Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Br J Cancer ; 119(11): 1392-1400, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30425352

RESUMO

BACKGROUND: Poly-ADP ribose polymerase (PARP) inhibitor-based cancer therapy selectively targets cells with deficient homologous recombination repair. Considering their long-term use in maintenance treatment, any potential mutagenic effect of PARP inhibitor treatment could accelerate the development of resistance or harm non-malignant somatic cells. METHODS: We tested the mutagenicity of long-term treatment with the PARP inhibitor niraparib using whole-genome sequencing of cultured cell clones and whole-exome sequencing of patient-derived breast cancer xenografts. RESULTS: We observed no significant increase in the number and alteration in the spectrum of base substitutions, short insertions and deletions and genomic rearrangements upon niraparib treatment of human DLD-1 colon adenocarcinoma cells, wild-type and BRCA1 mutant chicken DT40 lymphoblastoma cells and BRCA1-defective SUM149PT breast carcinoma cells, except for a minor increase in specific deletion classes. We also did not detect any contribution of in vivo niraparib treatment to subclonal mutations arising in breast cancer-derived xenografts. CONCLUSIONS: The results suggest that long-term inhibition of DNA repair with PARP inhibitors has no or only limited mutagenic effect. Mutagenesis due to prolonged use of PARP inhibitors in cancer treatment is therefore not expected to contribute to the genetic evolution of resistance, generate significant immunogenic neoepitopes or induce secondary malignancies.


Assuntos
Antineoplásicos/uso terapêutico , Indazóis/uso terapêutico , Mutação , Piperidinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos
2.
Proc Natl Acad Sci U S A ; 112(6): 1839-44, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25605917

RESUMO

Partial or even complete cancer regression can be achieved in some patients with current cancer treatments. However, such initial responses are almost always followed by relapse, with the recurrent cancer being resistant to further treatments. The discovery of therapeutic approaches that counteract relapse is, therefore, essential for advancing cancer medicine. Cancer cells are extremely heterogeneous, even in each individual patient, in terms of their malignant potential, drug sensitivity, and their potential to metastasize and cause relapse. Indeed, hypermalignant cancer cells, termed cancer stem cells or stemness-high cancer cells, that are highly tumorigenic and metastatic have been isolated from cancer patients with a variety of tumor types. Moreover, such stemness-high cancer cells are resistant to conventional chemotherapy and radiation. Here we show that BBI608, a small molecule identified by its ability to inhibit gene transcription driven by Stat3 and cancer stemness properties, can inhibit stemness gene expression and block spherogenesis of or kill stemness-high cancer cells isolated from a variety of cancer types. Moreover, cancer relapse and metastasis were effectively blocked by BBI608 in mice. These data demonstrate targeting cancer stemness as a novel approach to develop the next generation of cancer therapeutics to suppress cancer relapse and metastasis.


Assuntos
Antineoplásicos/farmacologia , Benzofuranos/farmacologia , Naftoquinonas/farmacologia , Metástase Neoplásica/prevenção & controle , Células-Tronco Neoplásicas/efeitos dos fármacos , Prevenção Secundária/métodos , Animais , Antineoplásicos/efeitos adversos , Benzofuranos/efeitos adversos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Xenoenxertos , Concentração Inibidora 50 , Camundongos , Naftoquinonas/efeitos adversos
3.
Bioorg Med Chem Lett ; 26(19): 4775-4780, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27578247

RESUMO

During the lead generation and optimization of PARP inhibitors blocking centrosome clustering, it was discovered that increasing hydrogen bond acceptor (HBA) strength improved cellular potency but led to elevated Caco2 and MDR1 efflux and thus poor oral bioavailability. Conversely, compounds with lower efflux had reduced potency. The project team was able to improve the bioavailability by reducing efflux through systematic modifications to the strength of the HBA by changing the electronic properties of neighboring groups, whilst maintaining sufficient acceptor strength for potency. Additionally, it was observed that enantiomers with different potency showed similar efflux, which is consistent with the promiscuity of efflux transporters. Eventually, a balance between potency and low efflux was achieved for a set of lead compounds with good bioavailability which allowed the project to progress towards establishing in vivo pharmacokinetic/pharmacodynamic relationships.


Assuntos
Centrossomo/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Células CACO-2 , Cães , Humanos , Ligação de Hidrogênio , Células Madin Darby de Rim Canino , Camundongos , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Ratos
4.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26546219

RESUMO

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Assuntos
Centrossomo/metabolismo , Ftalazinas/química , Inibidores de Poli(ADP-Ribose) Polimerases/química , Administração Oral , Animais , Sítios de Ligação , Células CACO-2 , Centrossomo/efeitos dos fármacos , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Células HeLa , Humanos , Microssomos/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Ftalazinas/administração & dosagem , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Estrutura Terciária de Proteína , Ratos , Tanquirases/antagonistas & inibidores , Tanquirases/metabolismo
5.
Nat Cell Biol ; 9(2): 160-70, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17330329

RESUMO

Centrosomes organize the microtubule cytoskeleton for both interphase and mitotic functions. They are implicated in cell-cycle progression but the mechanism is unknown. Here, we show that depletion of 14 out of 15 centrosome proteins arrests human diploid cells in G1 with reduced Cdk2-cyclin A activity and that expression of a centrosome-disrupting dominant-negative construct gives similar results. Cell-cycle arrest is always accompanied by defects in centrosome structure and function (for example, duplication and primary cilia assembly). The arrest occurs from within G1, excluding contributions from mitosis and cytokinesis. The arrest requires p38, p53 and p21, and is preceded by p38-dependent activation and centrosomal recruitment of p53. p53-deficient cells fail to arrest, leading to centrosome and spindle dysfunction and aneuploidy. We propose that loss of centrosome integrity activates a checkpoint that inhibits G1-S progression. This model satisfies the definition of a checkpoint in having three elements: a perturbation that is sensed, a transducer (p53) and a receiver (p21).


Assuntos
Centrossomo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fase G1/fisiologia , Fase S/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular , Células Cultivadas , Centrossomo/ultraestrutura , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Células HeLa , Humanos
6.
J Med Chem ; 64(15): 11302-11329, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34292726

RESUMO

Indoleamine 2,3-dioxygenase 1 (IDO1), a heme-containing enzyme that mediates the rate-limiting step in the metabolism of l-tryptophan to kynurenine, has been widely explored as a potential immunotherapeutic target in oncology. We developed a class of inhibitors with a conformationally constrained bicyclo[3.1.0]hexane core. These potently inhibited IDO1 in a cellular context by binding to the apoenzyme, as elucidated by biochemical characterization and X-ray crystallography. A SKOV3 tumor model was instrumental in differentiating compounds, leading to the identification of IACS-9779 (62) and IACS-70465 (71). IACS-70465 has excellent cellular potency, a robust pharmacodynamic response, and in a human whole blood assay was more potent than linrodostat (BMS-986205). IACS-9779 with a predicted human efficacious once daily dose below 1 mg/kg to sustain >90% inhibition of IDO1 displayed an acceptable safety margin in rodent toxicology and dog cardiovascular studies to support advancement into preclinical safety evaluation for human development.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Estrutura Molecular , Relação Estrutura-Atividade
7.
Trends Cell Biol ; 15(6): 303-11, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15953548

RESUMO

Early observations of centrosomes, made a century ago, revealed a tiny dark structure surrounded by a radial array of cytoplasmic fibers. We now know that the fibers are microtubules and that the dark organelles are centrosomes that mediate functions far beyond the more conventional role of microtubule organization. More recent evidence demonstrates that the centrosome serves as a scaffold for anchoring an extensive number of regulatory proteins. Among these are cell-cycle regulators whose association with the centrosome is an essential step in cell-cycle control. Such studies show that the centrosome is required for several cell-cycle transitions, including G(1) to S-phase, G(2) to mitosis and metaphase to anaphase. In this review (which is part of the Chromosome Segregation and Aneuploidy series), we discuss recent data that provide the most direct links between centrosomes and cell-cycle progression.


Assuntos
Ciclo Celular , Centrossomo/metabolismo , Aneuploidia , Animais , Humanos , Transdução de Sinais , Tubulina (Proteína)/metabolismo
8.
J Med Chem ; 63(17): 9888-9911, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787110

RESUMO

Tumor-associated macrophages (TAMs) have a significant presence in the tumor stroma across multiple human malignancies and are believed to be beneficial to tumor growth. Targeting CSF1R has been proposed as a potential therapy to reduce TAMs, especially the protumor, immune-suppressive M2 TAMs. Additionally, the high expression of CSF1R on tumor cells has been associated with poor survival in certain cancers, suggesting tumor dependency and therefore a potential therapeutic target. The CSF1-CSF1R signaling pathway modulates the production, differentiation, and function of TAMs; however, the discovery of selective CSF1R inhibitors devoid of type III kinase activity has proven to be challenging. We discovered a potent, highly selective, and orally bioavailable CSF1R inhibitor, IACS-9439 (1). Treatment with 1 led to a dose-dependent reduction in macrophages, promoted macrophage polarization toward the M1 phenotype, and led to tumor growth inhibition in MC38 and PANC02 syngeneic tumor models.


Assuntos
Antineoplásicos/uso terapêutico , Benzotiazóis/uso terapêutico , Neoplasias/tratamento farmacológico , Pirimidinas/uso terapêutico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Benzotiazóis/síntese química , Benzotiazóis/farmacocinética , Estabilidade de Medicamentos , Humanos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Pirimidinas/síntese química , Pirimidinas/farmacocinética , Relação Estrutura-Atividade , Células THP-1 , Macrófagos Associados a Tumor/efeitos dos fármacos
9.
Neurooncol Adv ; 1(1): vdz005, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32642648

RESUMO

BACKGROUND: Despite the poor prognosis of triple-negative breast cancer (TNBC) brain metastases, there are no approved systemic therapies. We explored the DNA-damaging poly(ADP-ribose) polymerase inhibitor (PARPi) niraparib in intracranial mouse models of breast cancer susceptibility protein (BRCA)-mutant TNBC. METHODS: Mice bearing intracranial human-derived TNBC cell lines (SUM149, MDA-MB-231Br, or MDA-MB-436) were treated with niraparib and monitored for survival; intracranial tissues were analyzed for PAR levels and niraparib concentration by mass spectrometry. RNASeq data of primary breast cancers using The Cancer Genome Atlas were analyzed for DNA damage signatures. Combined RAD51 and PARP inhibition in TNBC cell lines was assessed in vitro by colony-forming assays. RESULTS: Daily niraparib increased median survival and decreased tumor burden in the BRCA-mutant MDA-MB-436 model, but not in the BRCA-mutant SUM149 or BRCA-wild-type MDA-MB-231Br models despite high concentrations in intracranial tumors. RAD51 inhibitor B02 was shown to sensitize all cell lines to PARP inhibition (PARPi). In the analysis of BRCA-mutant primary human TNBCs, gene expression predictors of PARPi sensitivity and DNA repair signatures demonstrate widespread heterogeneity, which may explain the differential response to PARPi. Interestingly, these signatures are significantly correlated to RAD51 expression including PARPi sensitivity (R 2 = 0.602, R 2= 0.758). CONCLUSIONS: Niraparib penetrates intracranial tumor tissues in mouse models of TNBC with impressive single-agent efficacy in BRCA-mutant MDA-MB-436. Clinical evaluation of niraparib to treat TNBC brain metastases, an unmet clinical need desperate for improved therapies, is warranted. Further compromising DNA repair through RAD51 inhibition may further augment TNBC's response to PARPi.

10.
Sci Rep ; 9(1): 1853, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755715

RESUMO

PARP inhibitors have been proven clinically efficacious in platinum-responsive ovarian cancer regardless of BRCA1/2 status and in breast cancers with germline BRCA1/2 mutation. However, resistance to PARP inhibitors may preexist or evolve during treatment in many cancer types and may be overcome by combining PARP inhibitors with other therapies, such as immune checkpoint inhibitors, which confer durable responses and are rapidly becoming the standard of care for multiple tumor types. This study investigated the therapeutic potential of combining niraparib, a highly selective PARP1/2 inhibitor, with anti-PD-1 immune checkpoint inhibitors in preclinical tumor models. Our results indicate that niraparib treatment increases the activity of the type I (alpha) and type II (gamma) interferon pathways and enhances the infiltration of CD8+ cells and CD4+ cells in tumors. When coadministered in immunocompetent models, the combination of niraparib and anti-PD-1 demonstrated synergistic antitumor activities in both BRCA-proficient and BRCA-deficient tumors. Interestingly, mice with tumors cured by niraparib monotherapy completely rejected tumor growth upon rechallenge with the same tumor cell line, suggesting the potential establishment of immune memory in animals treated with niraparib monotherapy. Taken together, our findings uncovered immunomodulatory effects of niraparib that may sensitize tumors to immune checkpoint blockade therapies.


Assuntos
Neoplasias da Mama/imunologia , Indazóis/farmacologia , Interferons/imunologia , Neoplasias Ovarianas/imunologia , Piperidinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Anticorpos/imunologia , Proteína BRCA1/genética , Proteína BRCA2/genética , Sequência de Bases , Neoplasias da Mama/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Humanos , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Transplante de Neoplasias , Neoplasias Ovarianas/terapia
11.
Cancer Res ; 78(3): 742-757, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29180466

RESUMO

PARP inhibitors (PARPi) benefit only a fraction of breast cancer patients. Several of those patients exhibit intrinsic/acquired resistance mechanisms that limit efficacy of PARPi monotherapy. Here we show how the efficacy of PARPi in triple-negative breast cancers (TNBC) can be expanded by targeting MYC-induced oncogenic addiction. In BRCA-mutant/sporadic TNBC patients, amplification of the MYC gene is correlated with increased expression of the homologous DNA recombination enzyme RAD51 and tumors overexpressing both genes are associated with worse overall survival. Combining MYC blockade with PARPi yielded synthetic lethality in MYC-driven TNBC cells. Using the cyclin-dependent kinase inhibitor dinaciclib, which downregulates MYC expression, we found that combination with the PARPi niraparib increased DNA damage and downregulated homologous recombination, leading to subsequent downregulation of the epithelial-mesenchymal transition and cancer stem-like cell phenotypes. Notably, dinaciclib resensitized TBNC cells, which had acquired resistance to niraparib. We found that the synthetic lethal strategy employing dinaciclib and niraparib was also highly efficacious in ovarian, prostate, pancreatic, colon, and lung cancer cells. Taken together, our results show how blunting MYC oncogene addiction can leverage cancer cell sensitivity to PARPi, facilitating the clinical use of c-myc as a predictive biomarker for this treatment.Significance: Dual targeting of MYC-regulated homologous recombination and PARP-mediated DNA repair yields potent synthetic lethality in triple-negative breast tumors and other aggressive tumors characterized by MYC overexpression. Cancer Res; 78(3); 742-57. ©2017 AACR.


Assuntos
Proteína BRCA1/genética , Proteína BRCA2/genética , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Compostos de Piridínio/farmacologia , Mutações Sintéticas Letais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Apoptose , Proliferação de Células , Óxidos N-Cíclicos , Dano ao DNA , Reparo do DNA , Quimioterapia Combinada , Feminino , Humanos , Indolizinas , Camundongos , Camundongos Nus , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Oncotarget ; 9(98): 37080-37096, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-30647846

RESUMO

Niraparib is an orally bioavailable and selective poly (ADP-ribose) polymerase (PARP)-1/-2 inhibitor approved for maintenance treatment of both BRCA mutant (mut) and BRCA wildtype (wt) adult patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancers who have demonstrated a complete or partial response to platinum-based chemotherapy. In patients without germline BRCA mutations (non-gBRCAmut), niraparib improved progression-free survival (PFS) by 5.4 months, whereas another PARP inhibitor (PARPi) olaparib supplied only 1.9 months of improvement in a similar patient population. Previous studies revealed higher cell membrane permeability and volume of distribution (VD) as unique features of niraparib in comparison to other PARPi including olaparib. Here, we explore the potential correlation of these pharmacokinetic properties to preclinical antitumor effects in BRCAwt tumors. Our results show that at steady state, tumor exposure to niraparib is 3.3 times greater than plasma exposure in tumor xenograft mouse models. In comparison, the tumor exposure to olaparib is less than observed in plasma. In addition, niraparib crosses the blood-brain barrier and shows good sustainability in the brain, whereas sustained brain exposure to olaparib is not observed in the same models. Consistent with its favorable tumor and brain distribution, niraparib achieves more potent tumor growth inhibition than olaparib in BRCAwt models and an intracranial tumor model at maximum tolerated doses (MTD). These findings demonstrate favorable pharmacokinetic profiles and potent antitumor effects of niraparib in BRCAwt tumors, consistent with its broader clinical effect in patients with both BRCAmut and BRCAwt tumors.

13.
Cancer Res ; 78(23): 6691-6702, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30297535

RESUMO

: PARP proteins represent a class of post-translational modification enzymes with diverse cellular functions. Targeting PARPs has proven to be efficacious clinically, but exploration of the therapeutic potential of PARP inhibition has been limited to targeting poly(ADP-ribose) generating PARP, including PARP1/2/3 and tankyrases. The cancer-related functions of mono(ADP-ribose) generating PARP, including PARP6, remain largely uncharacterized. Here, we report a novel therapeutic strategy targeting PARP6 using the first reported PARP6 inhibitors. By screening a collection of PARP compounds for their ability to induce mitotic defects, we uncovered a robust correlation between PARP6 inhibition and induction of multipolar spindle (MPS) formation, which was phenocopied by PARP6 knockdown. Treatment with AZ0108, a PARP6 inhibitor with a favorable pharmacokinetic profile, potently induced the MPS phenotype, leading to apoptosis in a subset of breast cancer cells in vitro and antitumor effects in vivo. In addition, Chk1 was identified as a specific substrate of PARP6 and was further confirmed by enzymatic assays and by mass spectrometry. Furthermore, when modification of Chk1 was inhibited with AZ0108 in breast cancer cells, we observed marked upregulation of p-S345 Chk1 accompanied by defects in mitotic signaling. Together, these results establish proof-of-concept antitumor efficacy through PARP6 inhibition and highlight a novel function of PARP6 in maintaining centrosome integrity via direct ADP-ribosylation of Chk1 and modulation of its activity. SIGNIFICANCE: These findings describe a new inhibitor of PARP6 and identify a novel function of PARP6 in regulating activation of Chk1 in breast cancer cells.


Assuntos
ADP Ribose Transferases/antagonistas & inibidores , Neoplasias da Mama/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Inibidores de Poli(ADP-Ribose) Polimerases/química , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Neurosci ; 22(12): 4932-41, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12077190

RESUMO

Detection of a repellent factor, such as a semaphorin (Sema), causes localized collapse of the growth cone and directs the neurite away from the repellent. Growth cone collapse results from concomitant cytoskeletal rearrangements and detachment of adhesion sites from the extracellular matrix, via mostly unknown signaling mechanisms. In cultures of dorsal root ganglion neurons, we found that Sema3A treatment stimulates the synthesis of the eicosanoid, 12(S)-hydroxyeicosatetraenoic acid (HETE), whereas Sema3A-induced growth cone collapse is prevented when 12(S)-HETE synthesis is blocked with an inhibitor of 12/15-lipoxygenase (LO). Exogenously applied product of 12/15-LO, 12(S)-HETE, mimics Sema3A-induced collapse. As observed by interference reflection and confocal microscopy, 12(S)-HETE causes the loss of growth cone adhesion sites. The adhesion site effect seems partially independent of the actin cytoskeleton because growth cones treated with Sema3A and 12/15-LO inhibitor remain spread despite actin cytoskeleton loss. These studies demonstrate that 12/15-LO activity is a necessary step in Sema3A collapse signaling in growth cones and suggest a mechanism for its action.


Assuntos
Araquidonato 12-Lipoxigenase/fisiologia , Araquidonato 15-Lipoxigenase/fisiologia , Glicoproteínas/farmacologia , Cones de Crescimento/enzimologia , Cones de Crescimento/fisiologia , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biossíntese , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/ultraestrutura , Proteínas do Tecido Nervoso/análise , Neuropilina-1 , Ratos , Ratos Sprague-Dawley , Semaforina-3A
15.
J Med Chem ; 57(23): 9958-70, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25458601

RESUMO

KIFC1 (HSET), a member of the kinesin-14 family of motor proteins, plays an essential role in centrosomal bundling in cancer cells, but its function is not required for normal diploid cell division. To explore the potential of KIFC1 as a therapeutic target for human cancers, a series of potent KIFC1 inhibitors featuring a phenylalanine scaffold was developed from hits identified through high-throughput screening (HTS). Optimization of the initial hits combined both design-synthesis-test cycles and an integrated high-throughput synthesis and biochemical screening method. An important aspect of this integrated method was the utilization of DMSO stock solutions of compounds registered in the corporate compound collection as synthetic reactants. Using this method, over 1500 compounds selected for structural diversity were quickly assembled in assay-ready 384-well plates and were directly tested after the necessary dilutions. Our efforts led to the discovery of a potent KIFC1 inhibitor, AZ82, which demonstrated the desired centrosome declustering mode of action in cell studies.


Assuntos
Alanina/análogos & derivados , Cinesinas/antagonistas & inibidores , Piridinas/síntese química , Alanina/síntese química , Alanina/farmacologia , Animais , Células HeLa , Ensaios de Triagem em Larga Escala/métodos , Humanos , Concentração Inibidora 50 , Camundongos , Fenilalanina/análogos & derivados , Piridinas/farmacologia , Ratos , Relação Estrutura-Atividade
16.
ACS Chem Biol ; 8(10): 2201-8, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23895133

RESUMO

Centrosome amplification is observed in many human cancers and has been proposed to be a driver of both genetic instability and tumorigenesis. Cancer cells have evolved mechanisms to bundle multiple centrosomes into two spindle poles to avoid multipolar mitosis that can lead to chromosomal segregation defects and eventually cell death. KIFC1, a kinesin-14 family protein, plays an essential role in centrosomal bundling in cancer cells, but its function is not required for normal diploid cell division, suggesting that KIFC1 is an attractive therapeutic target for human cancers. To this end, we have identified the first reported small molecule inhibitor AZ82 for KIFC1. AZ82 bound specifically to the KIFC1/microtubule (MT) binary complex and inhibited the MT-stimulated KIFC1 enzymatic activity in an ATP-competitive and MT-noncompetitive manner with a Ki of 0.043 µM. AZ82 effectively engaged with the minus end-directed KIFC1 motor inside cells to reverse the monopolar spindle phenotype induced by the inhibition of the plus end-directed kinesin Eg5. Treatment with AZ82 caused centrosome declustering in BT-549 breast cancer cells with amplified centrosomes. Consistent with genetic studies, our data confirmed that KIFC1 inhibition by a small molecule holds promise for targeting cancer cells with amplified centrosomes and provided evidence that functional suppression of KIFC1 by inhibiting its enzymatic activity could be an effective means for developing cancer therapeutics.


Assuntos
Alanina/análogos & derivados , Descoberta de Drogas , Cinesinas/antagonistas & inibidores , Piridinas/química , Piridinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Alanina/química , Alanina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Modelos Moleculares
17.
Cell Cycle ; 6(20): 2531-40, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17704647

RESUMO

The IkappaB kinase (IKK) complex controls processes such as inflammation, immune responses, cell survival and the proliferation of both normal and tumor cells. By activating NFkappaB, the IKK complex contributes to G1/S transition and first evidence has been presented that IKKalpha also regulates entry into mitosis. At what stage IKK is required and whether IKK also contributes to progression through mitosis and cytokinesis, however, has not yet been determined. In this study, we use BMS-345541, a potent allosteric small molecule inhibitor of IKK, to inhibit IKK specifically during G2 and during mitosis. We show that BMS-345541 affects several mitotic cell cycle transitions, including mitotic entry, prometaphase to anaphase progression and cytokinesis. Adding BMS-345541 to the cells released from arrest in S-phase blocked the activation of Aurora A, B and C, Cdk1 activation and histone H3 phosphorylation. Additionally, treatment of the mitotic cells with BMS-345541 resulted in precocious cyclin B1 and securin degradation, defective chromosome separation and improper cytokinesis. BMS-345541 was also found to override the spindle checkpoint in nocodazole-arrested cells. In vitro kinase assays using BMS-345541 indicate that these effects are not primarily due to a direct inhibitory effect of BMS-345541 on mitotic kinases such as Cdk1, Aurora A or B, Plk1 or NEK2. This study points towards a new potential role of IKK in cell cycle progression. Since deregulation of the cell cycle is one of the hallmarks of tumor formation and progression, the newly discovered level of BMS-345541 function could be useful for cell cycle control studies and may provide valuable clues for the design of future therapeutics.


Assuntos
Quinase I-kappa B/metabolismo , Imidazóis/farmacologia , Mitose/efeitos dos fármacos , Quinoxalinas/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cromossomos de Mamíferos/genética , Ciclina A/metabolismo , Ciclina B/metabolismo , Ciclina B1 , Humanos , Fuso Acromático/metabolismo , Telomerase/genética , Telomerase/metabolismo
18.
Cell Cycle ; 5(17): 2029-35, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16969131

RESUMO

Most efforts thus far have been devoted to develop apoptosis inducers for cancer treatment. However, apoptotic pathway deficiencies are a hallmark of cancer cells. We propose that one way to bypass defective apoptotic pathways in cancer cells is to induce necrotic cell death. Here we show that selective induction of necrotic cell death can be achieved by activation of the DNA damage response pathways. While beta-lapachone induces apoptosis through E2F1 checkpoint pathways, necrotic cell death can be selectively induced by beta-lapachone in a variety of cancer cells. We found that beta-lapachone, unlike DNA damaging chemotherapeutic agents, transiently activates PARP1, a main regulator of the DNA damage response pathway, both in vitro and in vivo. This occurs within minutes of exposure to beta-lapachone, resulting in selective necrotic cell death. Inhibition of PAR blocked beta-lapachone-induced necrosis. Furthermore, necrotic cell death induced by beta-lapachone was significantly reduced in PARP1 knockout cell lines. Our data suggest that selective necrotic cell death can be induced through activation of DNA damage response pathways, supporting the idea of selective necrotic cell death as a therapeutic strategy to eliminate cancer cells.


Assuntos
Antineoplásicos/toxicidade , Dano ao DNA , Naftoquinonas/toxicidade , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Necrose , Neoplasias/metabolismo , Neoplasias/patologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Biol Chem ; 278(23): 21168-77, 2003 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-12665507

RESUMO

Exposure of growing neurons to thrombin or semaphorin 3A stimulates a receptor-mediated signaling cascade that results in collapse of their growth cones. This collapse response necessitates eicosanoid production, as we have shown earlier. The present report investigates whether and which protein kinase C (PKC) isoforms may be activated by such eicosanoids. To examine these questions, we isolated growth cones from fetal rat brain and tested whether thrombin or the eicosanoid, 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE), could activate endogenous growth cone PKC. We show that both thrombin and 12(S)-HETE stimulate the phosphorylation of the myristoylated alanine-rich protein kinase C substrate, an 87-kDa adhesion site protein. Furthermore, we show both with immunoprecipitated and with recombinant PKC that 12(S)-HETE activation is selective for the epsilon isoform and does not require accessory proteins. Last, we demonstrate that PKC activation is necessary for thrombin-induced growth cone collapse. These data indicate that eicosanoid-mediated repellent effects result from the direct and selective activation of PKCepsilon and suggest the involvement of myristoylated alanine-rich protein kinase C substrate phosphorylation in growth cone collapse.


Assuntos
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacologia , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Proteína Quinase C/metabolismo , Animais , Adesão Celular/fisiologia , Ativação Enzimática/efeitos dos fármacos , Hemostáticos/farmacologia , Substrato Quinase C Rico em Alanina Miristoilada , Neurônios/enzimologia , Neurônios/ultraestrutura , Fosforilação , Testes de Precipitina , Proteína Quinase C/genética , Proteína Quinase C/isolamento & purificação , Proteína Quinase C-épsilon , Proteínas/metabolismo , Ratos , Proteínas Recombinantes/metabolismo , Trombina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA