Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Invest New Drugs ; 40(6): 1244-1253, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36306030

RESUMO

The endoplasmic reticulum (ER) is a critical organelle that preserves the protein homeostasis of cells. Under various stress conditions, cells evolve a degree of capacity to maintain ER proteostasis, which is usually augmented in tumor cells, including colorectal cancer (CRC) cells, to bolster their survival and resistance to apoptosis. Bortezomib (BTZ) is a promising drug used in CRC treatment; however, its main limitation result from drug resistance. Here, we identified the role of tripartite motif-containing protein 59 (TRIM59)-a protein localized on the ER membrane- in the prevention of BTZ-mediated CRC killing. Depletion of TRIM59 is associated with the enhancement of ER stress and a remarkable increase in unfolded protein response (UPR) signaling. Besides, TRIM59 strengthens ER-associated degradation (ERAD) and alleviates the generation of ROS. Of note, TRIM59 knockdown synergizes with the anti-cancer effect of BTZ both in vitro and in vivo. Our findings revealed a role for TRIM59 in the ER by guarding ER proteostasis and represents a novel therapeutic target of CRC.


Assuntos
Neoplasias Colorretais , Proteostase , Humanos , Bortezomib/farmacologia , Retículo Endoplasmático/metabolismo , Resposta a Proteínas não Dobradas , Estresse do Retículo Endoplasmático , Apoptose , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia
2.
J Org Chem ; 85(24): 16226-16235, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33264019

RESUMO

Two new ring-size-varying analogues (2 and 3) of ipomoeassin F were synthesized and evaluated. Improved cytotoxicity (IC50: from 1.8 nM) and in vitro protein translocation inhibition (IC50: 35 nM) derived from ring expansion imply that the binding pocket of Sec61α (isoform 1) can accommodate further structural modifications, likely in the fatty acid portion. Streamlined preparation of the key diol intermediate 5 enabled gram-scale production, allowing us to establish that ipomoeassin F is biologically active in vivo (MTD: ∼3 mg/kg).


Assuntos
Glicoconjugados , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Relação Estrutura-Atividade
3.
Genes Dev ; 26(12): 1300-5, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22677547

RESUMO

The Drosophila TEAD ortholog Scalloped is required for Yki-mediated overgrowth but is largely dispensable for normal tissue growth, suggesting that its mammalian counterpart may be exploited for selective inhibition of oncogenic growth driven by YAP hyperactivation. Here we test this hypothesis genetically and pharmacologically. We show that a dominant-negative TEAD molecule does not perturb normal liver growth but potently suppresses hepatomegaly/tumorigenesis resulting from YAP overexpression or Neurofibromin 2 (NF2)/Merlin inactivation. We further identify verteporfin as a small molecule that inhibits TEAD-YAP association and YAP-induced liver overgrowth. These findings provide proof of principle that inhibiting TEAD-YAP interactions is a pharmacologically viable strategy against the YAP oncoprotein.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/genética , Complexos Multiproteicos/metabolismo , Fosfoproteínas/genética , Porfirinas/farmacologia , Fatores de Transcrição/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Transformação Celular Neoplásica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genes Dominantes/genética , Células HEK293 , Hepatomegalia/metabolismo , Hepatomegalia/patologia , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Transgênicos , Neurofibromina 2/metabolismo , Fenótipo , Fosfoproteínas/metabolismo , Ligação Proteica/efeitos dos fármacos , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/metabolismo , Verteporfina , Proteínas de Sinalização YAP
4.
Proc Natl Acad Sci U S A ; 112(52): E7276-85, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26655341

RESUMO

Itraconazole, a clinically used antifungal drug, was found to possess potent antiangiogenic and anticancer activity that is unique among the azole antifungals. Previous mechanistic studies have shown that itraconazole inhibits the mechanistic target of rapamycin (mTOR) signaling pathway, which is known to be a critical regulator of endothelial cell function and angiogenesis. However, the molecular target of itraconazole that mediates this activity has remained unknown. Here we identify the major target of itraconazole in endothelial cells as the mitochondrial protein voltage-dependent anion channel 1 (VDAC1), which regulates mitochondrial metabolism by controlling the passage of ions and small metabolites through the outer mitochondrial membrane. VDAC1 knockdown profoundly inhibits mTOR activity and cell proliferation in human umbilical vein cells (HUVEC), uncovering a previously unknown connection between VDAC1 and mTOR. Inhibition of VDAC1 by itraconazole disrupts mitochondrial metabolism, leading to an increase in the cellular AMP:ATP ratio and activation of the AMP-activated protein kinase (AMPK), an upstream regulator of mTOR. VDAC1-knockout cells are resistant to AMPK activation and mTOR inhibition by itraconazole, demonstrating that VDAC1 is the mediator of this activity. In addition, another known VDAC-targeting compound, erastin, also activates AMPK and inhibits mTOR and proliferation in HUVEC. VDAC1 thus represents a novel upstream regulator of mTOR signaling in endothelial cells and a promising target for the development of angiogenesis inhibitors.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Itraconazol/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Canal de Ânion 1 Dependente de Voltagem/antagonistas & inibidores , Animais , Antifúngicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Células HeLa , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Microscopia de Fluorescência , Dilatação Mitocondrial/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Canal de Ânion 1 Dependente de Voltagem/genética , Canal de Ânion 1 Dependente de Voltagem/metabolismo
5.
Molecules ; 21(7)2016 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-27455225

RESUMO

Epithelial-mesenchymal transition (EMT) is known to play an important role in cancer progression, metastasis and drug resistance. Although there are controversies surrounding the causal relationship between EMT and cancer metastasis, the role of EMT in cancer drug resistance has been increasingly recognized. Numerous EMT-related signaling pathways are involved in drug resistance in cancer cells. Cells undergoing EMT show a feature similar to cancer stem cells (CSCs), such as an increase in drug efflux pumps and anti-apoptotic effects. Therefore, targeting EMT has been considered a novel opportunity to overcome cancer drug resistance. This review describes the mechanism by which EMT contributes to drug resistance in cancer cells and summarizes new advances in research in EMT-associated drug resistance.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Descoberta de Drogas , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral
6.
Biochem Biophys Res Commun ; 450(1): 561-7, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24928393

RESUMO

Adipocyte-derived leucine aminopeptidase (A-LAP) is a novel member of the M1 family of zinc metallopeptidases, which has been reported to play a crucial role in angiogenesis. In the present study, we conducted a target-based screening of natural products and synthetic chemical libraries using the purified enzyme to search novel inhibitors of A-LAP. Amongst several hits isolated, a natural product purpurin was identified as one of the most potent inhibitors of A-LAP from the screening. In vitro enzymatic analyses demonstrated that purpurin inhibited A-LAP activity in a non-competitive manner with a Ki value of 20 M. In addition, purpurin showed a strong selectivity toward A-LAP versus another member of M1 family of zinc metallopeptidase, aminopeptidase N (APN). In angiogenesis assays, purpurin inhibited the vascular endothelial growth factor (VEGF)-induced invasion and tube formation of human umbilical vein endothelial cells (HUVEC). Moreover, purpurin inhibited in vivo angiogenesis in zebrafish embryo without toxicity. These data demonstrate that purpurin is a novel specific inhibitor of A-LAP and could be developed as a new anti-angiogenic agent.


Assuntos
Adipócitos/enzimologia , Antraquinonas/farmacologia , Células Endoteliais/fisiologia , Leucil Aminopeptidase/antagonistas & inibidores , Leucil Aminopeptidase/metabolismo , Neovascularização Fisiológica/fisiologia , Peixe-Zebra/fisiologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Animais , Neovascularização Fisiológica/efeitos dos fármacos
7.
Hepatology ; 57(6): 2458-68, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23325555

RESUMO

UNLABELLED: Patient-specific induced pluripotent stem cells (iPSCs) represent a potential source for developing novel drug and cell therapies. Although increasing numbers of disease-specific iPSCs have been generated, there has been limited progress in iPSC-based drug screening/discovery for liver diseases, and the low gene-targeting efficiency in human iPSCs warrants further improvement. Using iPSC lines from patients with alpha-1 antitrypsin (AAT) deficiency, for which there is currently no drug or gene therapy available, we established a platform to discover new drug candidates and correct disease-causing mutation with a high efficiency. A high-throughput format screening assay, based on our hepatic differentiation protocol, was implemented to facilitate automated quantification of cellular AAT accumulation using a 96-well immunofluorescence reader. To expedite the eventual application of lead compounds to patients, we conducted drug screening utilizing our established library of clinical compounds (the Johns Hopkins Drug Library) with extensive safety profiles. Through a blind large-scale drug screening, five clinical drugs were identified to reduce AAT accumulation in diverse patient iPSC-derived hepatocyte-like cells. In addition, using the recently developed transcription activator-like effector nuclease technology, we achieved high gene-targeting efficiency in AAT-deficiency patient iPSCs with 25%-33% of the clones demonstrating simultaneous targeting at both diseased alleles. The hepatocyte-like cells derived from the gene-corrected iPSCs were functional without the mutant AAT accumulation. This highly efficient and cost-effective targeting technology will broadly benefit both basic and translational applications. CONCLUSIONS: Our results demonstrated the feasibility of effective large-scale drug screening using an iPSC-based disease model and highly robust gene targeting in human iPSCs, both of which are critical for translating the iPSC technology into novel therapies for untreatable diseases.


Assuntos
Hepatócitos/efeitos dos fármacos , Hepatopatias/terapia , Células-Tronco Pluripotentes/efeitos dos fármacos , Reparo Gênico Alvo-Dirigido/métodos , Deficiência de alfa 1-Antitripsina/terapia , Diferenciação Celular , Células Cultivadas , Hepatócitos/citologia , Humanos , Hepatopatias/genética
8.
Proc Natl Acad Sci U S A ; 108(48): 19264-9, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22084084

RESUMO

Pancreatic ß-cells are an essential source of insulin and their destruction because of autoimmunity causes type I diabetes. We conducted a chemical screen to identify compounds that would induce the differentiation of insulin-producing ß-cells in vivo. To do this screen, we brought together the use of transgenic zebrafish as a model of ß-cell differentiation, a unique multiwell plate that allows easy visualization of lateral views of swimming larval fish and a library of clinical drugs. We identified six hits that can induce precocious differentiation of secondary islets in larval zebrafish. Three of these six hits were known drugs with a considerable background of published data on mechanism of action. Using pharmacological approaches, we have identified and characterized two unique pathways in ß-cell differentiation in the zebrafish, including down-regulation of GTP production and retinoic acid biosynthesis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Descoberta de Drogas/métodos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Preparações Farmacêuticas/metabolismo , Acetanilidas/farmacologia , Animais , Animais Geneticamente Modificados , Ácidos Cafeicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Primers do DNA/genética , Dimetil Sulfóxido , Relação Dose-Resposta a Droga , Epirizol/farmacologia , Imunofluorescência , Proteínas de Fluorescência Verde , Guanosina Trifosfato/biossíntese , Proteína HMGB1/metabolismo , Larva/efeitos dos fármacos , Microscopia Confocal , Ácido Micofenólico/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Ácidos Sulfanílicos/farmacologia , Tretinoína/metabolismo , Peixe-Zebra , p-Aminoazobenzeno/análogos & derivados , p-Aminoazobenzeno/farmacologia
9.
Int J Biol Sci ; 20(6): 1978-1991, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617536

RESUMO

Loss of PTEN tumor suppressor is an important event during colorectal cancer (CRC) development and is a target for therapeutic exploitation. This study reports that bromodomain and extra-terminal motif (BET) is a synthetic lethal partner of PTEN in CRC. BET inhibition (BETi) selectively induced G1 cell cycle arrest and apoptosis in PTEN-/- CRC. Further, BETi selectively and dose-dependently suppressed the growth of PTEN-/- CRC tumor xenografts in mice and patient-derived organoids. Mechanistically, PTEN-deficient CRC cells elevated the level of cytoplasmic p21CIP1/WAF1 that is hyper-phosphorylated at Thr145 by AKT. BETi suppressed AKT activation in PTEN-deficient CRC cells, followed by the reduction in p21 phosphorylation at Thr145, thereby promoting its nuclear translocation. In addition, BETi suppressed MYC level and this in turn increased the total p21 level in the nuclei. Over-expression of a phospho-mimetic p21 mutant (T145D) significantly rescued the BETi effect on PTEN-deficient CRC. These results suggest that BETi has a dual action on p21: elevating the level of p21 by inhibiting MYC and converting the oncogenic (cytoplasmic) p21 into the tumor-suppressive (nuclear) p21 by inhibiting AKT. Taken together, this study identified the synthetic lethal interaction between PTEN and BET, and provides a potential actionable target for CRC with PTEN loss.


Assuntos
Neoplasias Colorretais , Mutações Sintéticas Letais , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas c-akt , Fosforilação , Citoplasma , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , PTEN Fosfo-Hidrolase/genética
10.
Bioorg Med Chem Lett ; 23(9): 2733-7, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23518276

RESUMO

Tricyclic thiazoleamine derivatives that were identified as hits in a screen against human umbilical vein endothelial cell proliferation were subjected to a structure-activity relationship study. Two structurally superimposable scaffolds-4H-thiochromeno[4,3-d]thiazol-2-amine and 5,6-dihydro-4H-benzo[6,7]cyclohepta[1,2-d]thiazol-2-amine derivatives-yielded low-micromolar inhibitors, and two among them 37 and 43 also exhibited antiangiogenic activity in an endothelial tube formation assay. Thus, 37 and 43 can serve as leads to develop a novel class of antiangiogenic agents.


Assuntos
Inibidores da Angiogênese/química , Tiazóis/química , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/farmacologia , Proliferação de Células/efeitos dos fármacos , Ciclização , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Células Jurkat , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/farmacologia
11.
Int J Biol Sci ; 19(11): 3544-3557, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37496993

RESUMO

Colorectal cancer (CRC) driven by PTEN deficiency exhibits high risk of metastasis, advancement of tumor stages and chemotherapy resistance, where no effective therapy has been developed. In this study, we performed a synthetic lethal drug screening in CRC and found that PTEN-deficient CRC cells are highly vulnerable to MDM2 inhibition. MDM2 inhibitor treatment or its silencing selectively inhibited the growth of PTEN-deficient CRC in vitro and in mice models. Mechanistically, PTEN loss increased the level of active AKT and subsequently increased MDM2 phosphorylation, thereby limiting the p53 functions in PTEN-/- CRC cells. MDM2 inhibition in turn activated p53 in CRC, particularly in PTEN-/- CRC cells. The synthetic lethal effect of MDM2 inhibitor was largely dependent on p53, because p53 silenced cells or cells lacking p53 failed to exhibit synthetic lethality in PTEN-deficient cells. We further showed that MDM2 inhibition led to the p53-dependent reversal of Bcl2-Bax ratio, which contributed to mitochondria-mediated apoptotic cell death in PTEN-deficient CRC. This study suggests that pharmacological targeting of MDM2 could be a potential therapeutic strategy for PTEN-deficient CRC.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Animais , Camundongos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Int J Biol Sci ; 19(13): 4020-4035, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37705743

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer where no effective therapy has been developed. Here, we report that the natural product ER translocon inhibitor ipomoeassin F is a selective inhibitor of TNBC cell growth. A proteomic analysis of TNBC cells revealed that ipomoeassin F significantly reduced the levels of ER molecular chaperones, including PDIA6 and PDIA4, and induced ER stress, unfolded protein response (UPR) and autophagy in TNBC cells. Mechanistically, ipomoeassin F, as an inhibitor of Sec61α-containing ER translocon, blocks ER translocation of PDIA6, inducing its proteasomal degradation. Silencing of PDIA6 or PDIA4 by RNA interferences or treatment with a small molecule inhibitor of the protein disulfide isomerases in TNBC cells successfully recapitulated the ipomoeassin F phenotypes, including the induction of ER stress, UPR and autophagy, suggesting that the reduction of PDIAs is the key mediator of the pharmacological effects of ipomoeassin F. Moreover, ipomoeassin F significantly suppressed TNBC growth in a mouse tumor xenograft model, with a marked reduction in PDIA6 and PDIA4 levels in the tumor samples. Our study demonstrates that Sec61α-containing ER translocon and PDIAs are potential drug targets for TNBC and suggests that ipomoeassin F could serve as a lead for developing ER translocon-targeted therapy for TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Proteômica , Glicoconjugados , Modelos Animais de Doenças , Chaperonas Moleculares
13.
J Clin Invest ; 133(9)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36928177

RESUMO

Aurora A plays a critical role in G2/M transition and mitosis, making it an attractive target for cancer treatment. Aurora A inhibitors showed remarkable antitumor effects in preclinical studies, but unsatisfactory outcomes in clinical trials have greatly limited their development. In this study, the Aurora A inhibitor alisertib upregulated programmed death ligand 1 (PD-L1) expression in a panel of tumor cells both in vitro and in vivo. Upregulation of the checkpoint protein PD-L1 reduced antitumor immunity in immune-competent mice, paradoxically inhibiting the antitumor effects of alisertib. Mechanistically, Aurora A directly bound to and phosphorylated cyclic GMP-AMP synthase (cGAS), suppressing PD-L1 expression in tumor cells. Aurora A inhibition by alisertib activated the cGAS/stimulator of IFN genes (STING)/NF-κB pathway and promoted PD-L1 expression. Combining alisertib with anti-PD-L1 antibody improved antitumor immunity and enhanced the antitumor effects of alisertib in immune-competent mice. Our results, which reveal the immunomodulatory functions of Aurora A inhibitors and provide a plausible explanation for the poor clinical outcomes with their use, offer a potential approach to improve the antitumor efficacy of these inhibitors.


Assuntos
Aurora Quinase A , Inibidores de Proteínas Quinases , Animais , Camundongos , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Nucleotidiltransferases , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Humanos
14.
Cancer Res ; 83(15): 2614-2633, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37227919

RESUMO

Cancer metastasis is an extremely complex process affected by many factors. An acidic microenvironment can drive cancer cell migration toward blood vessels while also hampering immune cell activity. Here, we identified a mechanism mediated by sialyltransferases that induces an acidic tumor-permissive microenvironment (ATPME) in BRCA1-mutant and most BRCA1-low breast cancers. Hypersialylation mediated by ST8SIA4 perturbed the mammary epithelial bilayer structure and generated an ATPME and immunosuppressive microenvironment with increased PD-L1 and PD1 expressions. Mechanistically, BRCA1 deficiency increased expression of VEGFA and IL6 to activate TGFß-ST8SIA4 signaling. High levels of ST8SIA4 led to accumulation of polysialic acid (PSA) on mammary epithelial membranes that facilitated escape of cancer cells from immunosurveillance, promoting metastasis and resistance to αPD1 treatment. The sialyltransferase inhibitor 3Fax-Peracetyl Neu5Ac neutralized the ATPME, sensitized cancers to immune checkpoint blockade by activating CD8 T cells, and inhibited tumor growth and metastasis. Together, these findings identify a potential therapeutic option for cancers with a high level of PSA. SIGNIFICANCE: BRCA1 deficiency generates an acidic microenvironment to promote cancer metastasis and immunotherapy resistance that can be reversed using a sialyltransferase inhibitor.


Assuntos
Neoplasias da Mama , Microambiente Tumoral , Humanos , Feminino , Imunoterapia , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Neoplasias da Mama/patologia , Sialiltransferases/genética , Linhagem Celular Tumoral , Proteína BRCA1/genética
15.
Org Biomol Chem ; 10(15): 2979-92, 2012 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-22391578

RESUMO

Two substituted oxines, nitroxoline (5) and 5-chloroquinolin-8-yl phenylcarbamate (22), were identified as hits in a high-throughput screen aimed at finding new anti-angiogenic agents. In a previous study, we have elucidated the molecular mechanism of antiproliferative activity of nitroxoline in endothelial cells, which comprises of a dual inhibition of type 2 human methionine aminopeptidase (MetAP2) and sirtuin 1 (SIRT1). Structure-activity relationship study (SAR) of nitroxoline offered many surprises where minor modifications yielded oxine derivatives with increased potency against human umbilical vein endothelial cells (HUVEC), but with entirely different as yet unknown mechanisms. For example, 5-nitrosoquinolin-8-ol (33) inhibited HUVEC growth with sub-micromolar IC(50), but did not affect MetAP2 or MetAP1, and it only showed weak inhibition against SIRT1. Other sub-micromolar inhibitors were derivatives of 5-aminoquinolin-8-ol (34) and 8-sulfonamidoquinoline (32). A sulfamate derivative of nitroxoline (48) was found to be more potent than nitroxoline with the retention of activities against MetAP2 and SIRT1. The bioactivity of the second hit, micromolar HUVEC and MetAP2 inhibitor carbamate 22 was improved further with an SAR study culminating in carbamate 24 which is a nanomolar inhibitor of HUVEC and MetAP2.


Assuntos
Inibidores da Angiogênese/síntese química , Inibidores Enzimáticos/síntese química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Hidroxiquinolinas/síntese química , Nitroquinolinas/síntese química , Fenilcarbamatos/síntese química , Aminopeptidases/antagonistas & inibidores , Aminopeptidases/metabolismo , Inibidores da Angiogênese/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Ensaios de Triagem em Larga Escala , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Hidroxiquinolinas/farmacologia , Metionil Aminopeptidases , Nitroquinolinas/farmacologia , Fenilcarbamatos/farmacologia , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Relação Estrutura-Atividade
16.
Open Biol ; 12(8): 220098, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35946309

RESUMO

Neurodegenerative diseases (NDs) are a diverse group of disorders characterized by the progressive degeneration of the structure and function of the central or peripheral nervous systems. One of the major features of NDs, such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD), is the aggregation of specific misfolded proteins, which induces cellular dysfunction, neuronal death, loss of synaptic connections and eventually brain damage. By far, a great amount of evidence has suggested that TRIM family proteins play crucial roles in the turnover of normal regulatory and misfolded proteins. To maintain cellular protein quality control, cells rely on two major classes of proteostasis: molecular chaperones and the degradative systems, the latter includes the ubiquitin-proteasome system (UPS) and autophagy; and their dysfunction has been established to result in various physiological disorders including NDs. Emerging evidence has shown that TRIM proteins are key players in facilitating the clearance of misfolded protein aggregates associated with neurodegenerative disorders. Understanding the different pathways these TRIM proteins employ during episodes of neurodegenerative disorder represents a promising therapeutic target. In this review, we elucidated and summarized the diverse roles with underlying mechanisms of members of the TRIM family proteins in NDs.


Assuntos
Doenças Neurodegenerativas , Humanos , Chaperonas Moleculares/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteostase , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina/metabolismo
17.
Oncogene ; 41(19): 2734-2748, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35393542

RESUMO

SMAD4 loss-of-function mutations have been frequently observed in colorectal cancer (CRC) and are recognized as a drug target for therapeutic exploitation. In this study, we performed a synthetic lethal drug screening with SMAD4-isogenic CRC cells and found that aurora kinase A (AURKA) inhibition is synthetic lethal with SMAD4 loss. Inhibition of AURKA selectively inhibited the growth of SMAD4-/- CRC in vitro and in vivo. Mechanistically, SMAD4 negatively regulated AURKA level, resulting in the significant elevation of AURKA in SMAD4-/- CRC cells. Inhibition of AURKA induced G2/M cell cycle delay in SMAD4+/+ CRC cells, but induced apoptosis in SMAD4-/- CRC cells. We further observed that a high level of AURKA in SMAD4-/- CRC cells led to abnormal mitotic spindles, leading to cellular aneuploidy. Moreover, SMAD4-/- CRC cells expressed high levels of spindle assembly checkpoint (SAC) proteins, suggesting the hyperactivation of SAC. The silencing of key SAC proteins significantly rescued the AURKA inhibition-induced cell death in SMAD4-/- cells, suggesting that SMAD4-/- CRC cells are hyper-dependent on AURKA activity for mitotic exit and survival during SAC hyperactivation. This study presents a unique synthetic lethal interaction between SMAD4 and AURKA and suggests that AURKA could be a potential drug target in SMAD4-deficient CRC.


Assuntos
Aurora Quinase A , Neoplasias Colorretais , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Proteína Smad4/genética , Proteína Smad4/metabolismo , Mutações Sintéticas Letais
18.
Adv Sci (Weinh) ; 9(26): e2200562, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35712764

RESUMO

G protein-coupled receptors (GPCRs) are the most common and important drug targets. However, >70% of GPCRs are undruggable or difficult to target using conventional chemical agonists/antagonists. Small nucleic acid molecules, which can sequence-specifically modulate any gene, offer a unique opportunity to effectively expand drug targets, especially those that are undruggable or difficult to address, such as GPCRs. Here, the authors report  for the first time that small activating RNAs (saRNAs) effectively modulate a GPCR for cancer treatment. Specifically, saRNAs promoting the expression of Mas receptor (MAS1), a GPCR that counteracts the classical angiotensin II pathway in cancer cell proliferation and migration, are identified. These saRNAs, delivered by an amphiphilic dendrimer vector, enhance MAS1 expression, counteracting the angiotensin II/angiotensin II Receptor Type 1 axis, and leading to significant suppression of tumorigenesis and the inhibition of tumor progression of multiple cancers in tumor-xenografted mouse models and patient-derived tumor models. This study provides not only a new strategy for cancer therapy by targeting the renin-angiotensin system, but also a new avenue to modulate GPCR signaling by RNA activation.


Assuntos
Angiotensina II , Neoplasias , Angiotensina II/metabolismo , Animais , Camundongos , Neoplasias/genética , Neoplasias/terapia , RNA/metabolismo , Receptores Acoplados a Proteínas G/genética , Sistema Renina-Angiotensina
19.
J Biol Chem ; 285(33): 25867-74, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20554536

RESUMO

Recent reports have shown that Ca(2+)/calmodulin (Ca(2+)/CaM) signaling plays a crucial role in angiogenesis. We previously developed a new Ca(2+)/CaM antagonist, HBC (4-{3,5-bis-[2-(4-hydroxy-3-methoxyphenyl)ethyl]-4,5-dihydropyrazol-1-yl}benzoic acid), from a curcumin-based synthetic chemical library. Here, we investigated its anti-angiogenic activity and mode of action. HBC potently inhibited the proliferation of human umbilical vascular endothelial cells with no cytotoxicity. Furthermore, HBC blocked in vitro characteristics of angiogenesis such as tube formation and chemoinvasion, as well as neovascularization of the chorioallantoic membrane of growing chick embryos in vivo. Notably, HBC markedly inhibited expression of hypoxia-inducible factor-1alpha (HIF-1alpha) at the translational level during hypoxia, thereby reducing HIF-1 transcriptional activity and expression of its major target gene, vascular endothelial growth factor. In addition, combination treatment with HBC and various HIF-1 inhibitors, including suberoylanilide hydroxamic acid, rapamycin, and terpestacin, had greater anti-angiogenic activity than treatment with each single agent. Collectively, our findings indicate that HBC is a new anti-angiogenic agent targeting HIF that can be used to explore the biological role of Ca(2+)/CaM in angiogenesis.


Assuntos
Ácido Benzoico/uso terapêutico , Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Patológica/tratamento farmacológico , Animais , Ácido Benzoico/química , Western Blotting , Compostos Bicíclicos com Pontes/uso terapêutico , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Galinhas , Ensaio de Imunoadsorção Enzimática , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Fator 1 Induzível por Hipóxia/genética , Neovascularização Patológica/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/uso terapêutico , Vorinostat
20.
J Biol Chem ; 285(15): 11584-95, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20145250

RESUMO

Cellular oxygen sensing is required for hypoxia-inducible factor-1alpha stabilization, which is important for tumor cell survival, proliferation, and angiogenesis. Here we find that terpestacin, a small molecule previously identified in a screen of microbial extracts, binds to the 13.4-kDa subunit (UQCRB) of mitochondrial Complex III, resulting in inhibition of hypoxia-induced reactive oxygen species generation. Consequently, such inhibition blocks hypoxia-inducible factor activation and tumor angiogenesis in vivo, without inhibiting mitochondrial respiration. Overexpression of UQCRB or its suppression using RNA interference demonstrates that it plays a crucial role in the oxygen sensing mechanism that regulates responses to hypoxia. These findings provide a novel molecular basis of terpestacin targeting UQCRB of Complex III in selective suppression of tumor progression.


Assuntos
Proteínas de Transporte/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Hipóxia , Mitocôndrias/metabolismo , Neovascularização Patológica , Oxigênio/metabolismo , Espécies Reativas de Oxigênio , Animais , Compostos Bicíclicos com Pontes/farmacologia , Endotélio Vascular/citologia , Feminino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA