Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Theor Appl Genet ; 132(9): 2579-2589, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31187154

RESUMO

KEY MESSAGE: qhkw5-3, a major QTL for kernel weight in maize, was mapped to an interval of 125.3 kb between the InDel markers InYM20 and InYM36, and the candidate genes were analysed. Yield, of which kernel weight is a major component, is the primary trait of interest in maize breeding programmes. In our previous study, a major QTL (named qhkw5-3), which controls hundred-kernel weight, was identified and mapped to the interval between simple sequence repeat (SSR) markers SYM033 and SYM108 on chromosome 5, using an F2:3 population derived from a cross between the maize inbred line Zheng58 and the single-segment substitution line Z22. In order to fine map qhkw5-3, a larger BC1F1 segregating population of 14,759 seeds, derived from a (Z22 × Zheng58) × Z22 cross, was screened using the SSR markers SYM036 and SYM119. Forty genotypes with donor chromosomal fragments of different lengths were obtained. Progeny testing results indicated that qhkw5-3 was mapped to an interval of 442.6 kb between the SSR markers SYM077 and SYM084. Overlap mapping results, based on seven homozygous recombinant lines, showed that qhkw5-3 was narrowed down to an interval of 125.3 kb between the InDel markers InYM20 and InYM36. Within this interval, six candidate genes were analysed using qRT-PCR. The results of this study lay the foundations for cloning and functional analysis of qhkw5-3 and will contribute to advancing our knowledge of the genetic basis of yield traits in maize.


Assuntos
Mapeamento Cromossômico/métodos , Cromossomos de Plantas/genética , Proteínas de Plantas/genética , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Sementes/genética , Zea mays/genética , Estudos de Associação Genética , Ligação Genética , Genótipo , Fenótipo
2.
Nature ; 485(7396): 55-61, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22367541

RESUMO

The mammalian target of rapamycin (mTOR) kinase is a master regulator of protein synthesis that couples nutrient sensing to cell growth and cancer. However, the downstream translationally regulated nodes of gene expression that may direct cancer development are poorly characterized. Using ribosome profiling, we uncover specialized translation of the prostate cancer genome by oncogenic mTOR signalling, revealing a remarkably specific repertoire of genes involved in cell proliferation, metabolism and invasion. We extend these findings by functionally characterizing a class of translationally controlled pro-invasion messenger RNAs that we show direct prostate cancer invasion and metastasis downstream of oncogenic mTOR signalling. Furthermore, we develop a clinically relevant ATP site inhibitor of mTOR, INK128, which reprograms this gene expression signature with therapeutic benefit for prostate cancer metastasis, for which there is presently no cure. Together, these findings extend our understanding of how the 'cancerous' translation machinery steers specific cancer cell behaviours, including metastasis, and may be therapeutically targeted.


Assuntos
Metástase Neoplásica , Neoplasias da Próstata/patologia , Biossíntese de Proteínas , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Benzoxazóis/farmacologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Genoma/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/genética , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/genética , Fosfoproteínas/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Pirimidinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores
3.
Cancer Cell ; 9(5): 367-78, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16697957

RESUMO

We demonstrate that PTEN loss causes reduced NKX3.1 expression in both murine and human prostate cancers. Restoration of Nkx3.1 expression in vivo in Pten null epithelium leads to decreased cell proliferation, increased cell death, and prevention of tumor initiation. Whereas androgen receptor (AR) positively regulates NKX3.1 expression, NKX3.1 negatively modulates AR transcription and consequently the AR-associated signaling events. Consistent with its tumor suppressor functions, NKX3.1 engages cell cycle and cell death machinery via association with HDAC1, leading to increased p53 acetylation and half-life through MDM2-dependent mechanisms. Importantly, overexpression of Nkx3.1 has little effect on Pten wild-type epithelium, suggesting that PTEN plays a predominant role in PTEN-NKX3.1 interplay. Manipulating NKX3.1 expression may serve as a therapeutic strategy for treating PTEN-deficient prostate cancers.


Assuntos
Proteínas de Homeodomínio/metabolismo , PTEN Fosfo-Hidrolase/deficiência , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Animais , Proliferação de Células , Regulação para Baixo/genética , Ativação Enzimática , Epitélio/metabolismo , Epitélio/patologia , Expressão Gênica , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Humanos , Masculino , Camundongos , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transplantes
4.
Prostate ; 73(13): 1378-90, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23728788

RESUMO

BACKGROUND: Prostate cancer stem cells (PCSC) offer theoretical explanations to many clinical and biological behaviors of the disease in human. In contrast to approaches of using side populations and cell-surface markers to isolate and characterize the putative PCSC, we hypothesize that androgen deprivation leads to functional enrichment of putative PCSC. METHODS AND RESULTS: Human prostate cancer lines LNCaP, LAPC4 and LAPC9 were depleted of androgen in cell cultures and in castrated SCID mice. The resultant androgen deprivation-resistant or castration-resistant populations, in particular in LNCaP and its derivative cell lines, displayed increased expression of pluripotency transactivators and significantly higher tumorigenicity. Individual tumor cell clones were isolated from castration-resistant bulk cultures of LNCaP (CR-LNCaP) and tested for tumorigenicity in male SCID mice under limiting dilution conditions. As few as 200 cells were able to form spheres in vitro, and generate tumors with similar growth kinetics as 10(6) LNCaP or 10(4) CR-LNCaP cells in vivo. These putative PCSC were CD44(+) /CD24(-) and lack the expression of prostate lineage proteins. When transplanted into the prostate of an intact male SCID mouse, these putative PCSC seemed to show limited differentiation into Ck5(+) , Ck8(+) , Ck5(+) /Ck8(+) , and AR(+) cells. On the other hand, stable transduction of LNCaP with retrovirus encoding Sox2 led to androgen-deprivation resistant growth and down-regulation of major prostate lineage gene products in vitro. CONCLUSION: Concurrence of overexpression of pluripotency transactivators and resistance to androgen deprivation supported the role of putative PCSC in the emergence of prostate cancer resistant to androgen deprivation.


Assuntos
Androgênios/metabolismo , Células-Tronco Neoplásicas/patologia , Próstata/patologia , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Regulação para Cima , Animais , Castração , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/genética
5.
Cancer Cell ; 4(3): 209-21, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14522255

RESUMO

The murine Pten prostate cancer model described in this study recapitulates the disease progression seen in humans: initiation of prostate cancer with prostatic intraepithelial neoplasia (PIN), followed by progression to invasive adenocarcinoma, and subsequent metastasis with defined kinetics. Furthermore, while Pten null prostate cancers regress after androgen ablation, they are capable of proliferating in the absence of androgen. Global assessment of molecular changes caused by homozygous Pten deletion identified key genes known to be relevant to human prostate cancer, including those "signature" genes associated with human cancer metastasis. This murine prostate cancer model provides a unique tool for both exploring the molecular mechanism underlying prostate cancer and for development of new targeted therapies.


Assuntos
Deleção de Genes , Monoéster Fosfórico Hidrolases/genética , Hiperplasia Prostática/genética , Neoplasias da Próstata/genética , Proteínas Serina-Treonina Quinases , Proteínas Supressoras de Tumor/genética , Animais , Divisão Celular , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Knockout , Metástase Neoplásica , PTEN Fosfo-Hidrolase , Hiperplasia Prostática/patologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt
6.
J Cancer Res Clin Oncol ; 148(9): 2561-2566, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35648195

RESUMO

Malignant pleural mesothelioma (MPM) is a rare and deadly malignancy with an extremely poor prognosis. The median overall survival (OS) of this disease is 12-18 months. However, the oncogenic driver mutations of MPM are rarely understood, and the targeted therapy for it is still under investigation. In this report, we describe a case of MPM with CD74-ROS1 fusion who obtains complete and durable response after receiving crizotinib. By the time of submission, the progression-free survival (PFS) with crizotinib has been 6.0 years, and the patient has survived for 7.6 years. Currently, he is still in complete remission (CR). To the best of our knowledge, this case represents the first report of CD74-ROS1 fusion identified in MPM. Meanwhile, it is also the first report of complete and long-term response to crizotinib in a patient with MPM positive for CD74-ROS1 fusion. This case report might contribute to the tumorigenesis and targeted therapy of this deadly disease.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Mesotelioma Maligno , Mesotelioma , Carcinoma Pulmonar de Células não Pequenas/patologia , Crizotinibe/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Mesotelioma/tratamento farmacológico , Mesotelioma/genética , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética
7.
Differentiation ; 77(1): 103-11, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19281769

RESUMO

Tumor suppressor gene PTEN is important in the initiation and progression of human prostate carcinoma, whereas the role of TP53 remains controversial. Since Pten/Trp53 double conditional knockout mice show earlier onset and fast progression of prostate cancer when compared to Pten knockout mice, we asked whether heterozygosity of these two tumor suppressor genes was sufficient to accelerate prostatic tumorigenesis. To answer this question we examined prostatic lesion progression of Pten/Trp53 double heterozygous mice and a series of controls such as Pten heterozygous, Pten conditional knockout, Trp53 heterozygous and Trp53 knockout mice. Tissue recombination of adult prostatic epithelium coupled with embryonic rat seminal vesicle mesenchyme was used as a tool to stimulate prostatic epithelial proliferation. In our study, high-grade prostatic intraepithelial neoplasia (PIN) was found with high frequency at 8 weeks post-tissue recombination transplantation. PIN lesions in Pten/Trp53 double heterozygous mice were more severe than those seen in Pten heterozygous alone. Furthermore, morphologic features attributable to Pten or Trp53 loss appeared to be enhanced in double heterozygous tissues. LOH analysis of Pten and Trp53 in genomic DNA collected from high-grade PIN lesions in Pten heterozygous and Pten/Trp53 double heterozygous mice showed an intact wild-type allele for both genes in all samples examined. In conclusion, simultaneous heterozygosity of Pten and Trp53 accelerates prostatic tumorigenesis in this mouse model of prostate cancer independently of loss of heterozygosity of either gene.


Assuntos
PTEN Fosfo-Hidrolase/metabolismo , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Modelos Animais de Doenças , Heterozigoto , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , Ratos , Proteína Supressora de Tumor p53/genética
8.
Methods Mol Biol ; 568: 151-60, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19582425

RESUMO

Prostate cancer is a major health concern in the Western world. Prostate cancer stem cells have been implicated to be involved in, if not solely responsible for prostate cancer initiation and relapse after surgical, hormonal, and chemotherapy. Until now, the identity of the presumed prostate cancer stem cell has been illusive and the efforts to characterize such cells have been hampered by the lack of efficient prostate stem cell activity assay. Anchorage-independent growth of brain and mammary stem cells in vitro has been proved to be a useful tool to enrich and characterize neural and mammary stem cells. Recently, we have successfully established a prostosphere cultivation method employing strategies similar to neurosphere and mammosphere cultures. Our results demonstrate that prostosphere-forming cells possess stem cell characters and are significantly over-represented in cancer tissues.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Neoplásicas/patologia , Neoplasias da Próstata/patologia , Animais , Adesão Celular , Proliferação de Células , Tamanho Celular , Dissecação , Humanos , Masculino , Camundongos , Células Tumorais Cultivadas
9.
Cancer Res ; 67(13): 6083-91, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17616663

RESUMO

PTEN mutations are among the most frequent genetic alterations found in human prostate cancers. Our previous works suggest that although precancerous lesions were found in Pten heterozygous mice, cancer progression and metastasis only happened when both alleles of Pten were deleted. To understand the molecular mechanisms underlying the role of PTEN in prostate cancer control, we generated two pairs of isogenic, androgen receptor (AR)-positive prostate epithelial lines from intact conditional Pten knock-out mice that are either heterozygous (PTEN-P2 and -P8) or homozygous (PTEN-CaP2 and PTEN-CaP8) for Pten deletion. Further characterization of these cells showed that loss of the second allele of Pten leads to increased anchorage-independent growth in vitro and tumorigenesis in vivo without obvious structural or numerical chromosome changes based on SKY karyotyping analysis. Despite no prior exposure to hormone ablation therapy, Pten null cells are tumorigenic in both male and female severe combined immunodeficiency mice. Furthermore, knocking down PTEN can convert the androgen-dependent Myc-CaP cell into androgen independence, suggesting that PTEN intrinsically controls androgen responsiveness, a critical step in the development of hormone refractory prostate cancer. Importantly, knocking down AR by shRNA in Pten null cells reverses androgen-independent growth in vitro and partially inhibited tumorigenesis in vivo, indicating that PTEN-controlled prostate tumorigenesis is AR dependent. These cell lines will serve as useful tools for understanding signaling pathways controlled by PTEN and elucidating the molecular mechanisms involved in hormone refractory prostate cancer formation.


Assuntos
Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/fisiologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ágar/química , Alelos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Cromossomos/ultraestrutura , Feminino , Cariotipagem , Masculino , Camundongos , Camundongos Knockout , Mutação , PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/etiologia
11.
Cancer Res ; 66(2): 883-8, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424021

RESUMO

Osteopontin is a secreted glycosylated phosphoprotein known to be involved in numerous physiologic functions and associated with the late stages of various cancers. We used preneoplastic and neoplastic mouse models of prostate cancer to determine the onset of elevated expression of osteopontin in the development of this disease. Osteopontin alterations occurred early in the disease with dysregulated expression observed in lesions of low-grade prostatic intraepithelial neoplasia (PIN). Over time, osteopontin expressing dysplastic cells seemed to increase in number in high-grade PIN and increased further in adenocarcinoma, and in metastasis, almost all of the cancer cells immunohistochemically stained positive for osteopontin overexpression. We examined the biological properties of human prostate cancer cell lines LNCaP and PC-3, in which osteopontin overexpression was achieved via lentiviral gene transduction. Evidence was obtained that osteopontin could contribute to a proliferative advantage in both cell types, although more significantly in LNCaP than PC-3. Osteopontin also influenced their in vitro invasive ability, and again, most strikingly in the weakly oncogenic LNCaP. Furthermore, excess osteopontin induced the LNCaP cells to acquire a strong intravasation potential in vivo in the chicken embryo chorioallantoic membrane assay for blood vessel penetration. These results establish a correlation between an increased gradient of osteopontin expression throughout the stages of murine prostate cancer, beginning from the preneoplastic lesions to distant metastases that suggests a proliferative and invasive advantages to those prostate tumor cells overexpressing osteopontin. Together, these findings support a strategy designed to target osteopontin in the context of prostate cancer therapy.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Perfilação da Expressão Gênica , Neoplasias da Próstata/genética , Sialoglicoproteínas/biossíntese , Animais , Embrião de Galinha , Progressão da Doença , Humanos , Masculino , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Osteopontina , Neoplasias da Próstata/patologia , Sialoglicoproteínas/fisiologia , Transdução Genética , Células Tumorais Cultivadas
12.
PLoS One ; 7(8): e42564, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22880034

RESUMO

New therapies for late stage and castration resistant prostate cancer (CRPC) depend on defining unique properties and pathways of cell sub-populations capable of sustaining the net growth of the cancer. One of the best enrichment schemes for isolating the putative stem/progenitor cell from the murine prostate gland is Lin(-);Sca1(+);CD49f(hi) (LSC(hi)), which results in a more than 10-fold enrichment for in vitro sphere-forming activity. We have shown previously that the LSC(hi) subpopulation is both necessary and sufficient for cancer initiation in the Pten-null prostate cancer model. To further improve this enrichment scheme, we searched for cell surface molecules upregulated upon castration of murine prostate and identified CD166 as a candidate gene. CD166 encodes a cell surface molecule that can further enrich sphere-forming activity of WT LSC(hi) and Pten null LSC(hi). Importantly, CD166 could enrich sphere-forming ability of benign primary human prostate cells in vitro and induce the formation of tubule-like structures in vivo. CD166 expression is upregulated in human prostate cancers, especially CRPC samples. Although genetic deletion of murine CD166 in the Pten null prostate cancer model does not interfere with sphere formation or block prostate cancer progression and CRPC development, the presence of CD166 on prostate stem/progenitors and castration resistant sub-populations suggest that it is a cell surface molecule with the potential for targeted delivery of human prostate cancer therapeutics.


Assuntos
Molécula de Adesão de Leucócito Ativado/metabolismo , Biomarcadores Tumorais/metabolismo , Membrana Celular/metabolismo , Células-Tronco Neoplásicas/patologia , Próstata/patologia , Neoplasias da Próstata/patologia , Animais , Castração , Modelos Animais de Doenças , Progressão da Doença , Epitélio/metabolismo , Epitélio/patologia , Deleção de Genes , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Células-Tronco Neoplásicas/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Regeneração , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Análise Serial de Tecidos , Regulação para Cima
13.
Cancer Cell ; 19(6): 792-804, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21620777

RESUMO

Alteration of the PTEN/PI3K pathway is associated with late-stage and castrate-resistant prostate cancer (CRPC). However, how PTEN loss is involved in CRPC development is not clear. Here, we show that castration-resistant growth is an intrinsic property of Pten null prostate cancer (CaP) cells, independent of cancer development stage. PTEN loss suppresses androgen-responsive gene expressions by modulating androgen receptor (AR) transcription factor activity. Conditional deletion of Ar in the epithelium promotes the proliferation of Pten null cancer cells, at least in part, by downregulating the androgen-responsive gene Fkbp5 and preventing PHLPP-mediated AKT inhibition. Our findings identify PI3K and AR pathway crosstalk as a mechanism of CRPC development, with potentially important implications for CaP etiology and therapy.


Assuntos
Orquiectomia , PTEN Fosfo-Hidrolase/fisiologia , Neoplasias da Próstata/patologia , Animais , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Histona-Lisina N-Metiltransferase/genética , Humanos , Masculino , Camundongos , Proteínas Nucleares/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Complexo Repressor Polycomb 2 , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/prevenção & controle , Neoplasias da Próstata/terapia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Receptores Androgênicos/fisiologia , Proteínas de Ligação a Tacrolimo/fisiologia
14.
Cancer Cell ; 14(2): 146-55, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18691549

RESUMO

Transgenic expression of activated AKT1 in the murine prostate induces prostatic intraepithelial neoplasia (PIN) that does not progress to invasive prostate cancer (CaP). In luminal epithelial cells of Akt-driven PIN, we show the concomitant induction of p27(Kip1) and senescence. Genetic ablation of p27(Kip1) led to downregulation of senescence markers and progression to cancer. In humans, p27(Kip1) and senescence markers were elevated in PIN not associated with CaP but were decreased or absent, respectively, in cancer-associated PIN and in CaP. Importantly, p27(Kip1) upregulation in mouse and human in situ lesions did not depend upon mTOR or Akt activation but was instead specifically associated with alterations in cell polarity, architecture, and adhesion molecules. These data suggest that a p27(Kip1)-driven checkpoint limits progression of PIN to CaP.


Assuntos
Senescência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Alelos , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Adesão Celular , Comunicação Celular , Linhagem Celular , Polaridade Celular , Proliferação de Células , Progressão da Doença , Células Epiteliais/patologia , Humanos , Masculino , Camundongos , Mutação/genética , Fenótipo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Serina-Treonina Quinases TOR
15.
Proc Natl Acad Sci U S A ; 103(5): 1480-5, 2006 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-16432235

RESUMO

PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a potent tumor suppressor gene frequently mutated in human prostate cancers. Deletion of Pten in a murine model of prostate cancer recapitulates the disease progression seen in humans. Using defined cell lineage markers, we demonstrate that PTEN negatively regulates p63-positive prostatic basal cell proliferation without blocking differentiation. Concomitant with basal cell proliferation is the expansion of a prostate stem/progenitor-like subpopulation as evidenced by the progressive increase of stem cell antigen-1 (Sca-1)- and BCL-2-positive cells. This observation provides strong evidence that basal cell proliferation can be an initiating event for precancerous lesions. Sca-1(+) and BCL-2(+) progenitors may serve as cancer-initiating cells in this model.


Assuntos
PTEN Fosfo-Hidrolase/fisiologia , Neoplasias da Próstata/genética , Células-Tronco/citologia , Animais , Bromodesoxiuridina/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Separação Celular , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Progressão da Doença , Citometria de Fluxo , Deleção de Genes , Genes Supressores de Tumor , Humanos , Imuno-Histoquímica , Antígeno Ki-67/biossíntese , Cinética , Masculino , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfoproteínas/metabolismo , Neoplasias da Próstata/patologia , Fatores de Tempo , Transativadores/metabolismo , Fatores de Transcrição , Proteínas Supressoras de Tumor
16.
Prostate ; 66(16): 1698-709, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16998820

RESUMO

BACKGROUND: Relaxin is a potent peptide hormone normally secreted by the prostate. This study characterized relaxin expression during prostate cancer progression to androgen independence (AI), and in response to androgens. METHODS: The prostate cancer cell line, LNCaP, was assayed by microarrays and confirmatory Northern analysis to assess changes in relaxin levels due to androgen treatment and in LNCaP xenografts following castration. Relaxin protein levels were examined by immunohistochemistry (IHC) in tissue microarrays of human prostate cancer samples following androgen ablation. RESULTS: Relaxin levels decreased in a time and concentration-dependent manner due to androgens in vitro, and increased in xenografts post-castration. Relaxin increased in radical prostatectomy specimens after 6 months of androgen ablation and in AI tumors, was highest in bone metastases. CONCLUSIONS: Relaxin is negatively regulated by androgens in vitro and in vivo, which correlates to clinical prostate cancer specimens following androgen ablation. The role of relaxin in angiogenesis and tissue remodeling suggests it may contribute to prostate cancer progression.


Assuntos
Metribolona/farmacologia , Neoplasias da Próstata/metabolismo , Relaxina/metabolismo , Congêneres da Testosterona/farmacologia , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Hormônio-Dependentes/metabolismo , Orquiectomia , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Relaxina/biossíntese , Relaxina/genética , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Dev Biol ; 273(2): 175-84, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15328005

RESUMO

Recent studies indicate that certain key molecules that are vital for various developmental processes, such as Wnt, Shh, and Notch, cause cancer when dysregulated. PTEN, a tumor suppressor that antagonizes the PI3 kinase pathway, is the newest one on the list. The biological function of PTEN is evolutionarily conserved from C. elegans to humans, and the PTEN-controlled signaling pathway regulates cellular processes crucial for normal development, including cell proliferation, soma growth, cell death, and cell migration. In this review, we will focus on the function of PTEN in murine development and its role in regulating stem cell self-renewal and proliferation. We will summarize the organomegaly phenotypes associated with Pten tissue-specific deletion and discuss how PTEN controls organ size, a fundamental aspect of development. Last, we will review the role of PTEN in hormone-dependent, adult-onset mammary and prostate gland development.


Assuntos
Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/fisiologia , Animais , Movimento Celular , Desenvolvimento Embrionário e Fetal/genética , Desenvolvimento Embrionário e Fetal/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hormônios/fisiologia , Humanos , Masculino , Camundongos , Modelos Biológicos , PTEN Fosfo-Hidrolase , Proteínas Tirosina Fosfatases/genética , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/fisiologia , Proteínas Supressoras de Tumor/genética
18.
Acta Pharmacol Sin ; 24(9): 864-72, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12956933

RESUMO

AIM: To investigate the global gene expression profile changes in Jurkat cells after triptolide treatment in order to find the possible triptolide targets. METHODS: Jurkat cells were treated with or without triptolide 10 microg/L for 2 h. Total RNA were isolated and used as templates for reverse transcriptional labeling of fluorescent cDNA probes. High density DNA microarray chips with a set of 13,872 human genes/Ests were used to generate the expression profile of triptolide-treated or untreated control Jurkat cells by hybridizing with fluorescent labeled probes. Array image was acquired and analyzed with array analyzing software GeneSpring. RESULTS: Triptolide significantly suppressed expression of 117 genes in Jurkat cells. Among these 117 genes, 30 % were Ests or genes without known functions, 13 % were transcription factors, 9 % were signal transduction pathway regulators, and 9 % were DNA binding proteins. Notably, the expression of mitogen-activated protein kinase kinase kinase kinase 5 (MAP kinase 5) and phosphoinositide-3-kinase (PI-3 kinase) was inhibited more than 100-fold. Moreover, the expression of genes involved in lipid transportation and metabolism was down-regulated by triptolide. CONCLUSION: High-density microarray provided an effective approach to identify drug targeting molecules. It is suggested that the widely known immune suppressive and antitumor effects of triptolide were mediated at least in part by suppression of MAP kinase and PI-3 kinase gene expression.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Diterpenos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , MAP Quinase Quinase Quinases/biossíntese , Fenantrenos/farmacologia , Fosfatidilinositol 3-Quinases/biossíntese , Diterpenos/isolamento & purificação , Compostos de Epóxi , Humanos , Imunossupressores/farmacologia , Células Jurkat , MAP Quinase Quinase Quinase 5 , MAP Quinase Quinase Quinases/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fenantrenos/isolamento & purificação , Fosfatidilinositol 3-Quinases/genética , Plantas Medicinais/química , RNA Mensageiro/genética , Tripterygium/química
19.
Genomics ; 82(3): 331-42, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12906858

RESUMO

Androgens regulate important processes involved in the normal development and function of the human and rodent prostate glands. Here we report the isolation and characterization of a new androgen-regulated gene, designated WDR19, that encodes repeating sequence motifs found in the WD-repeat family of proteins. The WD repeat is a conserved domain of approximately 40 amino acids that is typically bracketed by glycine-histidine and tryptophan-aspartic acid (WD) dipeptides. WD-repeat proteins are a large group of structurally related proteins that participate in a wide range of cellular functions, including transmembrane signaling, mRNA modification, vesicle formation, and vesicular trafficking. The WDR19 gene comprises 36 exons and is located on chromosome 4p15-4p11. The predicted protein contains six WD repeats, a clathrin heavy-chain repeat, and three transmembrane domains. Sequence analysis reveals that the WDR19 gene is conserved from Caenorhabditis elegans to human. WDR19 is expressed in normal and neoplastic prostate epithelium as demonstrated by RNA in situ hybridization and is regulated by androgenic hormones. WDR19 transcripts exhibit alternative splicing in which two isoforms appear to be prostate restricted, a property that could be exploited for designing diagnostic or therapeutic strategies for prostate carcinoma.


Assuntos
Androgênios/metabolismo , Próstata/metabolismo , Proteínas/genética , Proteínas/isolamento & purificação , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência Conservada , Proteínas do Citoesqueleto , Epitélio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa