Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 411-418, 2024 May.
Artigo em Chinês | MEDLINE | ID: mdl-38790097

RESUMO

Objective To explore the effects of aloperine (Alo) on cigarette smoke-induced injury in human bronchial epithelial cells and its potential mechanism. Methods After human bronchial epithelial 16HBE cells were co-treated by 100 mL/L cigarette smoke extract (CSE) and various concentrations (50,100 and 200 µmol/L) of Alo, cell viability was assessed using CCK-8 assay. Lactate dehydrogenase (LDH) activity was measured with a related kit. Cell apoptosis was evaluated using the terminal-deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) and Western blot analysis. The levels of inflammatory factors were detected by ELISA. Oxidative stress levels were assessed using 2'7'-dichlorofluorescin diacetate (DCFH-DA) staining. The expression of Toll-like receptor 4 (TLR4)/nuclear factor-kappaB (NF-κB)/NLR family pyrin domain containing 3 (NLRP3) signaling-associated proteins was measured by Western blot analysis. After cells were co-treated with 100 mL/L CSE and 200 µmol/L Alo, the aforementioned assays were applied to evaluate the effects of TLR4 overexpression on the TLR4/NF-κB/NLRP3 signaling, LDH activity, apoptosis, inflammatory response and oxidative stress in cells. Results CSE exposure might inhibit 16HBE cell viability, increase LDH activity, apoptosis, inflammatory response and oxidative stress levels and activate TLR4/NF-κB/NLRP3 signaling. Treatment with Alo promoted cell viability, decreased LDH activity, cell apoptosis, inflammation and oxidative stress levels, and inactivated TLR4/NF-κB/NLRP3 signaling. Furthermore, TLR4 overexpression might reverse the protective role of Alo treatment in CSE-induced injury in 16HBE cells. Conclusion Alo may ameliorate CSE-induced injury in human bronchial epithelial cells via inhibiting TLR4/NF-κB/NLRP3 signaling.


Assuntos
Apoptose , Brônquios , Células Epiteliais , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Quinolizidinas , Transdução de Sinais , Receptor 4 Toll-Like , Humanos , Receptor 4 Toll-Like/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Brônquios/citologia , Brônquios/metabolismo , Brônquios/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Apoptose/efeitos dos fármacos , Quinolizidinas/farmacologia , Fumaça/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Linhagem Celular , Nicotiana/efeitos adversos
2.
Bioorg Chem ; 147: 107317, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38583252

RESUMO

By inducing steric activation of the 10CH bond with a 12-acyl group to form a key imine oxime intermediate, 20 novel (10S)-10,12-disubstituted aloperine derivatives were successfully synthesized and assessed for their antiviral efficacy against HCoV-OC43. Of them, compound 3i exhibited the moderate activities against HCoV-OC43, as well as against the SARS-CoV-2 variant EG.5.1 with the comparable EC50 values of 4.7 and 4.1 µM. A mechanism study revealed that it inhibited the protease activity of host TMPRSS2 by binding to an allosteric site, rather than the known catalytic center, different from that of camostat. Also, the combination of compound 3i and molnupiravir, as an RdRp inhibitor, showed an additive antiviral effect against HCoV-OC43. The results provide a new binding mode and lead compound for targeting TMPRSS2, with an advantage in combating broad-spectrum coronavirus.


Assuntos
Sítio Alostérico , Antivirais , Coronavirus Humano OC43 , Quinolizidinas , Serina Endopeptidases , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Serina Endopeptidases/metabolismo , Humanos , Coronavirus Humano OC43/efeitos dos fármacos , Coronavirus Humano OC43/química , Quinolizidinas/química , Quinolizidinas/farmacologia , Quinolizidinas/síntese química , Sítio Alostérico/efeitos dos fármacos , Relação Estrutura-Atividade , Descoberta de Drogas , SARS-CoV-2/efeitos dos fármacos , Estrutura Molecular , Testes de Sensibilidade Microbiana , Relação Dose-Resposta a Droga
3.
Molecules ; 28(19)2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37836759

RESUMO

Fourteen quinolizidine derivatives, structurally related to the alkaloids lupinine and cytisine and previously studied for other pharmacological purposes, were presently tested for antiarrhythmic, and other cardiovascular effects on isolated guinea pig heart tissues in comparison to well-established reference drugs. According to their structures, the tested compounds are assembled into three subsets: (a) N-(quinolizidinyl-alkyl)-benzamides; (b) 2-(benzotriazol-2-yl)methyl-1-(quinolizidinyl)alkyl-benzimidazoles; (c) N-substituted cytisines. All compounds but two displayed antiarrhythmic activity that was potent for compounds 4, 1, 6, and 5 (in ascending order). The last compound (N-(3,4,5-trimethoxybenzoyl)aminohomolupinane) was outstanding, exhibiting a nanomolar potency (EC50 = 0.017 µM) for the increase in the threshold of ac-arrhythmia. The tested compounds shared strong negative inotropic activity; however, this does not compromise the value of their antiarrhythmic action. On the other hand, only moderate or modest negative chronotropic and vasorelaxant activities were commonly observed. Compound 5, which has high antiarrhythmic potency, a favorable cardiovascular profile, and is devoid of antihypertensive activity in spontaneously hypertensive rats, represents a lead worthy of further investigation.


Assuntos
Alcaloides , Quinolizidinas , Esparteína , Ratos , Animais , Cobaias , Quinolizidinas/farmacologia , Antiarrítmicos/farmacologia , Antiarrítmicos/química , Coração , Esparteína/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Alcaloides/farmacologia
4.
Alkaloids Chem Biol ; 89: 1-37, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36731966

RESUMO

Quinolizidine alkaloids isolated from various marine and terrestrial animals and plants are primarily composed of lupinine-, matrine-, and sparteine-type alkaloids. Matrine, phenanthroquinolizidines, bis-quinolizidines, and small molecules from amphibian skins are representative compounds of such alkaloids. Quinolizidine alkaloids harbor anticancer, antibacterial, antiinflammatory, antifibrosis, antiviral, and anti-arrhythmia. In this chapter, we comprehensively outline the biological activity and pharmacological action of quinolizidine alkaloids and discuss new avenues toward the discovery of novel and more efficient drugs based on these naturally occurring compounds. It is urgent for basic research and clinical practice to conduct more targeted comprehensive research based on the lead drugs of quinolizidine alkaloids with significant pharmacological activity.


Assuntos
Alcaloides , Quinolizidinas , Esparteína , Animais , Alcaloides Quinolizidínicos , Alcaloides/farmacologia , Quinolizidinas/farmacologia , Anti-Inflamatórios , Matrinas
5.
Protein Pept Lett ; 30(3): 250-259, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36734907

RESUMO

BACKGROUND: Aloperine (ALO) is an important active component of quinolizidine alkaloids in Sophora flavescens A and Sophora alopecuroides L, and has effective anticancer activity against multiple cancers. However, the influence and mechanism of ALO on migration, invasion, and adhesion in bladder cancer cells remain unclear. OBJECTIVE: The aim of this study is to determine the anticancer effect of ALO on migration, invasion, and adhesion in bladder cancer cells and to investigate its potential TIMP-4-related mechanism. METHODS: Cell viability, cytotoxicity, wound healing, Transwell invasion, cell adhesion, real-time qPCR, western blot, and ELISA assays were performed to analyze the effect of ALO on migration, invasion, and adhesion in bladder cancer 5637 and UM-UC-3 cells. Furthermore, the anti-TIMP-4 antibody was used to explore the potential effect on ALO-inhibited bladder cancer cells. RESULTS: We have found that ALO significantly suppressed migration, invasion, and adhesion in bladder cancer cells. Furthermore, ALO could downregulate the expression of MMP-2 and MMP-9 mRNAs and proteins, and increase the expression of TIMP-4 mRNA and protein. Moreover, the anti- TIMP-4 antibody reversed the prevention of migration, invasion, and adhesion in ALO-treated bladder cancer cells. CONCLUSION: The data in this study suggest that ALO suppressed migration, invasion, and adhesion in bladder cancer cells by upregulating the expression of TIMP-4.


Assuntos
Quinolizidinas , Neoplasias da Bexiga Urinária , Humanos , Quinolizidinas/farmacologia , Linhagem Celular Tumoral , Neoplasias da Bexiga Urinária/tratamento farmacológico , Movimento Celular
6.
Int J Mol Sci ; 23(17)2022 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-36077056

RESUMO

COVID-19, caused by the highly transmissible severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has rapidly spread and become a pandemic since its outbreak in 2019. We have previously discovered that aloperine is a new privileged scaffold that can be modified to become a specific antiviral compound with markedly improved potency against different viruses, such as the influenza virus. In this study, we have identified a collection of aloperine derivatives that can inhibit the entry of SARS-CoV-2 into host cells. Compound 5 is the most potent tested aloperine derivative that inhibited the entry of SARS-CoV-2 (D614G variant) spike protein-pseudotyped virus with an IC50 of 0.5 µM. The compound was also active against several other SARS-CoV-2 variants including Delta and Omicron. Results of a confocal microscopy study suggest that compound 5 inhibited the viral entry before fusion to the cell or endosomal membrane. The results are consistent with the notion that aloperine is a privileged scaffold that can be used to develop potent anti-SARS-CoV-2 entry inhibitors.


Assuntos
Tratamento Farmacológico da COVID-19 , Inibidores da Fusão de HIV , Quinolizidinas , Humanos , Pandemias , Quinolizidinas/farmacologia , SARS-CoV-2 , Internalização do Vírus
7.
Fitoterapia ; 162: 105278, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35970410

RESUMO

Asthma is a high-incidence disease in the world. Oxysophocarpine (OSC), a quinolizidine alkaloid displays various pharmacological functions including anti-inflammation, neuroprotective, anti-virus and antioxidant. Here, we established mice and cell asthmatic model to explore the effects of OSC for asthma treatment. Mice were sensitized and challenged with ovalbumin (OVA) and treated with OSC before challenge. Enzyme-linked immuno sorbent assay (ELISA), hematoxylin and eosin (H&E), periodic acid-schiff (PAS), tolonium chloride staining and immunohistochemical assay were performed. OSC treatment inhibited inflammatory cell infiltration and mucus secretion in the airway, reduced IgE level in mouse serum and decreased IL-4, IL-5 production in bronchoalveolar lavage fluid (BALF). OSC also reduced the spleen index to regulate immune function. Meanwhile, NCI-H292 cells were induced by lipopolysaccharide (LPS) to simulate airway epithelial injury. OSC pretreatment decreased the IL-6 and IL-8 cytokine levels, mucin 5 AC expression, and mucin 5 AC mRNA level in the cell model. Further, OSC suppressed the phosphorylation of c-Jun N-terminal kinase (JNK), and activator protein 1 (AP-1, Fos and Jun). These findings revealed that OSC alleviated bronchial asthma associated with JNK/AP-1 signaling pathway.


Assuntos
Alcaloides , Asma , Quinolizidinas , Alcaloides/metabolismo , Alcaloides/farmacologia , Animais , Antioxidantes/farmacologia , Asma/tratamento farmacológico , Citocinas/metabolismo , Modelos Animais de Doenças , Amarelo de Eosina-(YS)/metabolismo , Amarelo de Eosina-(YS)/farmacologia , Amarelo de Eosina-(YS)/uso terapêutico , Hematoxilina/metabolismo , Hematoxilina/farmacologia , Hematoxilina/uso terapêutico , Imunoglobulina E , Interleucina-4/metabolismo , Interleucina-4/farmacologia , Interleucina-4/uso terapêutico , Interleucina-5/metabolismo , Interleucina-5/farmacologia , Interleucina-5/uso terapêutico , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Mucinas/metabolismo , Mucinas/farmacologia , Mucinas/uso terapêutico , Muco/metabolismo , Ovalbumina/metabolismo , Ácido Periódico/metabolismo , Ácido Periódico/farmacologia , Ácido Periódico/uso terapêutico , Quinolizidinas/farmacologia , RNA Mensageiro/metabolismo , Cloreto de Tolônio/metabolismo , Cloreto de Tolônio/farmacologia , Cloreto de Tolônio/uso terapêutico , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-1/farmacologia , Fator de Transcrição AP-1/uso terapêutico
8.
Molecules ; 27(16)2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-36014321

RESUMO

The discovery and the development of safe and efficient therapeutics against arthritogenic alphaviruses (e.g., chikungunya virus) remain a continuous challenge. Alkaloids are structurally diverse and naturally occurring compounds in plants, with a wide range of biological activities including beneficial effects against prominent pathogenic viruses and inflammation. In this short review, we discuss the effects of some alkaloids of three biologically relevant structural classes (isoquinolines, indoles and quinolizidines). Based on various experimental models (viral infections and chronic diseases), we highlight the immunomodulatory effects of these alkaloids. The data established the capacity of these alkaloids to interfere in host antiviral and inflammatory responses through key components (antiviral interferon response, ROS production, inflammatory signaling pathways and pro- and anti-inflammatory cytokines production) also involved in alphavirus infection and resulting inflammation. Thus, these data may provide a convincing perspective of research for the use of alkaloids as immunomodulators against arthritogenic alphavirus infection and induced inflammation.


Assuntos
Alcaloides , Infecções por Alphavirus , Vírus Chikungunya , Quinolizidinas , Alcaloides/farmacologia , Alcaloides/uso terapêutico , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/patologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus Chikungunya/fisiologia , Humanos , Indóis/uso terapêutico , Inflamação , Isoquinolinas , Quinolizidinas/farmacologia
9.
J Agric Food Chem ; 70(29): 9214-9226, 2022 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-35849433

RESUMO

As part of our ongoing investigation of pesticide active quinolizidine alkaloids (QAs) from the family Fabaceae, the chemical constituents of the seeds of Thermopsis lanceolata R. Br. were systematically investigated. Bioassay-guided fractionation and purification of the crude extract led to the isolation of seventeen new QAs (1-17), including three new naturally occurring compounds (15-17), along with 15 known compounds (18-32). Their structures were elucidated by comprehensive spectroscopic data analysis (IR, UV, NMR, and HRESIMS) and quantum chemistry calculations (13C NMR and ECD). The antitomato spotted wilt virus activities and insecticidal activities against Aphis fabae, Nilaparvata lugens (Stal), and Tetranychus urticae of compounds 1-32 were screened using the lesion counting method, spray method, and rice-stem dipping method, respectively. Biological tests indicated that compounds 6, 9, 10, and 18 displayed significant anti-TSWV activities compared with the positive control ningnanmycin. Compounds 3, 4, and 5 showed better insecticidal activities against A. fabae with LC50 values of 10.07, 12.07, and 6.56 mg/L, respectively. Moreover, compounds 5, 18, and 24 exhibited moderate insecticidal activities against N. lugens (Stal) with LC50 values of 37.91, 53.44, and 31.21 mg/L, respectively. Furthermore, compounds 9 and 10 exhibited moderate insecticidal activities against T. urticae.


Assuntos
Alcaloides , Afídeos , Fabaceae , Inseticidas , Quinolizidinas , Alcaloides/análise , Alcaloides/farmacologia , Animais , Inseticidas/química , Quinolizidinas/farmacologia , Sementes/química
10.
Molecules ; 27(1)2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-35011535

RESUMO

Fusarium oxysporum is an aggressive phytopathogen that affects various plant species, resulting in extensive local and global economic losses. Therefore, the search for competent alternatives is a constant pursuit. Quinolizidine alkaloids (QA) are naturally occurring compounds with diverse biological activities. The structural diversity of quinolizidines is mainly contributed by species of the family Fabaceae, particularly the genus Lupinus. This quinolizidine-based chemo diversity can be explored to find antifungals and even mixtures to address concomitant effects on F. oxysporum. Thus, the antifungal activity of quinolizidine-rich extracts (QREs) from the leaves of eight greenhouse-propagated Lupinus species was evaluated to outline promising QA mixtures against F. oxysporum. Thirteen main compounds were identified and quantified using an external standard. Quantitative analysis revealed different contents per quinolizidine depending on the Lupinus plant, ranging from 0.003 to 32.8 mg/g fresh leaves. Bioautography showed that all extracts were active at the maximum concentration (5 µg/µL). They also exhibited >50% mycelium growth inhibition. All QREs were fungistatic except for the fungicidal QRE of L. polyphyllus Lindl. Angustifoline, matrine, 13α-hydroxylupanine, and 17-oxolupanine were ranked to act jointly against the phytopathogen. Our findings constitute reference information to better understand the antifungal activity of naturally afforded QA mixtures from these globally important plants.


Assuntos
Antifúngicos/farmacologia , Lupinus/química , Extratos Vegetais/farmacologia , Quinolizidinas/farmacologia , Antifúngicos/química , Cromatografia Gasosa-Espectrometria de Massas , Efeito Estufa , Lupinus/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacologia , Extratos Vegetais/química , Quinolizidinas/química
11.
Mini Rev Med Chem ; 22(5): 729-742, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34488611

RESUMO

In this review, an effort towards presenting an all-around account of the recent progress on the natural product, aloperine, is made, and the antivirus structure-activity relationship of its derivatives is also summarized comprehensively. In addition, the principal pharmacological effects and corresponding molecular mechanisms of aloperine are discussed. Some new structural modifications of aloperine are also given, which might provide brief guidance for further investigations on the natural product aloperine.


Assuntos
Produtos Biológicos , Quinolizidinas , Produtos Biológicos/farmacologia , Piperidinas/química , Quinolizidinas/farmacologia , Relação Estrutura-Atividade
12.
Nat Prod Res ; 36(7): 1781-1788, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32924588

RESUMO

Seventeen quinolizidine alkaloids, including a new matrine-type one, sophcence A (1), were isolated from the roots of Sophora flavescens Alt. The structure of compound 1 was elucidated by means of 1D and 2D NMR, as well as HR-ESI-MS spectroscopic data. The NMR data of (-)-Δ7-dehydrosophoramine (10) and oxy-N-methylcytisine (12) were reported for the first time. In addition, (+)-sophoranol (4) exhibited moderate inhibition on lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW 264.7 macrophages with IC50 value of 22.14 µM, while lupanine (17) was found to inhibit the growth of human glioma stem cells GSC-3# at 20 µg/mL.


Assuntos
Alcaloides , Quinolizidinas , Sophora , Alcaloides/química , Humanos , Lipopolissacarídeos/farmacologia , Raízes de Plantas/química , Quinolizidinas/farmacologia , Quinolizinas/química , Sophora/química
13.
Eur J Pharmacol ; 916: 174721, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-34954231

RESUMO

Type 1 diabetes (T1D) is a metabolic dysfunction characterized by the selective destruction of islet ß-cells, with oxidative stress playing an essential role in the manifestation of this disease state. Aloperine (ALO) represents the main active alkaloid extracted from the traditional Chinese herbal Sophora alopecuroides L. and features outstanding antioxidative properties. In this study, T1D was induced by a single high dose streptozotocin (STZ, 150 mg/kg, intraperitoneal) in mice. Diabetic animals were intragastrically administered ALO at a dose of 50 mg/kg/day. Notably, treatment of ALO (50 mg/kg/day) for seven consecutive days could observably reverse the onset of diabetes induced by STZ accompanied by weight gain, lower blood glucose levels, and relief of ß-cells damage. Our in vitro study further demonstrated that ALO protected ß-cells from STZ/hydrogen peroxide-induced oxidative damage as manifested by increased expression of MnSOD and CAT. Furthermore, a network pharmacology study revealed that NOS1 represented the main target of ALO. Mechanistic studies subsequently showed that treatment of ALO increased the expression of NOS1, whereas NOS2 was decreased. Moreover, a docking study carried out suggested that ALO could fit into the binding pocket of human NOS1 and molecular dynamics simulation further validated this docking event. Collectively, the administration of ALO prior to diabetes could be a viable approach to the prevention of ß-cell injury. This study may offer a novel potential herbal medicine against T1D and may further help improve the understanding of the underlying molecular mechanisms of ALO-mediated protection against oxidative stress.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Óxido Nítrico Sintase Tipo I , Quinolizidinas , Animais , Glicemia/metabolismo , Citoproteção , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Camundongos , Óxido Nítrico Sintase Tipo I/metabolismo , Estresse Oxidativo , Piperidinas/farmacologia , Quinolizidinas/administração & dosagem , Quinolizidinas/farmacologia , Quinolizidinas/uso terapêutico , Estreptozocina
14.
Tissue Cell ; 74: 101706, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34883316

RESUMO

Anti-tumorous effect of Aloperine (ALO) has been previously found. This study examined the role and the underlying mechanism of ALO in colorectal cancer (CRC). CRC cells were processed by different concentrations of ALO, and subsequently the cell proliferation was detected by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and miR-296-5p expression was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Moreover, the target gene of miR-296-5p was predicted by TargetScan and confirmed by dual-luciferase reporter assay. The expressions of signal transducer and activator of transcription 3 (STAT3), apoptosis-related proteins and epithelial-mesenchymal transition (EMT)-related markers were measured by Western blot. Clone formation assay, flow cytometry, wound-healing and Transwell assays were respectively employed to detect cell proliferation, apoptosis, migration and invasion. ALO inhibited CRC cell proliferation in a dose-dependent manner. MiR-296-5p was low-expressed in CRC tissues and cells, and ALO promoted miR-296-5p expression. STAT3 was targeted by miR-296-5p. Up-regulation of miR-296-5p and ALO treatment both suppressed STAT3 expression, inhibited CRC cell proliferation, migration, invasion as well as the expressions of Bcl-2 and N-cadherin, but promoted apoptosis and expressions of Bax and E-cadherin, which were all reversed by overexpressed STAT3. ALO inhibited CRC cell proliferation, metastasis and EMT but promoted apoptosis via regulating miR-296-5p/STAT3 axis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Quinolizidinas/farmacologia , RNA Neoplásico/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células HCT116 , Células HT29 , Humanos , MicroRNAs/genética , Metástase Neoplásica , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Fator de Transcrição STAT3/genética
15.
Chin J Nat Med ; 19(11): 815-824, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34844720

RESUMO

Cervical cancer (CC) is recognized as the most common neoplasm in the female reproductive system worldwide. The lack of chemotherapeutic agents with outstanding effectiveness and safety severely compromises the anti-cipated prognosis of patients. Aloperine (ALO) is a natural quinolizidine alkaloid with marked anti-cancer effects on multiple malignancies as well as favorable activity in relieving inflammation, allergies and infection. However, its therapeutic efficacy and underlying mechanism in CC are still unclear. In the current study, MTT assay was employed to evaluate the viability of HeLa cells exposed to ALO to preliminarily estimate the effectiveness of ALO in CC. Then, the effects of ALO on the proliferation and apoptosis of HeLa cells were further investigated by plate colony formation and flow cytometry, respectively, while the migration and invasion of ALO-treated HeLa cells were evaluated using Transwell assay. Moreover, nude mice were subcutaneously inoculated with HeLa cells to demonstrate the anti-CC properties of ALO in vivo. The molecular mechanisms underlying these effects of ALO were evaluated by Western blot and immunohistochemical analysis. This study experimentally demonstrated that ALO inhibited the proliferation of HeLa cells via G2 phase cell cycle arrest. Simultaneously, ALO promoted an increase in the percentage of apoptotic HeLa cells by increasing the Bax/Bcl-2 ratio. Additionally, the migration and invasion of HeLa cells were attenuated by ALO treatment, which was considered to result from inhibition of epithelial-to-mesenchymal transition. For molecular mechanisms, the expression and activation of the IL-6-JAK1-STAT3 feedback loop were markedly suppressed by ALO treatment. This study indicated that ALO markedly suppresses the proliferation, migration and invasion and enhances the apoptosis of HeLa cells. In addition, these prominent anti-CC properties of ALO are associated with repression of the IL-6-JAK1-STAT3 feedback loop.


Assuntos
Quinolizidinas/farmacologia , Neoplasias do Colo do Útero , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Retroalimentação , Feminino , Células HeLa , Humanos , Interleucina-6/genética , Janus Quinase 1/genética , Camundongos , Camundongos Nus , Fator de Transcrição STAT3/genética , Transdução de Sinais , Neoplasias do Colo do Útero/tratamento farmacológico
16.
Bioorg Chem ; 117: 105432, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34678602

RESUMO

Twenty-nine 12 N-substituted aloperine derivatives were synthesized and screened for suppression on PD-L1 expression in H460 cells, as a continuation of our work. Systematic structural modifications led to the identification of compound 6b as the most active PD-L1 modulator. Compound 6b could significantly down-regulate both constitutive and inductive PD-L1 expression in NSCLC cells, and successively enhance the cytotoxicity of co-cultured T cells against tumor cells at the concentration of 20 µM. Also, it exhibited a moderate in vivo anticancer efficacy against Lewis tumor xenograft with a stable PK and safety profile. The mechanism study indicated that 6b mediated the degradation of PD-L1 through a proteasome pathway, rather than a lysosome route. These results provided the powerful information for cancer immunotherapy of aloperine derivatives with unique endocyclic skeleton by targeting PD-L1 to activate immune cells to kill cancer cells.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Regulação para Baixo/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Quinolizidinas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Inibidores de Checkpoint Imunológico/síntese química , Inibidores de Checkpoint Imunológico/química , Camundongos , Camundongos Endogâmicos , Estrutura Molecular , Quinolizidinas/síntese química , Quinolizidinas/química , Relação Estrutura-Atividade
17.
Bioorg Chem ; 115: 105196, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34333425

RESUMO

So far, there is still no specific drug against COVID-19. Taking compound 1 with anti-EBOV activity as the lead, fifty-four 12N-substituted aloperine derivatives were synthesized and evaluated for the anti-SARS-CoV-2 activities using pseudotyped virus model. Among them, 8a exhibited the most potential effects against both pseudotyped and authentic SARS-CoV-2, as well as SARS-CoV and MERS-CoV, indicating a broad-spectrum anti-coronavirus profile. The mechanism study disclosed that 8a might block a late stage of viral entry, mainly via inhibiting host cathepsin B activity rather than directly targeting cathepsin B protein. Also, 8a could significantly reduce the release of multiple inflammatory cytokines in a time- and dose-dependent manner, such as IL-6, IL-1ß, IL-8 and MCP-1, the major contributors to cytokine storm. Therefore, 8a is a promising agent with the advantages of broad-spectrum anti-coronavirus and anti-cytokine effects, thus worthy of further investigation.


Assuntos
Antivirais/farmacologia , Piperidinas/farmacologia , Quinolizidinas/farmacologia , SARS-CoV-2/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/síntese química , Antivirais/farmacocinética , Antivirais/toxicidade , Catepsina B/antagonistas & inibidores , Chlorocebus aethiops , Citocinas/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Piperidinas/síntese química , Piperidinas/farmacocinética , Piperidinas/toxicidade , Quinolizidinas/síntese química , Quinolizidinas/farmacocinética , Quinolizidinas/toxicidade , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Células Vero
18.
Int Immunopharmacol ; 97: 107720, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33945918

RESUMO

Presently, postmenopausal osteoporosis mainly caused by excessive activation of in vivo osteoclasts has become a global public health burden. Natural compounds have gradually become the potential drugs for the treatment of postmenopausal osteoporosis. Aloperine is a new alkaloid extracted from the leaves and seeds of sophora bean. The current studies have proved that aloperine has many biological activities, including anti-inflammatory, antiviral and anticancer activities. This study shows that aloperine can inhibit activity and formation of osteoclast mediated by RANKL in a dose-dependent manner without affecting the activity of bone marrow macrophages (BMM). In addition, it is found that aloperine can inhibit the expression of osteoclast specific marker genes, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), tartrate resistant acid phosphatase (TRAcP), matrix metallopeptidase 9 (MMP9), cathepsin K (Ctsk), V-ATPase d2 and calcitonin receptor. The in vitro experiment of aloperine proved that aloperine can inhibit the degradation of IκBα and the phosphorylation of P65, ERK and JNK. Additionally, aloperine improves bone loss in ovariectomized (OVX) mice by inhibiting osteoclast activity. This project proved that aloperine can affect the formation of osteoclasts by inhibiting RANKL signaling channel, and it is indicated that aloperine has the potential to be developed as a new drug for the prevention and treatment of postmenopausal osteoporosis.


Assuntos
Osteogênese/efeitos dos fármacos , Osteoporose Pós-Menopausa/tratamento farmacológico , Quinolizidinas/farmacologia , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Fêmur/efeitos dos fármacos , Fêmur/patologia , Humanos , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoporose Pós-Menopausa/diagnóstico , Osteoporose Pós-Menopausa/etiologia , Osteoporose Pós-Menopausa/patologia , Ovariectomia , Quinolizidinas/uso terapêutico , Ligante RANK/antagonistas & inibidores , Ligante RANK/metabolismo , Microtomografia por Raio-X
19.
Biomolecules ; 11(3)2021 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805605

RESUMO

Cystic fibrosis is a monogenic, autosomal, recessive disease characterized by an alteration of chloride transport caused by mutations in the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene. The loss of Phe residue in position 508 (ΔF508-CFTR) causes an incorrect folding of the protein causing its degradation and electrolyte imbalance. CF patients are extremely predisposed to the development of a chronic inflammatory process of the bronchopulmonary system. When the cells of a tissue are damaged, the immune cells are activated and trigger the production of free radicals, provoking an inflammatory process. In addition to routine therapies, today drugs called correctors are available for mutations such as ΔF508-CFTR as well as for others less frequent ones. These active molecules are supposed to facilitate the maturation of the mutant CFTR protein, allowing it to reach the apical membrane of the epithelial cell. Matrine induces ΔF508-CFTR release from the endoplasmic reticulum to cell cytosol and its localization on the cell membrane. We now have evidence that Matrine and Lumacaftor not only restore the transport of mutant CFTR protein, but probably also counteract the inflammatory process by improving the course of the disease.


Assuntos
Alcaloides/uso terapêutico , Aminopiridinas/uso terapêutico , Benzodioxóis/uso terapêutico , Fibrose Cística/tratamento farmacológico , Inflamação/patologia , Quinolizinas/uso terapêutico , Células A549 , Alcaloides/farmacologia , Aminopiridinas/farmacologia , Benzodioxóis/farmacologia , Morte Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Quinolizidinas/farmacologia , Quinolizinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Matrinas
20.
Drug Des Devel Ther ; 15: 857-870, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664565

RESUMO

BACKGROUND: Aloperine can regulate miR-296-5p/Signal Transducer and Activator of Transcription 3 (STAT3) pathway to inhibit the malignant development of colorectal cancer (CRC), but the regulatory mechanism is unclear. This study explored the upstream mechanism of Aloperine in reducing CRC damage from the perspective of the circRNA-miRNA-mRNA regulatory network. METHODS: After treatment with gradient concentrations of Aloperine (0.1 mmol/L, 0.2 mmol/L, 0.4 mmol/L, 0.8 mmol/L and 1 mmol/L) for 24 hours, changes in CRC cell proliferation and apoptosis were detected by functional experiments. Data of the differential expression of miR-296-5p in CRC patients and healthy people were obtained from Starbase. The effects of Aloperine on 12 differentially expressed circRNAs were detected. The binding of miR-296-5p with NOP2/Sun RNA methyltransferase 2 (circNSUN2) and STAT3 was predicted by TargetScan and confirmed through dual-luciferase experiments. The expressions of circNSUN2, miR-296-5p and STAT3 as well as apoptosis-related genes in CRC cells were detected by qRT-PCR and Western blot as needed. Rescue experiments were conducted to test the regulatory effects of circNSUN2, miR-296-5p and STAT3 on CRC cells. RESULTS: Aloperine at a concentration gradient inhibited proliferation and promoted apoptosis in CRC cells. The abnormally low expression of miR-296-5p in CRC could be upregulated by Aloperine. Among the differentially expressed circRNAs in CRC, only circNSUN2 not only targets miR-296-5p, but also can be regulated by Aloperine. The up-regulation of circNSUN2 offset the inhibitory effect of Aloperine on cancer cells. The rescue experiments finally confirmed the regulation of circNSUN2/miR-296-5p/STAT3 axis in CRC cells. CONCLUSION: By regulating the circNSUN2/miR-296-5p/STAT3 pathway, Aloperine prevents the malignant development of CRC cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Quinolizidinas/farmacologia , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Biologia Computacional , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Metiltransferases/genética , Metiltransferases/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Estrutura Molecular , Quinolizidinas/química , RNA Circular/genética , RNA Circular/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA