Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Bioorg Med Chem ; 61: 116740, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35396128

RESUMO

Human African trypanosomiasis is caused by a protozoan parasite Trypanosoma brucei majorly infecting people living in sub-Saharan Africa. Current limited available treatments suffer from drug resistance, severe adverse effects, low efficacy, and costly administrative procedures in African countries with limited medical resources. Therefore, there is always a perpetual demand for advanced drug development and invention of new strategies to combat the disease. Previous work in our lab generated a library of sulfonamide analogs as selective tubulin inhibitors, based on the structural difference between mammalian and trypanosome tubulin proteins. Further lead derivatization was performed in the current study and generated 25 potential drug candidates to improve the drug efficacy and uptake by selectively targeting the parasite's P2 membrane transporter protein with imidamide moiety. One of the newly synthesized analogs, compound 25 with a di-imidamide moiety, has shown greater potency with an IC50 of 1 nM to selectively inhibit the growth of trypanosome cells without affecting the viability of mammalian cells. Western blot analyses reveal that the compound suppressed tubulin polymerization in T. brucei cells. A detailed structure-activity relationship (SAR) was summarized that will be used to guide future lead optimization.


Assuntos
Tripanossomicidas , Trypanosoma brucei brucei , Tripanossomíase Africana , Animais , Humanos , Mamíferos/metabolismo , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/parasitologia , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacologia
2.
Bioorg Med Chem ; 25(12): 3215-3222, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28428042

RESUMO

African trypanosomiasis is still a threat to human health due to the severe side-effects of current drugs. We identified selective tubulin inhibitors that showed the promise to the treatment of this disease, which was based on the tubulin protein structural difference between mammalian and trypanosome cells. Further lead optimization was performed in the current study to improve the efficiency of the drug candidates. We used Trypanosoma brucei brucei cells as the parasite model, and human normal kidney cells and mouse macrophage cells as the host model to evaluate the compounds. One new analog showed great potency with an IC50 of 70nM to inhibit the growth of trypanosome cells and did not affect the viability of mammalian cells. Western blot analyses reveal that the compound decreased tubulin polymerization in T. brucei cells. A detailed structure activity relationship (SAR) was summarized that will be used to guide future lead optimization.


Assuntos
Tripanossomicidas/química , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Tripanossomíase Africana/tratamento farmacológico , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Animais , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/metabolismo , Tripanossomicidas/síntese química , Trypanosoma brucei brucei/metabolismo , Tripanossomíase Africana/parasitologia , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntese química
3.
J Med Chem ; 64(3): 1570-1583, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33523674

RESUMO

Androgen receptor (AR) contributes to the progression of glioblastoma (GBM), and antiandrogen agents have the potential to be used for the treatment of GBM. However, AR mutation commonly happens in GBM, which makes the antiandrogen agents less effective. Heat shock 27 kDa protein (HSP27) is a well-documented chaperone protein to stabilize ARs. Inhibition of HSP27 results in AR degradation regardless of the mutation status of ARs, which makes HSP27 a good target to abolish ARs in GBM. Compound I is a HSP27 inhibitor that significantly induces AR degradation in GBM cells via the proteasomal pathway, and it selectively inhibits AR-overexpressed GBM cell growth with IC50 values around 5 nM. The compound also significantly inhibits in vivo GBM xenograft at 20 mg/kg and does not cause toxicity to mice up to 80 mg/kg. These results suggest that targeting HSP27 to induce AR degradation in GBM is a promising and novel treatment.


Assuntos
Antagonistas de Androgênios/farmacologia , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas de Choque Térmico HSP27/antagonistas & inibidores , Receptores Androgênicos/efeitos dos fármacos , Antagonistas de Androgênios/toxicidade , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , Mutação , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Receptores Androgênicos/genética , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Eur J Med Chem ; 143: 1261-1276, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29128116

RESUMO

Ligand-independent activation of EphA2 receptor kinase promotes cancer metastasis and invasion. Activating EphA2 receptor tyrosine kinase with small molecule agonist is a novel strategy to treat EphA2 overexpressing cancer. In this study, we performed a lead optimization of a small molecule Doxazosin that was identified as an EphA2 receptor agonist. 33 new analogs were developed and evaluated; a structure-activity relationship was summarized based on the EphA2 activation of these derivatives. Two new derivative compounds 24 and 27 showed much improved activity compared to Doxazosin. Compound 24 possesses a bulky amide moiety, and compound 27 has a dimeric structure that is very different to the parental compound. Compound 27 with a twelve-carbon linker of the dimer activated the kinase and induced receptor internalization and cell death with the best potency. Another dimer with a six-carbon linker has significantly reduced potency compared to the dimer with a longer linker, suggesting that the length of the linker is critical for the activity of the dimeric agonist. To explore the receptor binding characteristics of the new molecules, we applied a docking study to examine how the small molecule binds to the EphA2 receptor. The results reveal that compounds 24 and 27 form more hydrogen bonds to EphA2 than Doxazosin, suggesting that they may have higher binding affinity to the receptor.


Assuntos
Doxazossina/farmacologia , Desenho de Fármacos , Efrina-A2/agonistas , Bibliotecas de Moléculas Pequenas/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Doxazossina/síntese química , Doxazossina/química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Receptor EphA2 , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
5.
J Steroid Biochem Mol Biol ; 168: 91-101, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28213333

RESUMO

Hexamethylene bis-acetamide inducible protein 1 (HEXIM1) is identified as a novel inhibitor of estrogen stimulated breast cell growth, and it suppresses estrogen receptor-α transcriptional activity. HEXIM1 protein level has been found to be downregulated by estrogens. Recently, HEXIM1 has been found to inhibit androgen receptor transcriptional activity as well. Researchers have used Hexamethylene bis-acetamide (HMBA) for decades to stimulate HEXIM1 expression, which also inhibit estrogen stimulated breast cancer cell gene activation and androgen stimulated prostate cancer gene activation. However, the direct molecular targets of HMBA that modulate the induction of HEXIM1 expression in mammalian cells have not been identified. Based on HMBA and its more potent analog 4a1, we designed molecular probes to pull down the binding proteins of these compounds. Via proteomic approach and biological assays, we demonstrate that HMBA and 4a1 are actually heat shock protein 70 (HSP70) binders. The known HSP70 activator showed similar activity as HMBA and 4a1 to induce HEXIM1 expression, suggesting that HMBA and 4a1 might be putative HSP70 activators. Molecular target identification of HMBA and 4a1 could lead to further structural optimization of the parental compound to generate more potent derivatives to stimulate HEXIM1 expression, which could be a novel approach for hormone dependent breast cancer and prostate cancer treatment.


Assuntos
Acetamidas/química , Benzenoacetamidas/química , Estrogênios/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores de Estrogênio/metabolismo , Biotinilação , Neoplasias da Mama/tratamento farmacológico , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Espectrometria de Massas , Neoplasias da Próstata/tratamento farmacológico , Ligação Proteica , Proteômica , Espectrometria de Massas por Ionização por Electrospray , Fatores de Transcrição
6.
PLoS One ; 11(1): e0146289, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26771307

RESUMO

OBJECTIVES: There is an urgent need to develop a safe, effective, orally active, and inexpensive therapy for African trypanosomiasis due to the drawbacks of current drugs. Selective tubulin inhibitors have the potential to be promising drug candidates for the treatment of this disease, which is based on the tubulin protein structural difference between mammalian and trypanosome cells. We propose to identify novel tubulin inhibitors from a compound library developed based on the lead compounds that selectively target trypanosomiasis. METHODS: We used Trypanosoma brucei brucei as the parasite model, and human normal kidney cells and mouse microphage cells as the host model. Growth rates of both trypanosomes and mammalian cells were determined as a means to screen compounds that selectively inhibit the proliferation of parasites. Furthermore, we examined the cell cycle profile of the parasite and compared tubulin polymerization dynamics before and after the treatment using identified compounds. Last, in vivo anti-parasite activities of these compounds were determined in T. brucei-infected mice. RESULTS: Three compounds were selected that are 100 fold more effective against the growth of T. brucei cells than mammalian cells. These compounds caused cell cycle progression defects in T. brucei cells. Western analyses indicated that these compounds decreased tubulin polymerization in T. brucei cells. The in vivo investigation revealed that these compounds, when admitted orally, inhibited T. brucei cell proliferation in mouse blood. However, they were not potent enough to clear up the infection completely. CONCLUSIONS: These compounds are promising lead compounds as orally active agents for drug development of anti-trypanosome agents. A more detail structure activity relationship (SAR) was summarized that will be used to guide future lead optimization to improve the selectivity and potency of the current compounds.


Assuntos
Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Moduladores de Tubulina/farmacologia , Administração Oral , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Camundongos , Proteínas de Protozoários/metabolismo , Relação Estrutura-Atividade , Tripanossomicidas/efeitos adversos , Tripanossomíase Africana/tratamento farmacológico , Moduladores de Tubulina/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA