely used painkillers in the world. Unfortunately, the clinical use of opioid analgesics is limited by serious adverse effects. Many researchers have been working on designing and optimizing structures in search of novel µ opioid receptor(MOR) agonists with improved analgesic activity and reduced incidence of adverse effects. There are many strategies to develop MOR drugs, mainly focusing on new low efficacy agonists (potentially G protein biased agonists), MOR agonists acting on different Gα subtype, targeting opioid receptors in the periphery, acting on multiple opioid receptor, and targeting allosteric sites of opioid receptors, and others. This review summarizes the design methods, clinical applications, and structure-activity relationships of small-molecule agonists for MOR based on these different design strategies, providing ideas for the development of safer novel opioid ligands with therapeutic potential.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Humanos , Relação Estrutura-Atividade , Analgésicos Opioides/farmacologia , Analgésicos Opioides/química , Animais , Estrutura Molecular
6.
Bioorg Chem ; 143: 106999, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38035515

RESUMO

Renal fibrosis is the pathological change process of chronic kidney disease deteriorating continuously. When the renal organ is stimulated by external stimuli, it will trigger the damage and phenotypic changes of some intrinsic cells in the kidney. When the body's autoimmune regulation or external treatment is not prompted enough to restore the organ, the pathological process is gradually aggravating, inducing a large amount of intracellular collagen deposition, which leads to the appearance of fibrosis and scarring. The renal parenchyma (including glomeruli and tubules) begins to harden, making it difficult to repair the kidney lesions. In the process of gradual changes in the kidney tissue, the kidney units are severely damaged and the kidney function shows a progressive decline, eventually resulting in the clinical manifestation of end-stage renal failure, namely uremia. This review provides a brief description of the diagnosis, pathogenesis, and potential therapeutic inhibitors of renal fibrosis. Since renal fibrosis has not yet had a clear therapeutic target and related drugs, some potential targets and relevant inhibitors are discussed, especially pharmacological effects and interactions with targets. Some existing natural products have potential efficacy for renal fibrosis, which is also roughly summarized, hoping that this article would have reference significance for the treatment of renal fibrosis.


Assuntos
Produtos Biológicos , Nefropatias , Humanos , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Rim , Nefropatias/tratamento farmacológico , Nefropatias/patologia , Fibrose
7.
Bioorg Chem ; 146: 107278, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38484586

RESUMO

VEGFR, a receptor tyrosine kinase inhibitor (TKI), is an important regulatory factor that promotes angiogenesis and vascular permeability. It plays a significant role in processes such as tumor angiogenesis, tumor cell invasion, and metastasis. VEGFR is mainly composed of three subtypes: VEGFR-1, VEGFR-2, and VEGFR-3. Among them, VEGFR-2 is the crucial signaling receptor for VEGF, which is involved in various pathological and physiological functions. At present, VEGFR-2 is closely related to a variety of cancers, such as non-small cell lung cancer (NSCLC), Hepatocellular carcinoma, Renal cell carcinoma, breast cancer, gastric cancer, glioma, etc. Consequently, VEGFR-2 serves as a crucial target for various cancer treatments. An increasing number of VEGFR inhibitors have been discovered to treat cancer, and they have achieved tremendous success in the clinic. Nevertheless, VEGFR inhibitors often exhibit severe cytotoxicity, resistance, and limitations in indications, which weaken the clinical therapeutic effect. In recent years, many small molecule inhibitors targeting VEGFR have been identified with anti-drug resistance, lower cytotoxicity, and better affinity. Here, we provide an overview of the structure and physiological functions of VEGFR, as well as some VEGFR inhibitors currently in clinical use. Also, we summarize the in vivo and in vitro activities, selectivity, structure-activity relationship, and therapeutic or preventive use of VEGFR small molecule inhibitors reported in patents in the past three years (2021-2023), thereby presenting the prospects and insights for the future development of targeted VEGFR inhibitors.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Renais , Neoplasias Pulmonares , Humanos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/química
8.
Acta Pharmacol Sin ; 45(1): 209-222, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37749236

RESUMO

Glioblastoma (GBM) is the most common malignant tumor in the brain with temozolomide (TMZ) as the only approved chemotherapy agent. GBM is characterized by susceptibility to radiation and chemotherapy resistance and recurrence as well as low immunological response. There is an urgent need for new therapy to improve the outcome of GBM patients. We previously reported that 3-O-acetyl-11-keto-ß-boswellic acid (AKBA) inhibited the growth of GBM. In this study we characterized the anti-GBM effect of S670, a synthesized amide derivative of AKBA, and investigated the underlying mechanisms. We showed that S670 dose-dependently inhibited the proliferation of human GBM cell lines U87 and U251 with IC50 values of around 6 µM. Furthermore, we found that S670 (6 µM) markedly stimulated mitochondrial ROS generation and induced ferroptosis in the GBM cells. Moreover, S670 treatment induced ROS-mediated Nrf2 activation and TFEB nuclear translocation, promoting protective autophagosome and lysosome biogenesis in the GBM cells. On the other hand, S670 treatment significantly inhibited the expression of SXT17, thus impairing autophagosome-lysosome fusion and blocking autophagy flux, which exacerbated ROS accumulation and enhanced ferroptosis in the GBM cells. Administration of S670 (50 mg·kg-1·d-1, i.g.) for 12 days in a U87 mouse xenograft model significantly inhibited tumor growth with reduced Ki67 expression and increased LC3 and LAMP2 expression in the tumor tissues. Taken together, S670 induces ferroptosis by generating ROS and inhibiting STX17-mediated fusion of autophagosome and lysosome in GBM cells. S670 could serve as a drug candidate for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Ferroptose , Glioblastoma , Humanos , Animais , Camundongos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Autofagossomos/metabolismo , Amidas/farmacologia , Transdução de Sinais , Lisossomos/metabolismo , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Proteínas Qa-SNARE
9.
Bioorg Chem ; 141: 106869, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37797454

RESUMO

The opioids have been used for more than a thousand years and are not only the most widely prescribed drugs for moderate to severe pain and acute pain, but also the preferred drugs. However, their non-analgesic effects, especially respiratory depression and potential addiction, are important factors that plague the safety of clinical use and are an urgent problem for pharmacological researchers to address. Current research on analgesic drugs has evolved into different directions: de-opioidization; application of pharmacogenomics to individualize the use of opioids; development of new opioids with less adverse effects. The development of new opioid drugs remains a hot research topic, and with the in-depth study of opioid receptors and intracellular signal transduction mechanisms, new research ideas have been provided for the development of new opioid analgesics with less side effects and stronger analgesic effects. The development of novel opioid drugs in turn includes selective opioid receptor ligands, biased opioid receptor ligands, and multi-target opioid receptor ligands and positive allosteric modulators (PAMs) or antagonists and the single compound as multi-targeted agnoists/antagonists for different receptors. PAMs strategies are also getting newer and are the current research hotspots, including the BMS series of compounds and others, which are extensive and beyond the scope of this review. This review mainly focuses on the selective/biased/multi-targeted MOR/DOR/KOR (mu opioid receptor/delta opioid receptor/kappa opioid receptor) small molecule ligands and involves some cryo-electron microscopy (cryoEM) and structure-based approaches as well as the single compound as multi-targeted agnoists/antagonists for different receptors from 2019 to 2022, including discovery history, activities in vitro and vivo, and clinical studies, in an attempt to provide ideas for the development of novel opioid analgesics with fewer side effects.


Assuntos
Analgésicos Opioides , Receptores Opioides kappa , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Receptores Opioides mu , Microscopia Crioeletrônica , Analgésicos , Ligantes
10.
Bioorg Chem ; 141: 106889, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37813074

RESUMO

Most patients with advanced prostate cancer (PCa) will develop metastatic castration-resistant prostate cancer (mCRPC) after androgen deprivation therapy, at this time the tumor enters the end stage, and the clinical treatment is very complicated, which requires rationalization of drugs to prolong the life of patients while improving their quality of life. Prostate-specific membrane antigen (PSMA) is a promising biological target for drug delivery in mCRPC due to its high level of specific expression in PCa cell membranes and low expression in normal tissues. Non-radioactive PSMA-targeted small molecule-drug conjugates (SMDCs) are gradually becoming a heat of discovery due to their good affinity and specificity; simple synthesis steps and transport management methods. Non-radioactive PSMA-targeted SMDCs under investigation can be divided into two categories: SMDCs and dual-ligand coupled drugs, among which SMDCs are the most widespread form of this type of conjugate. SMDCs have three key components: cytotoxic load, linker, and small molecule targeting ligands. SMDCs are internalized into the cell after binding to PSMA on the cell membrane and stored in endosomes and lysosomes, where they are usually enzymatically cleaved to allow precise release of cytotoxic molecules and uniform diffusion into the tumor tissue. More than a dozen non-radioactive PSMA-targeted SMDCs have been developed, many of which have shown favorable properties in both in vitro and in vivo evaluations, demonstrating more favorable results than unmodified cytotoxic drugs. Therefore, non-radioactive PSMA-targeted SMDCs have great therapeutic potential for mCRPC as a form of targeted therapy.


Assuntos
Antineoplásicos , Neoplasias de Próstata Resistentes à Castração , Humanos , Masculino , Antagonistas de Androgênios , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Preparações Farmacêuticas , Qualidade de Vida
11.
Bioorg Chem ; 138: 106577, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37178649

RESUMO

Protein kinases constitute the largest group within the kinase family, and mutations and translocations of protein kinases due to genetic alterations are intimately linked to the pathogenesis of numerous diseases. Bruton's tyrosine kinase (BTK) is a member of the protein kinases and plays a pivotal role in the development and function of B cells. BTK belongs to the tyrosine TEC family. The aberrant activation of BTK is closely associated with the pathogenesis of B-cell lymphoma. Consequently, BTK has always been a critical target for treating hematological malignancies. To date, two generations of small-molecule covalent irreversible BTK inhibitors have been employed to treat malignant B-cell tumors, and have exhibited clinical efficacy in hitherto refractory diseases. However, these drugs are covalent BTK inhibitors, which inevitably lead to drug resistance after prolonged use, resulting in poor tolerance in patients. The third-generation non-covalent BTK inhibitor Pirtobrutinib has obtained approval for marketing in the United States, thereby circumventing drug resistance caused by C481 mutation. Currently, enhancing safety and tolerance constitutes the primary issue in developing novel BTK inhibitors. This article systematically summarizes recently discovered covalent and non-covalent BTK inhibitors and classifies them according to their structures. This article also provides a detailed discussion of binding modes, structural features, pharmacological activities, advantages and limitations of typical compounds within each structure type, providing valuable references and insights for developing safer, more effective and more targeted BTK inhibitors in future studies.


Assuntos
Neoplasias , Humanos , Relação Estrutura-Atividade , Tirosina Quinase da Agamaglobulinemia , Neoplasias/tratamento farmacológico , Linfócitos B/metabolismo , Inibidores de Proteínas Quinases/química
12.
Bioorg Chem ; 138: 106592, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37178650

RESUMO

Pulmonary fibrosis is the end-stage change of a large class of lung diseases characterized by the proliferation of fibroblasts and the accumulation of a large amount of extracellular matrix, accompanied by inflammatory damage and tissue structure destruction, which also shows the normal alveolar tissue is damaged and then abnormally repaired resulting in structural abnormalities (scarring). Pulmonary fibrosis has a serious impact on the respiratory function of the human body, and the clinical manifestation is progressive dyspnea. The incidence of pulmonary fibrosis-related diseases is increasing year by year, and no curative drugs have appeared so far. Nevertheless, research on pulmonary fibrosis have also increased in recent years, but there are no breakthrough results. Pathological changes of pulmonary fibrosis appear in the lungs of patients with coronavirus disease 2019 (COVID-19) that have not yet ended, and whether to improve the condition of patients with COVID-19 by means of the anti-fibrosis therapy, which are the questions we need to address now. This review systematically sheds light on the current state of research on fibrosis from multiple perspectives, hoping to provide some references for design and optimization of subsequent drugs and the selection of anti-fibrosis treatment plans and strategies.


Assuntos
COVID-19 , Fibrose Pulmonar , Humanos , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , COVID-19/patologia , Pulmão , Fibrose , Fibroblastos
13.
Pharmacol Res ; 181: 106259, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35577307

RESUMO

Lung cancer is by far the leading cause of cancer death worldwide, and 85% of patients are diagnosed with non-small cell lung cancer (NSCLC), which is still very difficult to treat. Skp2 functions as an oncogene that participates in processes of many cancers. Here, we report a novel Skp2 inhibitor AAA-237 that binds to Skp2 protein and inhibits the proliferation of the NSCLC cells. We further investigated the anti-NSCLC mechanism of AAA-237 and found that it arrested the cell cycle at the G0/G1 phase by targeting Skp2 to reduce the degradation of p21Cip1 and p27Kip1 or by transcriptionally activating FOXO1 to increase the mRNA expression of p21Cip1 and p27Kip1. More importantly, we found that treatment of a high concentration AAA-237 could induce apoptosis of NSCLC cells and treatment of a low AAA-237 concentration for a longer time could induce senescence of NSCLC cells. Similar results were found in nude mice xenografted with A549 cells. AAA-237 inhibited tumor growth by inducing apoptosis and senescence in a dose-dependent manner. Considering these results, we propose that AAA-237 could be a promising therapeutic drug for treating patients with NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Pontos de Checagem do Ciclo Celular , Neoplasias Pulmonares , Proteínas Quinases Associadas a Fase S , Células A549 , Animais , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Fase G1 , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Proteínas Quinases Associadas a Fase S/antagonistas & inibidores
14.
Bioorg Chem ; 129: 106181, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36302332

RESUMO

In recent years, the development of dual target drugs has become a research hotspot in cancer treatment and the reasonable design of the drugs is critical. The nonclearable linked pharmacophore mode is one of the commonly used strategies for designing dual target drugs, it can connect the pharmacophores of two synergistic target inhibitors into one molecule through the linker, which greatly improves the utilization of drugs. Epigenetic modifications as a potential treatment for multiple diseases have always been a subject of great concern, and Histone deacetylases (HDAC) plays an important role. Janus Kinase (JAK) is a family of intracellular non-receptor tyrosine kinases that transduce cytokine-mediated signals through the JAK-signal transducers and the activators of transcription (STAT) pathway. Studies showed the combination of HDAC and JAK inhibitors exhibited synergistic effects in breast cancer treatment [1]. In addition, the pharmacophore models of the aforementioned two inhibitors indicate similar essential features. Further investigation on recent years' progress in the field demonstrated the nonclearable linked pharmacophore mode, using different length carbon chains as linkers to connect the pharmacophores of the two inhibitors, is the main strategy to design HDAC/JAK dual-target inhibitors which has been verified to be effective in biological activity tests. This review takes recent years' HDAC/JAK dual target inhibitors' development details as an example to summarize the general ideas behind the scene. We wish to provide the readers a theoretical basis for the development of more efficient dual-target or multi-target drugs in future.


Assuntos
Inibidores de Janus Quinases , Neoplasias , Histona Desacetilases/metabolismo , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Pirimidinas/farmacologia , Janus Quinases/metabolismo , Janus Quinases/farmacologia , Transdução de Sinais , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
15.
Invest New Drugs ; 37(4): 616-624, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30168013

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the most serious life-threatening malignancies. Although chemotherapeutic targets and agents for ESCC have made much progress recently, the efficacy is still unsatisfactory. Therefore, there is still an unmet medical need for patients with ESCC. Here, we report the expression status of HDAC1 in human ESCC and matched paracancerous tissues, and the results indicated that HDAC1 was generally upregulated in ESCC specimens. Furthermore, we comprehensively assessed the anti-ESCC activity of a highly active HDAC1 inhibitor quisinostat. Quisinostat could effectively suppress cellular viability and proliferation of ESCC cells, as well as induce cell cycle arrest and apoptosis even at low treatment concentrations. The effectiveness was also observed in KYSE150 xenograft model when quisinostat was administered at tolerated doses (3 mg/kg and 10 mg/kg). Meanwhile, quisinostat also had the ability to suppress the migration and invasion (pivotal steps of tumor metastasis) of ESCC cells. Western blot analysis indicated that quisinostat exerted its anti-ESCC effects mainly through blockade of Akt/mTOR and MAPK/ERK signaling cascades. Overall, HDAC1 may serve as a potential therapeutic target for ESCC, and quisinostat deserves to be further assessed as a promising drug candidate for the treatment of ESCC.


Assuntos
Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Histona Desacetilase 1/antagonistas & inibidores , Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos SCID , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral
17.
Chem Biodivers ; 16(11): e1900400, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31482617

RESUMO

The Keap1-Nrf2/ARE signaling pathway is an important defense system against exogenous and endogenous oxidative stress injury. The dysregulation of the signaling pathway is associated with many diseases, such as cancer, diabetes, and respiratory diseases. Over the years, a wide range of natural products has provided sufficient resources for the discovery of potential therapeutic drugs. Among them, polyphenols possess Nrf2 activation, not only inhibit the production of ROS, inhibit Keap1-Nrf2 protein-protein interaction, but also degrade Keap1 and regulate the Nrf2 related pathway. In fact, with the continuous improvement of natural polyphenols separation and purification technology and further studies on the Keap1-Nrf2 molecular mechanism, more and more natural polyphenols monomer components of Nrf2 activators have been gradually discovered. In this view, we summarize the research status of natural polyphenols that have been found with apparent Nrf2 activation and their action modes. On the whole, this review may guide the design of novel Keap1-Nrf2 activator.


Assuntos
Produtos Biológicos/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Polifenóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Produtos Biológicos/química , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Estrutura Molecular , Fator 2 Relacionado a NF-E2/metabolismo , Polifenóis/química
18.
Chem Biodivers ; 16(9): e1900304, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31338947

RESUMO

Glycogen synthase kinase-3 (GSK-3) plays an important regulatory role in various signaling pathways; such as PI3 K/AKT, which is closely related to the occurrence and development of tumors. At present, the most reported active GSK-3 inhibitors have the same structure: lactam ring or amide structure. To find out the GSK-3ß small molecule inhibitor with novel, safe, efficient and more uncomplicated synthesis method, we analyzed in-depth reported crystal-binding patterns of GSK-3ß small molecule inhibitor with GSK-3ß protein, and designed and synthesized 17 non-reported 3,5-diamino-N-substituted benzamide compounds. Their structures were confirmed by 1 H-NMR, 13 C-NMR, and HR-MS. The preliminary screening of tumor cytotoxicity of compounds in vitro was detected by MTT, and their structure-activity relationships were illustrated. The results have shown that 3,5-diamino-N-[3-(trifluoromethyl)phenyl]benzamide (4d) exhibited significant tumor cytotoxicity against human colon cancer cells (HCT-116) with IC50 of 8.3 µm and showed commendable selectivity to GSK-3ß. In addition, Compound 4d induced apoptosis to some extent and possessed modest PK properties.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Desenho de Fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Benzamidas/síntese química , Benzamidas/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Masculino , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Ratos , Ratos Sprague-Dawley , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
19.
Biochemistry ; 57(42): 6070-6077, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30231198

RESUMO

The cAMP signaling system plays important roles in the physiological processes of pathogen yeast Candida albicans, but its functional mechanism has not been well illustrated. Here, we report the enzymatic characterization and crystal structures of C. albicans phosphodiesterase 2 (caPDE2) in the unliganded and 3-isobutyl-1-methylxanthine-complexed forms. caPDE2 is a monomer in liquid and crystal states and specifically hydrolyzes cAMP with a KM of 35 nM. It does not effectively hydrolyze cGMP as shown by the 1.32 × 105-fold specificity of cAMP/cGMP. The crystal structure of caPDE2 shows significant differences from those of human PDEs. First, the N-terminal fragment of caPDE2 (residues 1-201) tightly associates with the catalytic domain to form a rigid molecular entity, implying its stable molecular conformation for C. albicans to resist environmental stresses. Second, the M-loop, a critical fragment for binding of the substrate and inhibitors to human PDEs, is not a part of the caPDE2 active site. This feature of caPDE2 may provide a structural basis for the design of selective inhibitors for the treatment of yeast infection.


Assuntos
Candida albicans/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/química , Proteínas Fúngicas/química , Cristalografia por Raios X , Domínios Proteicos , Estrutura Secundária de Proteína , Relação Estrutura-Atividade
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(3): 437-439, 2018 Jun 10.
Artigo em Zh | MEDLINE | ID: mdl-29896750

RESUMO

OBJECTIVE: To determine the genetic cause of an infant with multiple congenital anomalies. METHODS: Routine karyotype analysis and chromosome microarray analysis (CMA) were carried out for the infant and her parents. RESULTS: CMA has detected a 9.3 Mb duplication at 9q34.11-q34.3. G-banding analysis suggested that the infant has a 46,XX,der(1)add(1)(p34.1) karyotype, while her father was 46, XY, t(1,9)(p36.3;q34.1). Fluorescence in situ hybridization (FISH) analysis confirmed that the 9q34 duplication has derived from the balanced translocation carried by the father. CONCLUSION: A 9.3 Mb duplication was detected within the 9q34 region in an infant featuring multiple congenital anomalies. CMA and FISH have enabled detection of this duplication and facilitated genetic counseling and prevention of birth of further affected offspring.


Assuntos
Anormalidades Múltiplas/genética , Trissomia/genética , Bandeamento Cromossômico , Cromossomos Humanos Par 9/genética , Feminino , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Translocação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA