Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Immunol ; 20(11): 1352-1366, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37752225

RESUMO

Thiostrepton (TST) is a natural antibiotic with pleiotropic properties. This study aimed to elucidate the therapeutic effect of TST on experimental colitis and identify its targets. The effect of TST on colon inflammation was evaluated in a dextran sulfate sodium (DSS)-induced colitis model and a T-cell transfer colitis model. The therapeutic targets of TST were investigated by cytokine profiling, immunophenotyping and biochemical approaches. The effect of TST on the gut microbiota and its contribution to colitis were evaluated in mice with DSS-induced colitis that were subjected to gut microbiota depletion and fecal microbiota transplantation (FMT). Alterations in the gut microbiota caused by TST were determined by 16S rDNA and metagenomic sequencing. Here, we showed that TST treatment significantly ameliorated colitis in the DSS-induced and T-cell transfer models. Specifically, TST targeted the retinoic acid-related orphan nuclear receptor RORγt to reduce the production of IL-17A by γδ T cells, type 3 innate lymphoid cells (ILC3s) and Th17 cells in mice with DSS-induced colitis. Similarly, TST selectively prevented the development of Th17 cells in the T-cell transfer colitis model and the differentiation of naïve CD4+ T cells into Th17 cells in vitro. Mechanistically, TST induced the ubiquitination and degradation of RORγt by promoting the binding of Itch to RORγt. Moreover, TST also reversed dysbiosis to control colonic inflammation. Taken together, these results from our study describe the previously unexplored role of TST in alleviating colonic inflammation by reducing IL-17A production and modulating dysbiosis, suggesting that TST is a promising candidate drug for the treatment of IBD.


Assuntos
Colite , Interleucina-17 , Animais , Camundongos , Interleucina-17/metabolismo , Tioestreptona/metabolismo , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico , Imunidade Inata , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Disbiose , Linfócitos/metabolismo , Colite/induzido quimicamente , Colite/terapia , Colo/metabolismo , Inflamação/metabolismo , Ubiquitinação , Sulfato de Dextrana , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
2.
Mol Biol Rep ; 49(11): 10387-10397, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36097108

RESUMO

OBJECTIVE: FoxM1 transcription factor contributes to tumor metastasis and poor prognosis in many cancers including triple-negative breast cancer (TNBC). In this study, we examined the effects of FoxM1 inhibitor Thiostrepton (THIO) alone or in combination with MEK inhibitor Selumetinib (SEL) on metastatic parameters in vitro and in vivo. METHODS: Cell viability was determined by MTT assay. Immunoblotting and immunohistochemistry was used to assess metastasis-related protein expressions in 4T1 cells and its allograft tumor model in BALB/c mice. In vivo uPA activity was determined by enzymatic methods. RESULTS: Both inhibitors were effective on the expressions of FoxM1, ERK, p-ERK, Twist, E-cadherin, and Vimentin alone or in combination in vitro. THIO significantly decreased 4T1 cell migration and changed the cell morphology from mesenchymal-like to epithelial-like structure. THIO was more effective than in combination with SEL in terms of metastatic protein expressions in vivo. THIO alone significantly inhibited mean tumor growth, decreased lung metastasis rate and tumor foci, however, no significant changes in these parameters were observed in the combined group. Immunohistochemically, FoxM1 expression intensity was decreased with THIO and its combination with SEL in the tumors. CONCLUSIONS: This study suggests that inhibiting FoxM1 as a single target is more effective than combined treatment with MEK in theTNBC allograft model. The therapeutic efficacy of THIO should be investigated with further studies on appropriate drug delivery systems.


Assuntos
Tioestreptona , Neoplasias de Mama Triplo Negativas , Humanos , Camundongos , Animais , Tioestreptona/farmacologia , Tioestreptona/química , Tioestreptona/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Proteína Forkhead Box M1/genética , Linhagem Celular Tumoral , Proliferação de Células , Quinases de Proteína Quinase Ativadas por Mitógeno
3.
Free Radic Biol Med ; 193(Pt 1): 385-404, 2022 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-36152915

RESUMO

Gastric cancer is a leading cause of tumor-associated death worldwide. Metastasis and chemoresistance are crucial barriers for gastric cancer treatment. The Forkhead Box M1 (FOXM1) transcription factor has been reported as a promising treatment target for various types of tumors, but its effects on gastric cancer progression are not fully understood. In the present study, we found that FOXM1 expression levels were significantly up-regulated in human gastric cancer cell lines and tissues, and its expression was much higher in patients with metastasis. We then found that suppressing FOXM1 with its inhibitor thiostrepton (THIO) significantly reduced the proliferation of gastric cancer cells, while induced G0/G1 and apoptosis. Moreover, reactive oxygen species (ROS) production, mitochondrial impair and autophagy were remarkably provoked in gastric cancer cells treated with THIO, which were required for the regulation of apoptotic cell death. Furthermore, THIO exposure considerably suppressed the migration, invasion and angiogenesis in gastric cancer cells. The inhibitory effects of THIO on tumor growth and metastasis were confirmed in an established gastric cancer xenograft mouse model without detectable toxicity. Intriguingly, our in vitro studies showed that the anti-cancer effects of THIO on gastric cancer were almost abolished upon FOXM1 over-expression, indicating the necessity of FOXM1 suppression in THIO-inhibited tumor growth. In addition, higher FOXM1 expression was detected in gastric cancer cells with chemoresistance. Both in vitro and in vivo studies illustrated that THIO strongly promoted the drug-resistant gastric cancer cells to chemotherapies, proved by the considerably decreased cell proliferation and epithelial-mesenchymal transition (EMT) process. Together, these findings revealed that FOXM1 was a promising therapeutic target for gastric cancer treatment, and THIO exerted potential as an therapeutic agent for the disease.


Assuntos
Neoplasias Gástricas , Tioestreptona , Animais , Humanos , Camundongos , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico
4.
Adv Sci (Weinh) ; 9(29): e2202702, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35975458

RESUMO

Programmed death-ligand 1 (PD-L1) is a major target to cancer immunotherapy, and anti-PD-L1 and anti-PD-1 antibody-mediated immunotherapy are being increasingly used. However, immune checkpoint inhibitors (ICIs) are ineffective in treating large tumors and cause various immune-related adverse events in nontarget organs, including life-threatening cardiotoxicity. Therefore, the development of new therapeutic strategies to overcome these limitations is crucial. The focus of this study is the forkhead box protein M1 (FOXM1), which is identified as a potential therapeutic target for cancer immunotherapy and is associated with the modulation of PD-L1 expression. Selective small interfering RNA knockdown of FOXM1 or treatment with thiostrepton (TST) significantly reduces PD-L1 expression in non-small-cell lung cancer (NSCLC) cells and inhibits proliferation. Chromatin immunoprecipitation-PCR reveals that FOXM1 selectively upregulates PD-L1 expression by binding directly to the PD-L1 promoter. In vivo animal studies have shown that TST treatment significantly downregulates PD-L1 expression in human NSCLC tumors, while greatly reducing tumor size without side effects on normal tissues. Combined treatment with TST and anti-4-1BB antibody in the LLC-1 syngeneic tumor model induces synergistic therapeutic outcomes against immune resistant lung tumors as well as 2.72-folds higher CD3+ T cells in tumor tissues compared to that in the anti-4-1BB antibody treatment group.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Antígeno B7-H1 , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/uso terapêutico , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Receptor de Morte Celular Programada 1 , RNA Interferente Pequeno/uso terapêutico , Tioestreptona/uso terapêutico , Resultado do Tratamento
5.
Front Cell Infect Microbiol ; 12: 907043, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35873171

RESUMO

Trypanosoma cruzi, the causal agent of Chagas disease, has peroxiredoxins (PRXs) expressed in all stages of the parasite and whose function is to detoxify oxidizing agents, such as reactive oxygen species (ROS). These proteins are central for the survival and replication of the parasite and have been proposed as virulence factors. Because of their importance, they have also been considered as possible therapeutic targets, although there is no specific drug against them. One of them, the mitochondrial PRX (TcMPX), is important in the detoxification of ROS in this organelle and has a role in the infectivity of T. cruzi. However, their structural characteristics are unknown, and possible inhibitors have not been proposed. The aim was to describe in detail some structural characteristics of TcMPX and compare it with several PRXs to find possible similarities and repositioning the antibiotic Thiostrepton as a potential inhibitor molecule. It was found that, in addition to the characteristic active site of a 2-cys PRX, this protein has a possible transmembrane motif and motifs involved in resistance to hyper oxidation. The homology model suggests a high structural similarity with human PRX3. This similarity was corroborated by cross-recognition using an anti-human PRX antibody. In addition, molecular docking showed that Thiostrepton, a potent inhibitor of human PRX3, could bind to TcMPX and affect its function. Our results show that Thiostrepton reduces the proliferation of T. cruzi epimastigotes, cell-derived trypomastigotes, and blood trypomastigotes with low cytotoxicity on Vero cells. We also demonstrated a synergic effect of Thriostepton and Beznidazol. The convenience of seeking treatment alternatives against T. cruzi by repositioning compounds as Thiostrepton is discussed.


Assuntos
Doença de Chagas , Trypanosoma cruzi , Animais , Doença de Chagas/tratamento farmacológico , Doença de Chagas/parasitologia , Chlorocebus aethiops , Humanos , Simulação de Acoplamento Molecular , Peroxirredoxina III/metabolismo , Peroxirredoxina III/farmacologia , Peroxirredoxina III/uso terapêutico , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Tioestreptona/metabolismo , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico , Trypanosoma cruzi/metabolismo , Células Vero
6.
Acta Pharmacol Sin ; 43(11): 2956-2966, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35422085

RESUMO

The forkhead box M1 (FoxM1) protein, a transcription factor, plays critical roles in regulating tumor growth and drug resistance, while cellular FLICE-inhibitory protein (c-FLIP), an anti-apoptotic regulator, is involved in the ubiquitin-proteasome pathway. In this study, we investigated the effects of c-FLIP on the expression and ubiquitination levels of FoxM1 along with drug susceptibility in non-small-cell lung cancer (NSCLC) cells. We first showed that the expression levels of FoxM1 and c-FLIP were increased and positively correlated (R2 = 0.1106, P < 0.0001) in 90 NSCLC samples. The survival data from prognostic analysis demonstrated that high expression of c-FLIP and/or FoxM1 was related to poor prognosis in NSCLC patients and that the combination of FoxM1 and c-FLIP could be a more precise prognostic biomarker than either alone. Then, we explored the functions of c-FLIP/FoxM1 in drug resistance in NSCLC cell lines and a xenograft mouse model in vivo. We showed that c-FLIP stabilized FoxM1 by inhibiting its ubiquitination, thus upregulated the expression of FoxM1 at post-transcriptional level. In addition, a positive feedback loop composed of FoxM1, ß-catenin and p65 also participated in c-FLIP-FoxM1 axis. We revealed that c-FLIP promoted the resistance of NSCLC cells to thiostrepton and osimertinib by upregulating FoxM1. Taken together, these results reveal a new mechanism by which c-FLIP regulates FoxM1 and the function of this interaction in the development of thiostrepton and osimertinib resistance. This study provides experimental evidence for the potential therapeutic benefit of targeting the c-FLIP-FoxM1 axis for lung cancer treatment.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Carcinoma Pulmonar de Células não Pequenas , Resistencia a Medicamentos Antineoplásicos , Proteína Forkhead Box M1 , Animais , Humanos , Camundongos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico , Tioestreptona/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética
7.
Cancer Res ; 80(24): 5554-5568, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33087324

RESUMO

Peritoneal spread is the primary mechanism of metastasis of ovarian cancer, and survival of ovarian cancer cells in the peritoneal cavity as nonadherent spheroids and their adherence to the mesothelium of distant organs lead to cancer progression, metastasis, and mortality. However, the mechanisms that govern this metastatic process in ovarian cancer cells remain poorly understood. In this study, we cultured ovarian cancer cell lines in adherent and nonadherent conditions in vitro and analyzed changes in mRNA and protein levels to identify mechanisms of tumor cell survival and proliferation in adherent and nonadherent cells. EGFR or ERBB2 upregulated ZEB1 in nonadherent cells, which caused resistance to cell death and increased tumor-initiating capacity. Conversely, Forkhead box M1 (FOXM1) was required for the induction of integrin ß1, integrin-α V, and integrin-α 5 for adhesion of cancer cells. FOXM1 also upregulated ZEB1, which could act as a feedback inhibitor of FOXM1, and caused the transition of adherent cells to nonadherent cells. Strikingly, the combinatorial treatment with lapatinib [dual kinase inhibitor of EGFR (ERBB1) and ERBB2] and thiostrepton (FOXM1 inhibitor) reduced growth and peritoneal spread of ovarian cancer cells more effectively than either single-agent treatment in vivo. In conclusion, these results demonstrate that FOXM1 and EGFR/ERBB2 pathways are key points of vulnerability for therapy to disrupt peritoneal spread and adhesion of ovarian cancer cells. SIGNIFICANCE: This study describes the mechanism exhibited by ovarian cancer cells required for adherent cell transition to nonadherent form during peritoneal spread and metastasis. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/24/5554/F1.large.jpg.


Assuntos
Receptores ErbB/metabolismo , Proteína Forkhead Box M1/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Receptor ErbB-2/metabolismo , Transdução de Sinais/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Feminino , Proteína Forkhead Box M1/antagonistas & inibidores , Proteína Forkhead Box M1/genética , Técnicas de Silenciamento de Genes , Humanos , Lapatinib/farmacologia , Lapatinib/uso terapêutico , Camundongos , Neoplasias Peritoneais/prevenção & controle , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Transdução de Sinais/efeitos dos fármacos , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico , Transfecção
8.
Adv Mater ; 32(17): e2000964, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32162422

RESUMO

Fluorescent proteins are investigated extensively as markers for the imaging of cells and tissues that are treated by gene transfection. However, limited transfection efficiency and lack of targeting restrict the clinical application of this method rooted in the challenging development of robust fluorescent proteins for in vivo bioimaging. To address this, a new type of near-infrared (NIR) fluorescent protein assemblies manufactured by genetic engineering is presented. Due to the formation of well-defined nanoparticles and spectral operation within the phototherapeutic window, the NIR protein aggregates allow stable and specific tumor imaging via simple exogenous injection. Importantly, in vivo tumor metastases are tracked and this overcomes the limitations of in vivo imaging that can only be implemented relying on the gene transfection of fluorescent proteins. Concomitantly, the efficient loading of hydrophobic drugs into the protein nanoparticles is demonstrated facilitating the therapy of tumors in a mouse model. It is believed that these theranostic NIR fluorescent protein assemblies, hence, show great potential for the in vivo detection and therapy of cancer.


Assuntos
Antineoplásicos/química , Engenharia Genética , Proteínas Luminescentes/química , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Portadores de Fármacos/química , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Peptídeos/química , Polietilenoglicóis/química , Espectroscopia de Luz Próxima ao Infravermelho , Tioestreptona/química , Tioestreptona/metabolismo , Tioestreptona/uso terapêutico , Transplante Heterólogo
9.
Molecules ; 24(24)2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31835481

RESUMO

Mycobacterium abscessus is a rapid-growing, multidrug-resistant, non-tuberculous mycobacterial species responsible for a variety of human infections, such as cutaneous and pulmonary infections. M. abscessus infections are very difficult to eradicate due to the natural and acquired multidrug resistance profiles of M. abscessus. Thus, there is an urgent need for the development of effective drugs or regimens against M. abscessus infections. Here, we report the activity of a US Food and Drug Administration approved drug, thiostrepton, against M. abscessus. We found that thiostrepton significantly inhibited the growth of M. abscessus wild-type strains, subspecies, clinical isolates, and drug-resistant mutants in vitro and in macrophages. In addition, treatment of macrophages with thiostrepton significantly decreased proinflammatory cytokine production in a dose-dependent manner, suggesting an inhibitory effect of thiostrepton on inflammation induced during M. abscessus infection. We further showed that thiostrepton exhibits antimicrobial effects in vivo using a zebrafish model of M. abscessus infection.


Assuntos
Antibacterianos/farmacologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/efeitos dos fármacos , Tioestreptona/farmacologia , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Linhagem Celular , Citocinas/biossíntese , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Testes de Sensibilidade Microbiana , Estrutura Molecular , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/metabolismo , Mycobacterium abscessus/classificação , Mycobacterium abscessus/genética , Tioestreptona/uso terapêutico , Peixe-Zebra
10.
Anticancer Agents Med Chem ; 19(3): 323-329, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29308746

RESUMO

BACKGROUND/OBJECTIVE: Forkhead Box M1 (FOXM1) is frequently activated in tumors. We studied the expression and the possible mechanism of FOXM1 and evaluated the effects of thiostrepton in an endometriotic rat model. METHODS AND MATERIAL: This was a randomized study in a rat model of endometriosis. Fifty female Wistar rats were surgically induced with endometriosis. After 4 weeks of observation, twenty and thirty rats were randomly allocated to an ovariectomized (OVX) group and a treatment group, respectively. The OVX group was ovariectomized and randomly divided into an OVX-estrogen group and a control (OVX -oil) group. All rats were allowed a resting period of 3 days prior to any operation. The rats in the estrogen group were given estradiol (20 µg/kg, 0.1 ml /d), while the control group was treated with an equivalent amount of sesame oil. Every group was injected with subcutaneous injection for 7 days. The treatment group was randomly divided into three groups to receive the following: TST at 150 mg/kg, ip.; TST at 250 mg/kg, ip.; or sterile normal saline, ip. The groups received these dosages every 2 days for 2 weeks. Lesion growth, histological examination, and protein expression were subsequently analyzed using caliper measurement, histology, immunostaining, and Western blot after each rat received an injection in its own group. RESULTS: Our results showed that FOXM1 is enriched in nucleus of an ectopic endometrium when compared with a eutopic uterus. Furthermore, we found that an ERK/FOXM1/matrix metalloproteinase-9 (MMP9) signaling pathway might result in the establishment and development of endometriosis. Finally, a thiostrepton concentration dependently reduced the expression of FOXM1, MMP9 and Bcl-2 in endometriotic lesions of the treated rats. Statistical significance was accepted for a value of P < 0.05. CONCLUSION: We postulate that thiostrepton could inhibit the endometriotic lesions, at least in part, by decreasing the FOXM1 expression and exerting a pro-apoptotic effect. We reported for the first time that FOXM1 expresses in experimental endometriosis rat and thiostrepton may also be suitable for the administration of endometriosis by inhibiting the growth of endometriotic implants. More studies are needed to further evaluate thiostrepton's effect.


Assuntos
Endometriose/tratamento farmacológico , Tioestreptona/uso terapêutico , Animais , Relação Dose-Resposta a Droga , Endometriose/metabolismo , Feminino , Proteína Forkhead Box M1/antagonistas & inibidores , Proteína Forkhead Box M1/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Estrutura Molecular , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
11.
J Mol Med (Berl) ; 96(2): 223-235, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29290032

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive vascular remodeling disease characterized by a persistent elevation of pulmonary artery pressure, leading to right heart failure and premature death. Exaggerated proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is a key component of vascular remodeling. Despite major advances in the field, current therapies for PAH remain poorly effective in reversing the disease or significantly improving long-term survival. Because the transcription factor FOXM1 is necessary for PASMC proliferation during lung morphogenesis and its overexpression stimulates proliferation and evasion of apoptosis in cancer cells, we thus hypothesized that upregulation of FOXM1 in PAH-PASMCs promotes cell expansion and vascular remodeling. Our results showed that FOXM1 was markedly increased in distal pulmonary arteries and isolated PASMCs from PAH patients compared to controls as well as in two preclinical models. In vitro, we showed that miR-204 expression regulates FOXM1 levels and that inhibition of FOXM1 reduced cell proliferation and resistance to apoptosis through diminished DNA repair mechanisms and decreased expression of the pro-remodeling factor survivin. Accordingly, inhibition of FOXM1 with thiostrepton significantly improved established PAH in two rat models. Thus, we show for the first time that FOXM1 is implicated in PAH development and represents a new promising target. KEY MESSAGES: FOXM1 is overexpressed in human PAH-PASMCs and PAH animal models. FOXM1 promotes PAH-PASMC proliferation and resistance to apoptosis. Pharmacological inhibition of FOXM1 improves established PAH in the MCT and Su/Hx rat models. FOXM1 may be a novel therapeutic target in PAH.


Assuntos
Proteína Forkhead Box M1/fisiologia , Hipertensão Pulmonar/metabolismo , Miócitos de Músculo Liso/fisiologia , Artéria Pulmonar/fisiologia , Animais , Linhagem Celular , Proliferação de Células , Proteína Forkhead Box M1/antagonistas & inibidores , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/metabolismo , Masculino , MicroRNAs/metabolismo , Artéria Pulmonar/citologia , Ratos Sprague-Dawley , Tioestreptona/uso terapêutico , Remodelação Vascular
12.
BMC Cancer ; 16: 511, 2016 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-27439614

RESUMO

BACKGROUND: Synovial sarcoma (SS) is a soft tissue sarcoma of unknown histogenesis. Most metastatic or unresectable cases are incurable. Novel antitumor agents and precise prognostication are needed for SS patients. The protein forkhead box M1 (FOXM1), which belongs to the FOX family of transcription factors, is considered to be an independent predictor of poor survival in many cancers and sarcomas, but the prognostic implications and oncogenic roles of FOXM1 in SS are poorly understood. Here we examined the correlation between FOXM1 expression and clinicopathologic and prognostic factors, and we investigated the efficacy of FOXM1 target therapy in SS cases. METHODS: Immunohistochemical study of 106 tumor specimens was conducted to evaluate their immunohistochemical expression of FOXM1. An in vitro study examined the antitumor effect of the FOXM1 inhibitor thiostrepton and small interference RNA (siRNA) on two SS cell lines. We also assessed the efficacy of the combined use of doxorubicin (DOX) and thiostrepton. RESULTS: Univariate and multivariate analyses revealed that FOXM1 expression was associated with poor prognosis in SS. The cDNA microarray analysis using clinical samples revealed that the expression of cell cycle-associated genes was correlated with FOXM1 expression. FOXM1 inhibition by thiostrepton showed significant antitumor activity on the SS cell lines in vitro. FOXM1 interruption by siRNA increased the chemosensitivity for DOX in both SS cell lines. CONCLUSION: FOXM1 expression is a novel biomarker, and its inhibition is a potential treatment option for SS.


Assuntos
Antineoplásicos/uso terapêutico , Proteína Forkhead Box M1/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Sarcoma Sinovial/tratamento farmacológico , Adulto , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Doxorrubicina/uso terapêutico , Feminino , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Terapia de Alvo Molecular/métodos , Proteínas de Fusão Oncogênica/genética , Prognóstico , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma Sinovial/genética , Sarcoma Sinovial/metabolismo , Tioestreptona/uso terapêutico , Adulto Jovem
13.
J Comput Aided Mol Des ; 28(12): 1205-15, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25281020

RESUMO

The bacterial ribosome is a major target of naturally occurring thiopeptides antibiotics. Studying thiopeptide (e.g. thiostrepton) binding to the GAR's 23S·L11 ribosomal subunit using docking methods is challenging. Regarding the target, the binding site is composed of a flexible protein-RNA nonbonded interface whose available crystal structure is of medium resolution. Regarding the ligands, the thiopeptides are chemically complex, flexible, and contain macrocycles. In this study we developed a combined MD-docking-MD workflow that allows us to study thiopeptide-23S·L11 binding. It is shown that docking thiostrepton-like ligands to an MD-refined receptor structure instead of the medium resolution crystal leads to better convergence to the native-like docking pose and a better reproduction of experimental binding affinities. By applying an energy decomposition analysis, we identify key structural binding elements within GAR's rRNA-protein binding site and within the ligand structures.


Assuntos
Desenho de Fármacos , RNA Ribossômico/química , Relação Estrutura-Atividade , Tioestreptona/química , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Ligação Proteica , RNA Ribossômico/metabolismo , Ribossomos/química , Ribossomos/metabolismo , Tioestreptona/uso terapêutico
14.
J Cell Mol Med ; 18(5): 811-23, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24628949

RESUMO

Docetaxel is commonly used as an effective chemotherapeutic drug for gastric cancer patients recently. With the increasing emergence of docetaxel resistance nowadays, identification of suitable biomarkers for predicting chemosensitivity to docetaxel may be a key role for improving therapeutic effects for gastric cancer patients. In this study, we investigated the correlation between the expression of transcription factor forkhead box protein M1 (FOXM1) and chemotherapy response to docetaxel in gastric cancer, the possible mechanism for which was further explored. As a result, FOXM1 overexpression was shown to mediate resistance to docetaxel in gastric cancers. It altered microtubule dynamics to protect tumour cells from docetaxel-induced apoptosis. Mechanistic investigations revealed that tubulin-destabilizing protein Stathmin, which mediated docetaxel resistance in FOXM1-silenced gastric cancer cells, is a direct down-stream target of FOXM1, whereas another microtubule dynamics protein mitotic centromere-associated kinesin (MCAK), shown to be related to docetaxel resistance in gastric cancer cells, is not associated with FOXM1 expression significantly. These results were further provided by immunohistochemical analysis, indicating that FOXM1 and Stathmin expression levels were correlated in 103 post-operational gastric cancer specimens. Moreover, when we attenuated FOXM1 expression with FOXM1 inhibitor thiostrepton, docetaxel resistance in gastric cancers was found to be reversed, simultaneously with the down-regulation of FOXM1 and Stathmin. Therefore, FOXM1 can be a useful marker for predicting and monitoring docetaxel response. Through the inhibition of FOXM1, docetaxel resistance can be reversed, and thus FOXM1 could be a new therapeutic target in docetaxel-resistant gastric cancer.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Estatmina/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Taxoides/uso terapêutico , Regulação para Cima/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Docetaxel , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Gastrectomia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Cinesinas/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Análise Multivariada , Fenótipo , Prognóstico , Regiões Promotoras Genéticas/genética , Modelos de Riscos Proporcionais , Estatmina/metabolismo , Neoplasias Gástricas/cirurgia , Taxoides/farmacologia , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico , Regulação para Cima/efeitos dos fármacos
15.
Cell Cycle ; 11(18): 3370-2, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22894930

RESUMO

Nanoparticle-encapsulated thiazole antibiotic, thiostrepton, has been shown to be an effective agent for inhibiting tumor growth in solid tumor models through the inhibition of proteasomal activity by the induction of apoptosis in cancer cells. Here, we show the efficacy of thiostrepton-micelles in inhibiting tumor growth in a DEN/PB-induced liver cancer model. We also demonstrate an enhanced anticancer effect of the combination treatment of thiostrepton with bortezomib, another proteasome inhibitor in this liver cancer model.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ácidos Borônicos/uso terapêutico , Transformação Celular Neoplásica/patologia , Neoplasias Hepáticas/tratamento farmacológico , Pirazinas/uso terapêutico , Tioestreptona/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica/efeitos dos fármacos , Dietilnitrosamina , Modelos Animais de Doenças , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/patologia , Camundongos , Fenobarbital , Pirazinas/farmacologia , Tioestreptona/farmacologia
17.
Cell Cycle ; 7(12): 1851-5, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18583930

RESUMO

We previously described the identification of a nucleoside analog transcriptional inhibitor ARC (4-amino-6-hydrazino-7-beta-D-ribofuranosyl-7H-Pyrrolo[2,3-d]-pyrimidine-5-carboxamide) and FoxM1 inhibitor, thiazole antibiotic Siomycin A that were able to induce apoptosis in cancer cell lines of different origin. Here, we report the characterization of these drugs on a panel of melanoma cell lines. We found that in contrast to the common anti-melanoma drug dacarbazine (DTIC), ARC and thiazole antibiotics, Siomycin A and thiostrepton, efficiently inhibited growth and induced cell death in melanoma cell lines in low concentrations. Overexpression of the antiapoptotic protein Mcl-1 protected melanoma cells from apoptosis induced by these compounds. Furthermore, we found that ARC and Siomycin A synergistically induce apoptosis in DM833 melanoma cell line suggesting that they may antagonize different anti-apoptotic pathways in melanoma cells. In general, these drugs may represent important candidates for anti-cancer drug development against melanoma.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Melanoma/tratamento farmacológico , Nucleosídeos/farmacologia , Peptídeos/farmacologia , Pirimidinas/farmacologia , Tioestreptona/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Melanoma/patologia , Nucleosídeos/uso terapêutico , Peptídeos/uso terapêutico , Pirimidinas/uso terapêutico , Tioestreptona/uso terapêutico
18.
J Antibiot (Tokyo) ; 57(9): 590-6, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15580960

RESUMO

The antibiotic thiazole compound siomycin, which we have found from the culture broth of Actinomycetes (strain No.806097) in search of antibody production inhibitor, showed the in vitro immunosuppressive property against B-cells stimulated with T-cell independent antigen DNP-LPS (dinitrophenyl-lipopolysaccharide) while it also showed inhibitory effect against T-cell proliferation. Its inhibitory mechanism was considered to be different from that of FK506, the representative of T-cell immunosuppressant. Moreover, siomycin showed inhibitory effect in both T-cell dependent and independent murine antibody production models and decreased the severity in murine collagen arthritis model. Therefore, siomycin is a unique immunosuppressant which has potential for the treatment of some antibody-mediated diseases.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Artrite Experimental/tratamento farmacológico , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Linfócitos B/imunologia , Colágeno , Feminino , Humanos , Imunossupressores/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Peptídeos/uso terapêutico , Linfócitos T/imunologia , Tacrolimo/farmacologia , Tacrolimo/uso terapêutico , Tioestreptona/farmacologia , Tioestreptona/uso terapêutico
19.
Mol Biochem Parasitol ; 109(1): 17-23, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10924753

RESUMO

A chloroplast-like organelle is present in many species of the Apicomplexa phylum. We have previously demonstrated that the plastid organelle of Plasmodium faciparum is essential to the survival of the blood-stage malaria parasite in culture. One known function of the plastid organelle in another Apicomplexan, Toxoplasma gondii, involves the formation of the parasitophorous vacuole. The effects of interruption of plastid function on sporozoites and sexual-stage parasites have not been investigated. In our previous studies of the effects of thiostrepton, a polypeptide antibiotic from streptococcus spp., on erythrocytic schizongony of the human malaria P. falciparium, we found that this antibiotic appears to interact with the guanosine triphosphatase (GTPase) binding domain of the organellar large subunit ribosomal RNA, as it does in bacteria. We investigate here the effects of this drug on life-cycle stages of the malaria parasite in vivo. Preincubation of mature infective sporozoites with thiostrepton has no observable effect on their infectivity. Sporozoite infection both by mosquito bite and sporozoite injection was prevented by pretreatment of mice with thiostrepton. Thiostrepton eliminates infection with erythrocytic forms of Plasmodium berghei in mice. Clearance of infected red blood cells follows the delayed kinetics associated with drugs that interact with the apicoplast. Thiostrepton treatment of infected mice reduces transmission of parasites by more than ten-fold, indicating that the plastid has a role in sexual development of the parasite. These results indicate that the plastid function is accessible to drug action in vivo and important to the development of both sexual and asexual forms of the parasite.


Assuntos
Antibacterianos/uso terapêutico , Antimaláricos/uso terapêutico , Malária/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Tioestreptona/uso terapêutico , Animais , Culicidae/parasitologia , Vetores de Doenças , Relação Dose-Resposta a Droga , Feminino , Malária/parasitologia , Malária/transmissão , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/patogenicidade , Fatores de Tempo
20.
Aust Vet J ; 68(6): 193-5, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1888310

RESUMO

A topical preparation containing miconazole, polymyxin and prednisolone was shown to be more effective in the treatment of otitis externa in 167 dogs than 2 other ear preparations containing antibiotics, an antimycotic and a corticosteroid. With miconazole, polymyxin and prednisolone, the recurrence rate was 26.7% compared with 72.6% and 54.3% when the other products were used. The mean duration of treatment required to achieve resolution of clinical signs was 9.6 days, compared with 12.2 days and 13.0 days and no cases failed to respond to treatment, compared with 17.7% and 14.3%. Malassezia canis alone (71%) or in association with bacteria (18%) was recovered from 44 of 49 ears cultured.


Assuntos
Doenças do Cão/tratamento farmacológico , Miconazol/uso terapêutico , Otite Externa/veterinária , Polimixina B/uso terapêutico , Prednisolona/uso terapêutico , Administração Tópica , Animais , Betametasona/administração & dosagem , Betametasona/uso terapêutico , Dissulfiram/administração & dosagem , Dissulfiram/uso terapêutico , Cães , Combinação de Medicamentos , Miconazol/administração & dosagem , Neomicina/administração & dosagem , Neomicina/uso terapêutico , Nistatina/administração & dosagem , Nistatina/uso terapêutico , Otite Externa/tratamento farmacológico , Polimixina B/administração & dosagem , Prednisolona/administração & dosagem , Recidiva , Tioestreptona/administração & dosagem , Tioestreptona/uso terapêutico , Triancinolona/administração & dosagem , Triancinolona/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...