Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Total Environ ; 737: 139707, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32516662

RESUMO

An optimal flowering period (OFP) minimises the long-term combined risk of extreme weather events on crop yield and exists in all environments. With climate change, the frequency, timing and intensity of these events are likely to change, which in turn may shift the OFP. It is important to explore how the OFP would change under a future climate. Knowledge of the OFP is important for formulating breeding strategies and developing suitable varieties. Here, a simulation analysis was conducted at 4 sites in Western Australia to quantify any shift in the OFP due to climate change, by accounting for the effects of frost, heat and water stress on wheat yield. Three global climate models that projected the greatest precipitation decrease under the Representative Concentration Pathways 8.5 during 2061-2100 were ensembled to represent a dry future climate condition (dry scenario); and 3 models that predicted the smallest decrease in precipitation were ensembled to represent a wet future climate condition (wet scenario). The simulation results predicted that the timing of OFPs for wheat in Western Australia would occur earlier than the current OFP. On average the OFP was 29 days earlier in the dry scenario and 11 days earlier in the wet scenario. Early sowing of long-season varieties would be preferable to achieve the OFP in both climate scenarios due to greater yield potential. Early sowing opportunities were very limited under the dry scenario, and therefore fast maturing varieties for late sowing would also be necessary.


Assuntos
Triticum , Água , Mudança Climática , Estações do Ano , Temperatura , Austrália Ocidental
2.
Glob Chang Biol ; 24(6): 2403-2415, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29284201

RESUMO

Climate change threatens global wheat production and food security, including the wheat industry in Australia. Many studies have examined the impacts of changes in local climate on wheat yield per hectare, but there has been no assessment of changes in land area available for production due to changing climate. It is also unclear how total wheat production would change under future climate when autonomous adaptation options are adopted. We applied species distribution models to investigate future changes in areas climatically suitable for growing wheat in Australia. A crop model was used to assess wheat yield per hectare in these areas. Our results show that there is an overall tendency for a decrease in the areas suitable for growing wheat and a decline in the yield of the northeast Australian wheat belt. This results in reduced national wheat production although future climate change may benefit South Australia and Victoria. These projected outcomes infer that similar wheat-growing regions of the globe might also experience decreases in wheat production. Some cropping adaptation measures increase wheat yield per hectare and provide significant mitigation of the negative effects of climate change on national wheat production by 2041-2060. However, any positive effects will be insufficient to prevent a likely decline in production under a high CO2 emission scenario by 2081-2100 due to increasing losses in suitable wheat-growing areas. Therefore, additional adaptation strategies along with investment in wheat production are needed to maintain Australian agricultural production and enhance global food security. This scenario analysis provides a foundation towards understanding changes in Australia's wheat cropping systems, which will assist in developing adaptation strategies to mitigate climate change impacts on global wheat production.


Assuntos
Mudança Climática , Triticum/fisiologia , Aclimatação , Agricultura/métodos , Austrália , Abastecimento de Alimentos
3.
Oncol Rep ; 38(5): 3227-3237, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29048636

RESUMO

Estrogen receptor (ER) plays important roles in cell growth, development and tumorigenesis. Although ER-regulated genes have been extensively investigated, little is known about roles of ER-regulated lncRNAs in breast cancer. Here, we conducted genome-wide study of ER-regulated lncRNAs by using RNA-seq, ChIP-seq and TCGA data. A total of identified 114 ER-regulated lncRNAs were identified, many of them were overexpressed in ER+ breast cancer and co-expressed with some key regulators. Silencing one of most prominent lncRNA, AP000439.3, resulted in inhibition of cell cycle progression and proliferation. Further study revealed AP000439.3 can regulate expression of CCND1 through enhancing estrogen receptor induction of CCND1. This finding revealed lncRNAs may serve as important effectors of ER in regulation of gene expression and cell phenotype in breast cancer.


Assuntos
Neoplasias da Mama/genética , Ciclina D1/genética , Receptor alfa de Estrogênio/genética , RNA Longo não Codificante/genética , Neoplasias da Mama/patologia , Carcinogênese/genética , Ciclo Celular/genética , Proliferação de Células/genética , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células MCF-7
4.
PLoS One ; 7(8): e43891, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22937117

RESUMO

Nanosecond pulsed electric fields (nsPEF) induce apoptotic pathways in human cancer cells. The potential therapeutic effective of nsPEF has been reported in cell lines and in xenograft animal tumor model. The present study investigated the ability of nsPEF to cause cancer cell death in vivo using carcinogen-induced animal tumor model, and the pulse duration of nsPEF was only 7 and 14 nano second (ns). An nsPEF generator as a prototype medical device was used in our studies, which is capable of delivering 7-30 nanosecond pulses at various programmable amplitudes and frequencies. Seven cutaneous squamous cell carcinoma cell lines and five other types of cancer cell lines were used to detect the effect of nsPEF in vitro. Rate of cell death in these 12 different cancer cell lines was dependent on nsPEF voltage and pulse number. To examine the effect of nsPEF in vivo, carcinogen-induced cutaneous papillomas and squamous cell carcinomas in mice were exposed to nsPEF with three pulse numbers (50, 200, and 400 pulses), two nominal electric fields (40 KV/cm and 31 KV/cm), and two pulse durations (7 ns and 14 ns). Carcinogen-induced cutaneous papillomas and squamous carcinomas were eliminated efficiently using one treatment of nsPEF with 14 ns duration pulses (33/39 = 85%), and all remaining lesions were eliminated after a 2nd treatment (6/39 = 15%). 13.5% of carcinogen-induced tumors (5 of 37) were eliminated using 7 ns duration pulses after one treatment of nsPEF. Associated with tumor lysis, expression of the anti-apoptotic proteins Bcl-xl and Bcl-2 were markedly reduced and apoptosis increased (TUNEL assay) after nsPEF treatment. nsPEF efficiently causes cell death in vitro and removes papillomas and squamous cell carcinoma in vivo from skin of mice. nsPEF has the therapeutic potential to remove human squamous carcinoma.


Assuntos
Carcinoma de Células Escamosas/terapia , Eletroquimioterapia , Eletroporação , Papiloma/terapia , Neoplasias Cutâneas/terapia , Animais , Linhagem Celular Tumoral , Camundongos
5.
Blood Cells Mol Dis ; 45(4): 269-75, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20926322

RESUMO

The search for novel therapeutic candidates targeting fetal hemoglobin (HbF) activation to reduce the imbalance of globin genes is regarded as a promising approach for the clinical management of sickle cell disease and ß-thalassemia. For the first time, we identified cucurbitacin D (CuD), an oxygenated tetracyclic triterpenoid, as a molecular entity inducing γ-globin gene expression and HbF synthesis in K562 cells and human hematopoietic progenitors from a ß-thalassemia patient. CuD demonstrated a higher potency in HbF induction when compared with hydroxyurea, which was revealed by the evidence that CuD results in a higher fetal cell percentage and greater HbF content in K562 cells, in addition, to being less cytotoxic. Moreover, CuD also promotes higher HbF expression in primary erythroid cells. In the study to elucidate the molecular mechanisms of CuD's action, our data indicated that CuD-stimulated HbF synthesis was mediated by p38 pathway activation. At the post-transcriptional level, CuD treatment led to a significant elongation of the γ-globin mRNA half-life in K562 cells. Taken together, the results suggest that CuD may be a potential therapeutic agent for ß-hemoglobinopathies, including sickle cell anemia and ß-thalassemia.


Assuntos
Hemoglobina Fetal/biossíntese , Células-Tronco Hematopoéticas/metabolismo , Células K562/metabolismo , Triterpenos/farmacologia , gama-Globinas/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Hemoglobinopatias/tratamento farmacológico , Humanos , Estabilidade de RNA , RNA Mensageiro/metabolismo
6.
J Leukoc Biol ; 88(4): 699-706, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20571037

RESUMO

The M-CSFR (c-Fms) participates in proliferation, differentiation, and survival of macrophages and is involved in the regulation of distinct macrophage functions. Interaction with the ligand M-CSF results in phosphorylation of tyrosine residues on c-Fms, thereby creating binding sites for molecules containing SH2 domains. Lnk is a SH2 domain adaptor protein that negatively regulates hematopoietic cytokine receptors. Here, we show that Lnk binds to c-Fms. Biological and functional effects of this interaction were examined in macrophages from Lnk-deficient (KO) and WT mice. Clonogenic assays demonstrated an elevated number of M-CFUs in the bone marrow of Lnk KO mice. Furthermore, the M-CSF-induced phosphorylation of Akt in Lnk KO macrophages was increased and prolonged, whereas phosphorylation of Erk was diminished. Zymosan-stimulated production of ROS was increased dramatically in a M-CSF-dependent manner in Lnk KO macrophages. Lastly, Lnk inhibited M-CSF-induced migration of macrophages. In summary, we show that Lnk binds to c-Fms and can blunt M-CSF stimulation. Modulation of levels of Lnk in macrophages may provide a unique therapeutic approach to increase innate host defenses.


Assuntos
Macrófagos/metabolismo , Proteínas/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Western Blotting , Diferenciação Celular/imunologia , Quimiotaxia de Leucócito/imunologia , Ensaio de Imunoadsorção Enzimática , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/citologia , Macrófagos/imunologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Fagocitose/imunologia , Proteínas/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Leukoc Biol ; 85(6): 957-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19293402

RESUMO

The JAK2 mutation JAK2V617F is found frequently in patients with myeloproliferative disorders (MPD) and transforms hematopoietic cells to cytokine-independent proliferation when expressed with specific cytokine receptors. The Src homology 2 (SH2) and pleckstrin homology (PH) domain-containing adaptor protein Lnk (SH2B3) is a negative regulator of hematopoietic cytokine signaling. Here, we show that Lnk is a potent inhibitor of JAK2V617F constitutive activity. Lnk down-regulates JAK2V617F-mediated signaling and transformation in hematopoietic Ba/F3-erythropoietin receptor cells. Furthermore, in CFU assays, Lnk-deficient murine bone marrow cells are significantly more sensitive to transformation by JAK2V617F than wild-type (WT) cells. Lnk, through its SH2 and PH domains, interacts with WT and mutant JAK2 and is phosphorylated by constitutively activated JAK2V617F. Finally, we found that Lnk levels are high in CD34(+) hematopoietic progenitors from MPD patients and that Lnk expression is induced following JAK2 activation. Our data suggest that JAK2V617F is susceptible to endogenous negative-feedback regulation, providing new insights into the molecular pathogenesis of MPD.


Assuntos
Substituição de Aminoácidos/genética , Janus Quinase 2/antagonistas & inibidores , Proteínas Mutantes/antagonistas & inibidores , Transtornos Mieloproliferativos/enzimologia , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular Transformada , Proliferação de Células , Ativação Enzimática , Sistema Hematopoético/citologia , Sistema Hematopoético/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Janus Quinase 2/metabolismo , Proteínas de Membrana , Camundongos , Células Mieloides/citologia , Células Mieloides/enzimologia , Fenilalanina/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Valina/genética
8.
Cancer Sci ; 99(9): 1793-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18627377

RESUMO

Cucurbitacins are a group of diverse triterpenoid substances isolated from plants with medicinal properties. One particularly potent family member is cucurbitacin B (CuB). The antiproliferative effects of CuB against human breast cancer cells were tested. Six human breast cancer cell lines were examined because they represent a diverse mix of breast cancer subtypes varying in expression of estrogen receptor (ER), Her2/neu, and p53 mutation. The antiproliferative effect of CuB were also studied in vivo. The effective dose inhibiting 50% growth (ED(50)) was between 10(-8) M and 10(-7) M for this collection of breast cancer cell lines. These cells underwent rapid morphologic changes after 15-20 min exposure to CuB (5 x 10(-7) M), which was associated with disruption of the microtubules and F-actin, as observed by confocal microscopy. Human MDA-MB-231 (ER-, p53 mutated) breast cancer cells were orthotopically implanted into the breasts of nude mice who intraperitoneally received either CuB 1.0 mg/kg or vehicle. Tumor volume was reduced by 55% in the group that received CuB for 6 weeks compared with vehicle controls. No apparent organ tissue damage was observed by pathological assessment. Interestingly, the experimental mice had lower serum glucose levels, consistent with use of CuB as an antidiabetic drug in China. This drug appears to be a third in a family of drugs targeting the microtubules (taxanes [e.g. taxol], vinca alkaloid [e.g. vincristine], and now CuB). Our in vitro and in vivo results suggest that CuB may be an effective, new approach for the treatment of ER-, Her2/neu amplified, and p53 mutant breast cancers.


Assuntos
Antineoplásicos , Proliferação de Células/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Triterpenos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama , Linhagem Celular Tumoral , Feminino , Humanos , Técnicas In Vitro , Camundongos , Triterpenos/química , Triterpenos/farmacologia
9.
Int J Cancer ; 123(6): 1364-75, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18561312

RESUMO

Glioblastoma Multiforme (GBM) is almost inevitably a fatal tumor of the brain with most individuals dying within 1 year of diagnosis. It is the most frequent brain tumor in adults. Dose-response studies showed that Cucurbitacin B inhibited 50% growth (ED(50)) of 5 human GBM cell lines in liquid culture at approximately 10(-7) M. Soft-gel assays demonstrated that nearly all of the GBM clonogenic cells were inhibited at 10(-8) M of Cucurbitacin B. FACS analysis found that the compound (10(-7) M, 24 hr) caused G2/M arrest. The GBM cells underwent profound morphologic changes within 15-30 min after exposure to Cucurbitacin B (10(-7) M), rounding up and losing their pseudopodia associated with disruption of actin and microtubules, as observed by immunoflourescence. Cucurbitacin B (10(-7) M) caused prominent multinucleation of the cells after they were pulse-exposed (48 hr) to the drug, washed and cultured in normal medium for an additional 2 days. The drug (10(-7) M, 3-24 hr) increased levels of p-p38, p-JNK and p-JUN in U87 and T98G GBM cell lines as seen by Western blot. Interestingly, alterations in cell morphology caused by Cucurbitacin B (10(-7) M) were blocked by the JNK inhibitor SP600125. In summary, Cucurbitacin B has a prominent anti-proliferative activity on GBM cells; and at least in part, the mode of action is by affecting the cytoskeleton, as well as, the JNK pathway. Clinical trails of this drug should be pursued in GBM.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Fitoterapia , Trichosanthes/química , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Microscopia Confocal , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-jun/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos
10.
J Chemother ; 20(2): 246-52, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18467253

RESUMO

Prostate carcinoma and metastasis are common among male subjects worldwide. CKBM is a drug product targeting prostate cancer in multiple ways. Prostate cancer cell lines PC3 and DU145 were treated with CKBM. The effect of CKBM on the cell's viability, cell cycle, adhesive and invasive properties and its growth in an animal model were assessed. Results indicated that CKBM inhibited PC3 and DU145 cell growth in vitro at IC(50 )values 3.923 and 4.697% respectively, and it brought about cell cycle arrest at G2/M phase. CKBM also attenuated DU145 cells to invade and adhere to extracellular matrices including Matrigel, laminin, fibronectin and collagen IV. Moreover, PC3 tumor xenograft growth was inhibited by over 60% after 28-day of 0.2, 0.4 or 0.8 ml/day CKBM treatment. The present study indicates that CKBM is effective against prostate cancer cell growth in vitro and in vivo. Further studies are required to elucidate its mechanism of action.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Extratos Vegetais/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Adesão Celular , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/uso terapêutico , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Transplante de Neoplasias , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/patologia , Extratos Vegetais/uso terapêutico , Neoplasias da Próstata/patologia , Transplante Heterólogo
11.
Leuk Res ; 32(9): 1366-73, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18405967

RESUMO

Cucurbitacins have long been utilized for their abortifacient and anti-inflammatory effects; however, little is known about their mechanism of action. In this study, we have demonstrated robust antiproliferative effects of CuB on various leukemia and lymphoma cell lines, as well as on primary mononuclear bone marrow cells derived from patients with acute myeloid leukemia or myelodysplastic syndrome. Myeloid leukemic cells treated with CuB exhibit significant S-phase cell cycle arrest, enlarged cell size, multinucleation, and enhanced expression of a monocytic- and granulocytic-specific CD11b. Moreover, our data demonstrate that CuB prominently alters the cytoskeletal network of leukemic cells, inducing rapid and improper polymerization of the F-actin network. These encouraging results suggest the appropriateness of clinical trials of cucurbitacins for the treatment of hematopoietic malignancies.


Assuntos
Actinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Citoesqueleto/patologia , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicas/patologia , Fase S/efeitos dos fármacos , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Antígeno CD11b/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Granulócitos/metabolismo , Granulócitos/patologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA