Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Ann Oncol ; 31(5): 626-633, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32205017

RESUMO

BACKGROUND: This phase Ib study evaluated the safety, tolerability, pharmacokinetics, and preliminary efficacy of the oral AKT inhibitor ipatasertib and chemotherapy or hormonal therapy in patients with advanced or metastatic solid tumors to determine combined dose-limiting toxicities (DLTs), maximum tolerated dose, and recommended phase II doses and schedules. PATIENTS AND METHODS: The clinical study comprised four combination treatment arms: arm A (with docetaxel), arm B [with mFOLFOX6 (modified leucovorin, 5-fluorouracil, and oxaliplatin)], arm C (with paclitaxel), and arm D (with enzalutamide). Primary endpoints were safety and tolerability; secondary endpoints were pharmacokinetics, clinical activity per Response Evaluation Criteria in Solid Tumors v1.1, and prostate-specific antigen levels. RESULTS: In total, 122 patients were enrolled. Common adverse events were diarrhea, nausea, vomiting, decreased appetite, and fatigue. The safety profiles of the combination regimens were consistent with those of the background regimens, except for diarrhea, hyperglycemia, and rash, which were previously observed with ipatasertib treatment. The only combination DLT across all treatment arms was one event of grade 3 dehydration (ipatasertib 600 mg and paclitaxel). Recommended phase II doses for ipatasertib were 600 mg (and mFOLFOX6) and 400 mg (and paclitaxel), respectively. The maximum assessed dose of ipatasertib 600 mg combined with docetaxel or enzalutamide was well tolerated. Coadministration with enzalutamide (a cytochrome P450 3A inducer) resulted in approximately 50% lower ipatasertib exposure. CONCLUSIONS: Ipatasertib in combination with chemotherapy or hormonal therapy was well tolerated with a safety profile consistent with that of ATP-competitive AKT inhibitors. CLINICAL TRIAL NUMBER: NCT01362374.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Humanos , Masculino , Dose Máxima Tolerável , Neoplasias/tratamento farmacológico , Piperazinas , Pirimidinas/uso terapêutico
2.
Oncogene ; 36(37): 5309-5320, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28534509

RESUMO

Tumor recurrence in glioblastoma (GBM) is, in part, attributed to increased epithelial-to-mesenchymal transition (EMT) and enhanced tumor cell dissemination in adjacent brain parenchyma after ionizing radiation (IR). EMT is associated with aggressive behavior, increased stem-like characteristics and treatment resistance in malignancies; however, the underlying signaling mechanisms that regulate EMT are poorly understood. We identified grade-dependent p21-activated kinases 4 (PAK4) upregulation in gliomas and further determined its role in mesenchymal transition and radioresistance. IR treatment significantly elevated expression and nuclear localization of PAK4 in correlation with induction of reactive oxygen species (ROS) and mesenchymal transition in GBM cells. Stable PAK4 overexpression promoted mesenchymal transition by elevating EMT marker expression in these cells. Of note, transcription factor-DNA-binding arrays and chromatin immunoprecipitation experiments identified the formation of a novel nuclear PAK4/PPARγ complex which was recruited to the promoter of Nox1, a peroxisome proliferator-activated receptor gamma (PPARγ) target gene. In addition, IR further elevated PAK4/PPARγ complex co-recruitment to Nox1 promoter, and increased Nox1 expression and ROS levels associated with mesenchymal transition in these cells. Conversely, specific PAK4 downregulation decreased PPARγ-mediated Nox1 expression and suppressed EMT in IR-treated cells. In vivo orthotopic tumor experiments showed inhibition of growth and suppression of IR-induced PPARγ and Nox1 expression by PAK4 downregulation in tumors. Our results provide the first evidence of a novel role for PAK4 in IR-induced EMT and suggest potential therapeutic efficacy of targeting PAK4 to overcome radioresistance in gliomas.


Assuntos
Neoplasias Encefálicas/patologia , Transição Epitelial-Mesenquimal/efeitos da radiação , Glioma/patologia , NADPH Oxidases/metabolismo , PPAR gama/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , NADPH Oxidase 1 , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , PPAR gama/genética , Regiões Promotoras Genéticas , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transfecção , Quinases Ativadas por p21/genética
3.
Cardiovasc Toxicol ; 17(3): 307-318, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27783203

RESUMO

Nicotinamide phosphoribosyltransferase (NAMPT) is a pleiotropic protein that functions as an enzyme, cytokine, growth factor and hormone. As a target for oncology, NAMPT is particularly attractive, because it catalyzes the rate-limiting step in the salvage pathway to generate nicotinamide adenine dinucleotide (NAD), a universal energy- and signal-carrying molecule involved in cellular energy metabolism and many homeostatic functions. Inhibition of NAMPT generally results in NAD depletion, followed by ATP reduction and loss of cell viability. Herein, we describe NAMPT inhibitor (NAMPTi)-induced cardiac toxicity in rodents following short-term administration (2-7 days) of NAMPTi's. The cardiac toxicity was interpreted as a functional effect leading to congestive heart failure, characterized by sudden death, thoracic and abdominal effusion, and myocardial degeneration. Based on exposures in the initial in vivo safety rodent studies and cardiotoxicity observed, we conducted studies in rat and human in vitro cardiomyocyte cell systems. Based on those results, combined with human cell line potency data, we demonstrated the toxicity is both on-target and likely human relevant. This toxicity was mitigated in vitro by co-administration of nicotinic acid (NA), which can enable NAD production through the NAMPT-independent pathway; however, this resulted in only partial mitigation in in vivo studies. This work also highlights the usefulness and predictivity of in vitro cardiomyocyte assays using human cells to rank-order compounds against potency in cell-based pharmacology assays. Lastly, this work strengthens the correlation between cardiomyocyte cell viability and functionality, suggesting that these assays together may enable early assessment of cardiotoxicity in vitro prior to conduct of in vivo studies and potentially reduce subsequent attrition due to cardiotoxicity.


Assuntos
Citocinas/antagonistas & inibidores , Inibidores Enzimáticos/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Animais , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/enzimologia , Feminino , Compostos Heterocíclicos com 2 Anéis/toxicidade , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/enzimologia , Masculino , Nicotinamida Fosforribosiltransferase/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Sulfonas/toxicidade
4.
Leukemia ; 30(6): 1335-43, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26965285

RESUMO

Chronic and juvenile myelomonocytic leukemias (CMML and JMML) are myelodysplastic/myeloproliferative neoplasia (MDS/MPN) overlap syndromes that respond poorly to conventional treatments. Aberrant Ras activation because of NRAS, KRAS, PTPN11, CBL and NF1 mutations is common in CMML and JMML. However, no mechanism-based treatments currently exist for cancers with any of these mutations. An alternative therapeutic strategy involves targeting Ras-regulated effector pathways that are aberrantly activated in CMML and JMML, which include the Raf/MEK/ERK and phosphoinositide-3'-OH kinase (PI3K)/Akt cascades. Mx1-Cre, Kras(D12) and Mx1-Cre, Nf1(flox/)(-) mice accurately model many aspects of CMML and JMML. Treating Mx1-Cre, Kras(D12) mice with GDC-0941 (also referred to as pictilisib), an orally bioavailable inhibitor of class I PI3K isoforms, reduced leukocytosis, anemia and splenomegaly while extending survival. However, GDC-0941 treatment attenuated activation of both PI3K/Akt and Raf/MEK/ERK pathways in primary hematopoietic cells, suggesting it could be acting through suppression of Raf/MEK/ERK signals. To interrogate the importance of the PI3K/Akt pathway specifically, we treated mice with the allosteric Akt inhibitor MK-2206. This compound had no effect on Raf/MEK/ERK signaling, yet it also induced robust hematologic responses in Kras and Nf1 mice with MPN. These data support investigating PI3K/Akt pathway inhibitors as a therapeutic strategy in JMML and CMML patients.


Assuntos
Compostos Heterocíclicos com 3 Anéis/farmacologia , Síndromes Mielodisplásicas/metabolismo , Transtornos Mieloproliferativos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas ras/genética , Animais , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Indazóis , Leucemia Mielomonocítica Crônica , Leucemia Mielomonocítica Juvenil , Sistema de Sinalização das MAP Quinases , Camundongos , Síndromes Mielodisplásicas/tratamento farmacológico , Transtornos Mieloproliferativos/tratamento farmacológico , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas
5.
Cell Death Dis ; 6: e1590, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25590800

RESUMO

The anti-apoptotic protein MCL-1 is a key regulator of cancer cell survival and a known resistance factor for small-molecule BCL-2 family inhibitors such as ABT-263 (navitoclax), making it an attractive therapeutic target. However, directly inhibiting this target requires the disruption of high-affinity protein-protein interactions, and therefore designing small molecules potent enough to inhibit MCL-1 in cells has proven extremely challenging. Here, we describe a series of indole-2-carboxylic acids, exemplified by the compound A-1210477, that bind to MCL-1 selectively and with sufficient affinity to disrupt MCL-1-BIM complexes in living cells. A-1210477 induces the hallmarks of intrinsic apoptosis and demonstrates single agent killing of multiple myeloma and non-small cell lung cancer cell lines demonstrated to be MCL-1 dependent by BH3 profiling or siRNA rescue experiments. As predicted, A-1210477 synergizes with the BCL-2/BCL-XL inhibitor navitoclax to kill a variety of cancer cell lines. This work represents the first description of small-molecule MCL-1 inhibitors with sufficient potency to induce clear on-target cellular activity. It also demonstrates the utility of these molecules as chemical tools for dissecting the basic biology of MCL-1 and the promise of small-molecule MCL-1 inhibitors as potential therapeutics for the treatment of cancer.


Assuntos
Compostos de Anilina/farmacologia , Apoptose/efeitos dos fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides/antagonistas & inibidores , Neoplasias/patologia , Bibliotecas de Moléculas Pequenas/farmacologia , Sulfonamidas/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Ácidos Carboxílicos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Indóis/farmacologia , Proteínas de Membrana/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
7.
Oncogene ; 32(26): 3119-29, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-22945648

RESUMO

Tumor stem cells, postulated to be the source cells for malignancies, have been identified in several cancers using cell-surface expression of markers including CD133, a pentaspan membrane protein. CD133+ve cells form neurospheres, exhibit self-renewal and differentiation, and are tumorigenic. However, despite its association with stem cells, a causal relationship of CD133 to tumorigenesis remains to be defined. Hypothesizing that specific epigenetic and transcription factors implicated in driving the stem cell state may concurrently regulate CD133 expression in stem cells, we analyzed the structure and regulation of CD133 promoter in glioma stem cells and glioma cell lines. Initially, a minimal promoter region was identified by analyzing the activity of CD133 promoter-driven luciferase-expressing 5'-and 3'-deletion-constructs upstream of the transcription start site. This region contained a CpG island that was hypermethylated in CD133-ve glioma stem cells (GSC) and glioma cells but unmethylated in CD133+ve ones. Of several predicted TF-binding sites in this region, the role of tandem Sp1 (-242 and -221) and two Myc (-541 and -25)-binding sites were examined. Overexpression of Sp1 or Myc increased CD133 minimal promoter-driven luciferase activity and CD133 levels in GSC and in glioma cell line. Mithramycin, a Sp1 inhibitor, decreased minimal promoter activity and downregulated CD133 levels in GSC. Gel-shift assays demonstrated direct binding of Sp1 to their predicted sites that was competitively inhibited by oligonucleotide-binding-site sequences and supershifted by anti-Sp1 confirming the interaction. Sp1 and Myc-antibody chromatin immunoprecipitation (ChIP) analysis in GSC showed enrichment of regions with Sp1 and Myc-binding sites. In CD133-ve cells, ChIP analysis showed binding of the methyl-DNA-binding proteins, MBD1, MBD2 and MeCP2 to the methylated CpG island and repression of transcription. These results demonstrate that Sp1 and Myc regulate CD133 transcription in GSC and that promoter methylation and methyl-DNA-binding proteins cause repression of CD133 by excluding transcription-factor binding.


Assuntos
Antígenos CD/genética , Epigênese Genética , Glioma/genética , Glicoproteínas/genética , Células-Tronco Neoplásicas/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Peptídeos/genética , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/metabolismo , Antígeno AC133 , Antígenos CD/metabolismo , Sequência de Bases , Diferenciação Celular , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Glicoproteínas/metabolismo , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Proteína Oncogênica p55(v-myc)/imunologia , Peptídeos/metabolismo , Plicamicina/farmacologia , Análise de Sequência de DNA , Fator de Transcrição Sp1/antagonistas & inibidores , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/imunologia , Fatores de Transcrição/metabolismo , Transcrição Gênica
8.
Cell Death Dis ; 2: e121, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21368892

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in malignant cells, including gliomas, and is currently in anticancer clinical trials. However, the full-length and tagged forms of TRAIL, unlike the untagged ligand (soluble TRAIL (sTRAIL)), exhibits toxicity against normal cells. Here, we report the generation and testing of an adenovirus (AdsTRAIL) that expresses untagged sTRAIL in an intracranial xenograft model and a human glioma organotypic slice culture model. AdsTRAIL efficiently induced apoptosis in glioma cell lines, including those resistant to sTRAIL, but not in normal human astrocytes (NHAs). It inhibited anchorage-independent glioma growth and exerted a bystander effect in transwell assays. Intratumoral injections of AdsTRAIL in a rodent intracranial glioma model resulted in reduced tumor growth and improved survival compared with Ad-enhanced green fluorescent protein (EGFP)- or vehicle-treated controls without toxicity. Human glioma organotypic slices treated with AdsTRAIL demonstrated apoptosis induction and caspase activation.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/genética , Glioma/terapia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adenoviridae/metabolismo , Animais , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Secções Congeladas , Terapia Genética/instrumentação , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glioma/metabolismo , Glioma/fisiopatologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico
9.
Leukemia ; 24(11): 1893-900, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20882050

RESUMO

Ligation of CD40 on chronic lymphocytic leukemia (CLL) cells induces phenotypic and biochemical changes that facilitate CLL cell-T cell interactions and enhances the sensitivity of CLL cells to clearance by adaptive and innate immune-effector mechanisms. CLL cells can be transduced to express CD40 ligand (CD154) using a replication-defective adenovirus vector, thereby cross-linking CD40 on transduced and non-transduced, bystander CLL cells. In a previous study, patients received infusions of autologous CLL cells, transduced to express murine CD154 (mCD154), which induced anti-leukemic immune responses, but also anti-mCD154 antibodies. In this study, we report a phase I study, in which patients were infused with 1 × 10(8), 3 × 10(8) or 1 × 10(9) autologous CLL cells transduced ex vivo to express ISF35, a humanized, membrane-stable CD154. Infusions were well tolerated and consistently followed by reductions in blood lymphocyte counts and lymphadenopathy. After infusion, circulating CLL cells had enhanced or de novo expression of CD95, DR5, p73 and Bid, which enhanced their susceptibility to death-receptor-mediated or drug-induced apoptosis, including CLL cells with deletions at 17p13.1 (del(17p)). Two patients who had CLL with del(17p) had subsequent chemoimmunotherapy and responded well to treatment. In summary, infusions of autologous, ISF35-transduced CLL cells were well tolerated, had biological and clinical activity, and might enhance the susceptibility of CLL cells with del(17p) to chemoimmunotherapy.


Assuntos
Ligante de CD40/uso terapêutico , Terapia Genética/métodos , Leucemia Linfocítica Crônica de Células B/terapia , Adulto , Idoso , Antígenos CD40/imunologia , Ligante de CD40/efeitos adversos , Cromossomos Humanos Par 17/genética , Feminino , Terapia Genética/efeitos adversos , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Microscopia de Fluorescência/métodos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Deleção de Sequência , Receptor fas/imunologia
10.
Oncogene ; 27(50): 6522-37, 2008 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-18955977

RESUMO

Nucleoside analogs are structurally similar antimetabolites that have a broad range of action and are clinically active in both solid tumors and hematological malignancies. Many of these agents are incorporated into DNA by polymerases during normal DNA synthesis, an action that blocks further extension of the nascent strand and causes stalling of replication forks. The molecular mechanisms that sense stalled replication forks activate cell cycle checkpoints and DNA repair processes, which may contribute to drug resistance. When replication forks are not stabilized by these molecules or when subsequent DNA repair processes are overwhelmed, apoptosis is initiated either by these same DNA damage sensors or by alternative mechanisms. Recently, strategies aimed at targeting DNA damage checkpoints or DNA repair processes have demonstrated effectiveness in sensitizing cells to nucleoside analogs, thus offering a means to elude drug resistance. In addition to their DNA synthesis-directed actions many nucleoside analogs trigger apoptosis by unique mechanisms, such as causing epigenetic modifications or by direct activation of the apoptosome. A review of the cellular and molecular responses to clinically relevant agents provides an understanding of the mechanisms that cause apoptosis and may provide rationale for the development of novel therapeutic strategies.


Assuntos
Apoptose/efeitos dos fármacos , Nucleosídeos/farmacologia , Transdução de Sinais , Animais , Dano ao DNA , Metilação de DNA , Reparo do DNA , Replicação do DNA , Humanos , Nucleosídeos/química , Nucleosídeos/metabolismo
11.
Apoptosis ; 10(1): 233-43, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15711939

RESUMO

Patients with malignant gliomas have a poor prognosis and new treatment paradigms are needed against this disease. TRAIL/Apo2L selectively induces apoptosis in malignant cells sparing normal cells and is hence of interest as a potential therapeutic agent against gliomas. To determine the factors that modulate sensitivity to TRAIL, we examined the differences in TRAIL-activated signaling pathways in glioma cells with variable sensitivities to the agent. Apoptosis in response to TRAIL was unrelated to DR5 expression or endogenous p53 status in a panel of 8 glioma cell lines. TRAIL activated the extrinsic (cleavage of caspase-8, caspase-3 and PARP) and mitochondrial apoptotic pathways and reduced FLIP levels. It also induced caspase-dependent JNK activation, which did not influence TRAIL-induced apoptosis. Because the pro-survival PI3K/Akt pathway is highly relevant to gliomas, we assessed whether Akt could protect against TRAIL-induced apoptosis. Pretreatment with SH-6, a novel Akt inhibitor, enhanced TRAIL-induced apoptosis, suggesting a protective role for Akt. Conversely, TRAIL induced caspase-dependent cleavage of Akt neutralizing its anti-apoptotic effects. These results demonstrate that TRAIL-induced apoptosis in gliomas involves both activation of death pathways and downregulation of survival pathways. Additional studies are warranted to determine the therapeutic potential of TRAIL against gliomas.


Assuntos
Apoptose , Glioma/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Glicoproteínas de Membrana/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , MAP Quinase Quinase 4 , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF
12.
Curr Opin Oncol ; 13(6): 484-90, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11673689

RESUMO

The mammalian cell cycle is exquisitely controlled by the cyclin-dependent kinases, which regulate cell cycle progression. Cell cycle transitions are, in turn, controlled by checkpoints that monitor the integrity and replication status of the genetic material before cells commit to either replicate or segregate their DNA. On activation, checkpoints interface with cyclin-Cdk complexes to block the cell cycle. Pharmacologic compounds that exploit our current knowledge of cell cycle and checkpoint pathway regulation offer insights into the development of novel therapeutic strategies.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Replicação do DNA , Desenho de Fármacos , Animais , Dano ao DNA , Humanos , Mamíferos , Neoplasias/tratamento farmacológico
13.
Endocrinology ; 142(6): 2540-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356703

RESUMO

Cyr61, a member of the CCN (CTGF/Cyr61/NOV) family of growth regulators, is a secreted cysteine-rich proangiogenic factor that has been implicated in tumorigenesis. Previous studies have also demonstrated that Cyr61 is regulated by 17beta-estradiol (E(2)) in the uterus. Therefore, we hypothesized that hormonal regulation of Cyr61 may be important in estrogen-dependent pathogenic processes such as breast tumorigenesis. Our study demonstrates that both Cyr61 messenger RNA and protein are induced by E(2) in MCF-7 mammary adenocarcinoma cells that primarily overexpress estrogen receptor alpha (ERalpha) in a dose-dependent and immediate early fashion. Cyr61 gene induction by E(2) is transcriptionally regulated by ERalpha as the antiestrogen, ICI 182,780, and actinomycin D blocked induction completely. In addition, Cyr61 is up-regulated in MCF-7 cells by epidermal growth factor (EGF) in an immediate early fashion as well. The functional relevance of steroid induction of Cyr61 in breast cancer cell growth is demonstrated by anti-Cyr61 neutralizing antibodies, which diminished E(2) and EGF-dependent DNA synthesis and dramatically reduced E(2)-driven cell proliferation by more than 70%. Most importantly, Cyr61 is overexpressed in 70% (28 of 40) of breast cancer patients with infiltrating ductal carcinoma and is localized exclusively to hyperplastic ductal epithelial cells. Moreover, the levels of Cyr61 protein are higher in breast tumors that are ER(+)/EGF receptor(+) than those that are ER(-)/EGF receptor(+), suggesting that estrogens may mediate Cyr61 expression in vivo. Collectively, our data suggest that Cyr61 may play a critical role in estrogen- as well as growth factor-dependent breast tumor growth.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Divisão Celular , Estradiol/análogos & derivados , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Substâncias de Crescimento/genética , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular , Adenocarcinoma/genética , Adenocarcinoma/patologia , Anticorpos/farmacologia , Proteína Rica em Cisteína 61 , DNA de Neoplasias/biossíntese , Dactinomicina/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Fulvestranto , Substâncias de Crescimento/imunologia , Substâncias de Crescimento/fisiologia , Humanos , Proteínas Imediatamente Precoces/imunologia , Proteínas Imediatamente Precoces/fisiologia , Hibridização In Situ , RNA Mensageiro/análise , Receptores de Estrogênio/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ativação Transcricional , Células Tumorais Cultivadas
14.
J Clin Endocrinol Metab ; 86(4): 1707-15, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11297607

RESUMO

Uterine leiomyomas are the most common tumors of the reproductive tract, afflicting women between the ages of 30--55 yr. Although considered to be the leading cause of hysterectomies in the United States, little is known of the etiology and mechanisms of pathogenesis in leiomyomas. Accordingly, rapid analysis of differential expression (RADE) was employed to identify genes that are abnormally expressed in leiomyomas. Of the several genes identified, Cyr61, a member of the CCN family of growth and angiogenic regulators, was shown to be markedly down-regulated at the messenger ribonucleic acid (mRNA) and protein levels in leiomyoma tumors compared with the matched uterine myometrial controls (n = 38). In addition, in situ hybridization experiments corroborated the lack of Cyr61 expression in leiomyoma cells, whereas abundant transcript levels were identified in adjacent myometrial smooth muscle cells. To elucidate the mechanisms of Cyr61 gene regulation in leiomyomas, we determined the effects of ovarian steroids, basic fibroblast growth factor (bFGF), and serum, on Cyr61 expression using an ex vivo culture system. Treatment of human myometrial explants with 17 beta-estradiol and bFGF up-regulated Cyr61 transcripts, whereas the progesterone receptor agonist, R5020 (alone or in combination with 17 beta-estradiol), had no effect. Paradoxically, neither 17 beta-estradiol nor bFGF was capable of up-regulating Cyr61 mRNA in leiomyoma explants despite elevated levels of ER alpha mRNA, suggesting a possible defect in steroid and growth factor regulation. Thus, dysregulation of Cyr61 by estrogen and bFGF may contribute to down-regulation of Cyr61 in leiomyomas, which, in turn, may predispose uterine smooth muscle cells toward sustained growth.


Assuntos
Estradiol/fisiologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Substâncias de Crescimento/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Leiomioma/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo , Proteína Rica em Cisteína 61 , Regulação para Baixo , Estradiol/farmacologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Substâncias de Crescimento/genética , Humanos , Proteínas Imediatamente Precoces/genética , Leiomioma/patologia , Pessoa de Meia-Idade , Músculo Liso/metabolismo , Músculo Liso/patologia , Miométrio/citologia , Miométrio/metabolismo , Proteína Sobre-Expressa em Nefroblastoma , RNA Mensageiro/metabolismo , Valores de Referência , Distribuição Tecidual , Regulação para Cima , Neoplasias Uterinas/patologia
15.
Cancer Res ; 61(3): 1065-72, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11221834

RESUMO

The mechanisms of resistance to nucleoside analogues established in preclinical models are rarely found in primary tumors resistant to therapy with these agents. We tested the hypothesis that cells sense sublethal incorporation of analogues into DNA during replication and react by arresting further DNA synthesis and cell cycle progression. After removal of drug, cells may be able to repair damaged DNA and continue proliferation, thus escaping nucleoside analogue toxicity. As a corollary, we evaluated whether dysregulation of this mechanism causes cell death. Using gemcitabine as a model of S-phase-specific nucleoside analogues in human acute myelogenous leukemia ML-1 cells, we found that DNA synthesis decreased, cells arrested in S-phase transit, and 60-70% of the population accumulated in S-phase in response to cytostatic conditions. Proliferation continued after washing the cells into drug-free medium. S-phase-arrested cells were then treated with otherwise nontoxic concentrations of UCN-01, which caused rapid onset of apoptosis without cell cycle progression specifically in cells with an S-phase DNA content. Thus, S-phase arrest by nucleoside analogues sensitizes cells to UCN-01, which appears to activate signaling for death mechanisms and/or inhibit survival pathways. These results differ from those in cells arrested at the G2 checkpoint, in which UCN-01 abrogates cell cycle arrest, permitting cells to progress in the cell cycle before apoptosis.


Assuntos
Alcaloides/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Desoxicitidina/farmacologia , Fase S/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Estaurosporina/análogos & derivados , Células Tumorais Cultivadas , Gencitabina
16.
Cancer Res ; 60(22): 6408-15, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11103806

RESUMO

Activation of the c-Jun NH2-terminal kinase type 1 (JNK1) signaling pathway is often associated with apoptosis. In this report, we elucidated the role of this kinase in the programmed cell death induced by the nucleoside analogue 9-beta-D-arabinosyl-2-fluoroadenine (F-ara-A). Treatment of ML-1 cells with 3 or 10 microM F-ara-A specifically killed cells in the S-phase of the population. Incorporation of F-ara-ATP, the nucleoside triphosphate of F-ara-A, into DNA resulted in the activation of JNK1 in a time- and dose-dependent fashion. Activation of JNK1 temporally preceded DNA fragmentation. When incorporation of F-ara-A into DNA was blocked by pretreatment of the cells with aphidicolin to inhibit DNA synthesis, neither JNK1 signaling nor apoptosis was evident. Furthermore, inhibition of JNK1 by treatment of the cells with forskolin or by pretreatment with an antisense oligonucleotide directed against JNK1 mRNA resulted in a decrease in F-ara-A-induced apoptosis. Finally, the JNK1 signaling pathway appeared to be upstream to that of the effector caspases in nucleoside analogue-induced apoptosis. Thus, our data strongly suggest that JNK1 is involved in transduction of F-ara-A-induced distress signals into an apoptotic response.


Assuntos
Antineoplásicos/farmacologia , Apoptose/fisiologia , Dano ao DNA/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfato de Vidarabina/análogos & derivados , Vidarabina/análogos & derivados , Vidarabina/farmacologia , Apoptose/efeitos dos fármacos , Arabinonucleotídeos/metabolismo , Caspases/metabolismo , DNA/efeitos dos fármacos , DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Leucemia Mieloide , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fase S/fisiologia , Células Tumorais Cultivadas , Células U937 , Vidarabina/metabolismo , Fosfato de Vidarabina/metabolismo
17.
J Clin Invest ; 103(9): 1353-61, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10225979

RESUMO

Cytokine effects on immunity and inflammation often depend on the transcription factors termed signal transducers and activators of transcription (STATs), so STAT signaling pathways are candidates for influencing inflammatory disease. We reasoned that selective IFN responsiveness of the first STAT family member (Stat1) and Stat1-dependent immune-response genes such as intercellular adhesion molecule-1 (ICAM-1), IFN regulatory factor-1 (IRF-1), and Stat1 itself in airway epithelial cells provides a basis for detecting cytokine signaling abnormalities in inflammatory airway disease. On the basis of nuclear localization and phosphorylation, we found that epithelial Stat1 (but not other control transcription factors) was invariably activated in asthmatic compared with normal control or chronic bronchitis subjects. Furthermore, epithelial levels of activated Stat1 correlated with levels of expression for epithelial ICAM-1, IRF-1, and Stat1, and in turn, ICAM-1 levels correlated with T-cell accumulation in tissue. However, only low levels of IFN-gamma or IFN-gamma-producing cells were detected in airway tissue in all subjects. The results therefore provide initial evidence linking abnormal behavior of STAT pathways for cytokine signaling to the development of an inflammatory disease. In that context, the results also change the current scheme for asthma pathogenesis to one that must include a localized gain in transcriptional signal ordinarily used for a T helper 1-type cytokine (IFN-gamma) in combination with allergy-driven overproduction of T helper 2-type cytokines.


Assuntos
Asma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Transativadores/metabolismo , Asma/imunologia , Sequência de Bases , Brônquios/citologia , Brônquios/metabolismo , Estudos de Casos e Controles , Primers do DNA , Células Epiteliais/metabolismo , Humanos , Interferon gama/metabolismo , Fator de Transcrição STAT1 , Células Th1/imunologia
18.
J Exp Med ; 187(12): 1927-40, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9625753

RESUMO

Immune cell migration into and through mucosal barrier sites in general and airway sites in particular is a critical feature of immune and inflammatory responses, but the determinants of transepithelial (unlike transendothelial) immune cell traffic are poorly defined. Accordingly, we used primary culture airway epithelial cells and peripheral blood mononuclear cells to develop a cell monolayer system that allows for apical-to-basal and basal-to-apical T cell transmigration that can be monitored with quantitative immunofluorescence flow cytometry. In this system, T cell adhesion and subsequent transmigration were blocked in both directions by monoclonal antibodies (mAbs) against lymphocyte function-associated antigen 1 (LFA-1) or intercellular adhesion molecule 1 (ICAM-1) (induced by interferon gamma [IFN-gamma] treatment of epithelial cells). The total number of adherent plus transmigrated T cells was also similar in both directions, and this pattern fit with uniform presentation of ICAM-1 along the apical and basolateral cell surfaces. However, the relative number of transmigrated to adherent T cells (i.e., the efficiency of transmigration) was increased in the basal-to-apical relative to the apical-to-basal direction, so an additional mechanism was needed to mediate directional movement towards the apical surface. Screening for epithelial-derived beta-chemokines indicated that IFN-gamma treatment caused selective expression of RANTES (regulated upon activation, normal T cell expressed and secreted), and the functional significance of this finding was demonstrated by inhibition of epithelial-T cell adhesion and transepithelial migration by anti-RANTES mAbs. In addition, we found that epithelial (but not endothelial) cells preferentially secreted RANTES through the apical cell surface thereby establishing a chemical gradient for chemotaxis across the epithelium to a site where they may be retained by high levels of RANTES and apical ICAM-1. These patterns for epithelial presentation of ICAM-1 and secretion of RANTES appear preserved in airway epithelial tissue studied either ex vivo with expression induced by IFN-gamma treatment or in vivo with endogenous expression induced by inflammatory disease (i.e., asthma). Taken together, the results define how the patterns for uniform presentation of ICAM-1 along the cell surface and specific apical sorting of RANTES may serve to mediate the level and directionality of T cell traffic through epithelium (distinct from endothelium) and provide a basis for how this process is precisely coordinated to route immune cells to the mucosal surface and maintain them there under normal and stimulated conditions.


Assuntos
Quimiocina CCL5/metabolismo , Quimiotaxia de Leucócito , Células Epiteliais/imunologia , Molécula 1 de Adesão Intercelular/biossíntese , Linfócitos T/imunologia , Traqueia/imunologia , Adesão Celular , Polaridade Celular , Células Cultivadas , Células Epiteliais/citologia , Humanos , Antígeno-1 Associado à Função Linfocitária/metabolismo , Mucosa/citologia , Mucosa/imunologia , Traqueia/citologia
19.
Proc Assoc Am Physicians ; 110(1): 1-11, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9460078

RESUMO

A major goal of our research is to understand how immune cells (especially T cells) infiltrate the pulmonary airway during host defense and inflammatory disease (especially asthma). In that context, we have proposed that epithelial cells lining the airway provide critical biochemical signals for immune-cell influx and activation and that this epithelial-immune cell interaction is a critical feature of airway inflammation and hyperreactivity. In this brief report, we describe our progress in defining a subset of epithelial immune-response genes the expression of which is coordinated for viral defense both directly in response to replicating virus and indirectly under the control of a specific interferon-gamma signal transduction pathway featuring the Stat1 transcription factor as a critical relay signal between cytoplasm and nucleus. Unexpectedly, the same pathway is also activated during asthmatic airway inflammation in a setting where there is no apparent infection and no increase in interferon-gamma levels. The findings provide the first evidence of an overactive Stat1-dependent gene network in asthmatic airways and a novel molecular link between mucosal immunity and inflammation. The findings also offer the possibility that overactivity of Stat1-dependent genes might augment a subsequent T helper cell (Th1)-type response to virus or might combine with a heightened Th2-type response to allergen to account for more severe exacerbations of asthma.


Assuntos
Asma/imunologia , Mucosa Nasal/imunologia , Animais , Células Epiteliais/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade nas Mucosas/genética , Imunidade nas Mucosas/imunologia , Inflamação , Linfócitos T/imunologia
20.
Neurochem Res ; 22(4): 351-62, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9130244

RESUMO

The rapid decreases in viability seen in H2O2-treated PC12 cells reflect enhanced susceptibility of neural cell types to oxidant injury. The dose-response relationship between NGF concentration and survival after H2O2 treatment resembles that for NGF effects on PC12 survival in serumless medium. Previously we have shown that NGF treatment enhances the activity of GSH-Px and catalase which catalyze the degradation of H2O2. Here in order to ascertain whether NGF stimulates transcription, affects mRNA stability, or acts post-transcriptionally, we measured catalase and GSH-Px mRNA half-lives. While both catalase and GSH-Px transcripts are stable with a relatively long half life and a gradual decay in mRNA levels, NGF had different effects on their stability. NGF had marked effects on catalase mRNA stability. The catalase gene has a 3' flanking region with T-rich clusters and CA repeats known to be susceptible to regulation by destabilization or ubiquination. NGF maintained catalase mRNA levels of actinomycin D (ACT-D) treated PC12 cells at twice that of cells exposed to ACT-D alone, delaying the rate of decay for catalase mRNA for 24 h. The NGF induction of GSH-Px and catalase mRNA was inhibited by cycloheximide (CHX) treatment with a slight decrease in their mRNA levels due to prolonged exposure to CHX. When the CHX treatment was delayed relative to the NGF treatment there was no effect on NGF effects on catalase and GSH-Px. The GSH-Px gene has conserved sequences in the open reading frame and 3' untranslated region which forms a stem-loop structure necessary for the incorporation of Se into this selenoprotein. While Se is important in stabilizing GSH-Px transcripts, it did not affect transcription rates or mRNA stability. These results are consistent with the hypothesis that NGF regulates catalase and GSH-Px expression via a primary effect on transcription factor pathways.


Assuntos
Antioxidantes , Catalase/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/genética , Fatores de Crescimento Neural/farmacologia , Animais , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Células PC12 , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA