Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Clin Cancer Res ; 43(1): 15, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38195652

RESUMO

BACKGROUND: New drugs to tackle the next pathway or mutation fueling cancer are constantly proposed, but 97% of them are doomed to fail in clinical trials, largely because they are identified by cellular or in silico screens that cannot predict their in vivo effect. METHODS: We screened an Adeno-Associated Vector secretome library (> 1000 clones) directly in vivo in a mouse model of cancer and validated the therapeutic effect of the first hit, EMID2, in both orthotopic and genetic models of lung and pancreatic cancer. RESULTS: EMID2 overexpression inhibited both tumor growth and metastatic dissemination, consistent with prolonged survival of patients with high levels of EMID2 expression in the most aggressive human cancers. Mechanistically, EMID2 inhibited TGFß maturation and activation of cancer-associated fibroblasts, resulting in more elastic ECM and reduced levels of YAP in the nuclei of cancer cells. CONCLUSION: This is the first in vivo screening, precisely designed to identify proteins able to interfere with cancer cell invasiveness. EMID2 was selected as the most potent protein, in line with the emerging relevance of the tumor extracellular matrix in controlling cancer cell invasiveness and dissemination, which kills most of cancer patients.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Pancreáticas , Animais , Humanos , Camundongos , Núcleo Celular , Modelos Animais de Doenças , Detecção Precoce de Câncer , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Colágeno/metabolismo
3.
Nat Commun ; 14(1): 6777, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880212

RESUMO

Reprogramming of amino acid metabolism, sustained by oncogenic signaling, is crucial for cancer cell survival under nutrient limitation. Here we discovered that missense mutant p53 oncoproteins stimulate de novo serine/glycine synthesis and essential amino acids intake, promoting breast cancer growth. Mechanistically, mutant p53, unlike the wild-type counterpart, induces the expression of serine-synthesis-pathway enzymes and L-type amino acid transporter 1 (LAT1)/CD98 heavy chain heterodimer. This effect is exacerbated by amino acid shortage, representing a mutant p53-dependent metabolic adaptive response. When cells suffer amino acids scarcity, mutant p53 protein is stabilized and induces metabolic alterations and an amino acid transcriptional program that sustain cancer cell proliferation. In patient-derived tumor organoids, pharmacological targeting of either serine-synthesis-pathway and LAT1-mediated transport synergizes with amino acid shortage in blunting mutant p53-dependent growth. These findings reveal vulnerabilities potentially exploitable for tackling breast tumors bearing missense TP53 mutations.


Assuntos
Neoplasias da Mama , Proteína Supressora de Tumor p53 , Feminino , Humanos , Aminoácidos/metabolismo , Aminoácidos Essenciais , Neoplasias da Mama/patologia , Glicina , Transportador 1 de Aminoácidos Neutros Grandes/genética , Serina , Proteína Supressora de Tumor p53/genética
4.
Cell Rep ; 36(11): 109694, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525372

RESUMO

Chromatin organization plays a crucial role in tissue homeostasis. Heterochromatin relaxation and consequent unscheduled mobilization of transposable elements (TEs) are emerging as key contributors of aging and aging-related pathologies, including Alzheimer's disease (AD) and cancer. However, the mechanisms governing heterochromatin maintenance or its relaxation in pathological conditions remain poorly understood. Here we show that PIN1, the only phosphorylation-specific cis/trans prolyl isomerase, whose loss is associated with premature aging and AD, is essential to preserve heterochromatin. We demonstrate that this PIN1 function is conserved from Drosophila to humans and prevents TE mobilization-dependent neurodegeneration and cognitive defects. Mechanistically, PIN1 maintains nuclear type-B Lamin structure and anchoring function for heterochromatin protein 1α (HP1α). This mechanism prevents nuclear envelope alterations and heterochromatin relaxation under mechanical stress, which is a key contributor to aging-related pathologies.


Assuntos
Proteínas de Drosophila/metabolismo , Heterocromatina/metabolismo , Lamina Tipo B/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Peptidilprolil Isomerase/metabolismo , Estresse Mecânico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Homólogo 5 da Proteína Cromobox/genética , Homólogo 5 da Proteína Cromobox/metabolismo , Elementos de DNA Transponíveis/genética , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Humanos , Lamina Tipo B/química , Camundongos , Camundongos Endogâmicos C57BL , Peptidilprolil Isomerase de Interação com NIMA/antagonistas & inibidores , Peptidilprolil Isomerase de Interação com NIMA/genética , Neocórtex/citologia , Neocórtex/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Membrana Nuclear/química , Peptidilprolil Isomerase/antagonistas & inibidores , Peptidilprolil Isomerase/genética , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo
5.
Front Oncol ; 9: 94, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30873382

RESUMO

Cellular choices are determined by developmental and environmental stimuli through integrated signal transduction pathways. These critically depend on attainment of proper activation levels that in turn rely on post-translational modifications (PTMs) of single pathway members. Among these PTMs, post-phosphorylation prolyl-isomerization mediated by PIN1 represents a unique mechanism of spatial, temporal and quantitative control of signal transduction. Indeed PIN1 was shown to be crucial for determining activation levels of several pathways and biological outcomes downstream to a plethora of stimuli. Of note, studies performed in different model organisms and humans have shown that hormonal, nutrient, and oncogenic stimuli simultaneously affect both PIN1 activity and the pathways that depend on PIN1-mediated prolyl-isomerization, suggesting the existence of evolutionarily conserved molecular circuitries centered on this isomerase. This review focuses on molecular mechanisms and cellular processes like proliferation, metabolism, and stem cell fate, that are regulated by PIN1 in physiological conditions, discussing how these are subverted in and hijacked by cancer cells. Current status and open questions regarding the use of PIN1 as biomarker and target for cancer therapy as well as clinical development of PIN1 inhibitors are also addressed.

6.
Nature ; 550(7675): 249-254, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29022589

RESUMO

Adenosine-to-inosine (A-to-I) RNA editing is a conserved post-transcriptional mechanism mediated by ADAR enzymes that diversifies the transcriptome by altering selected nucleotides in RNA molecules. Although many editing sites have recently been discovered, the extent to which most sites are edited and how the editing is regulated in different biological contexts are not fully understood. Here we report dynamic spatiotemporal patterns and new regulators of RNA editing, discovered through an extensive profiling of A-to-I RNA editing in 8,551 human samples (representing 53 body sites from 552 individuals) from the Genotype-Tissue Expression (GTEx) project and in hundreds of other primate and mouse samples. We show that editing levels in non-repetitive coding regions vary more between tissues than editing levels in repetitive regions. Globally, ADAR1 is the primary editor of repetitive sites and ADAR2 is the primary editor of non-repetitive coding sites, whereas the catalytically inactive ADAR3 predominantly acts as an inhibitor of editing. Cross-species analysis of RNA editing in several tissues revealed that species, rather than tissue type, is the primary determinant of editing levels, suggesting stronger cis-directed regulation of RNA editing for most sites, although the small set of conserved coding sites is under stronger trans-regulation. In addition, we curated an extensive set of ADAR1 and ADAR2 targets and showed that many editing sites display distinct tissue-specific regulation by the ADAR enzymes in vivo. Further analysis of the GTEx data revealed several potential regulators of editing, such as AIMP2, which reduces editing in muscles by enhancing the degradation of the ADAR proteins. Collectively, our work provides insights into the complex cis- and trans-regulation of A-to-I editing.


Assuntos
Adenosina Desaminase , Primatas/genética , Edição de RNA/genética , Proteínas de Ligação a RNA , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Feminino , Genótipo , Células HEK293 , Humanos , Masculino , Camundongos , Músculos/metabolismo , Proteínas Nucleares/metabolismo , Especificidade de Órgãos/genética , Proteólise , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Análise Espaço-Temporal , Especificidade da Espécie , Transcriptoma/genética
7.
Nat Commun ; 8: 15772, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28598431

RESUMO

The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo.


Assuntos
Antineoplásicos/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Peptidilprolil Isomerase de Interação com NIMA/antagonistas & inibidores , Animais , Antineoplásicos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Inibidores Enzimáticos/química , Feminino , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Camundongos Nus , Peptidilprolil Isomerase de Interação com NIMA/química , Peptidilprolil Isomerase de Interação com NIMA/genética , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Cell Death Differ ; 24(2): 200-211, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27834957

RESUMO

Mammary gland development, various stages of mammary tumorigenesis and breast cancer progression have the peptidyl-prolyl cis/trans isomerase PIN1 at their centerpiece, in virtue of the ability of this unique enzyme to fine-tune the dynamic crosstalk between multiple molecular pathways. PIN1 exerts its action by inducing conformational and functional changes on key cellular proteins, following proline-directed phosphorylation. Through this post-phosphorylation signal transduction mechanism, PIN1 controls the extent and direction of the cellular response to a variety of inputs, in physiology and disease. This review discusses PIN1's roles in normal mammary development and cancerous progression, as well as the clinical impact of targeting this enzyme in breast cancer patients.


Assuntos
Neoplasias da Mama/patologia , Glândulas Mamárias Humanas/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição E2F/metabolismo , Inibidores Enzimáticos/uso terapêutico , Feminino , Instabilidade Genômica , Humanos , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Peptidilprolil Isomerase de Interação com NIMA/antagonistas & inibidores , Peptidilprolil Isomerase de Interação com NIMA/genética , Transdução de Sinais
9.
Nat Struct Mol Biol ; 22(5): 417-24, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25895060

RESUMO

The DNA-damage response (DDR) ensures genome stability and proper inheritance of genetic information, both of which are essential to survival. It is presently unclear to what extent other signaling pathways modulate DDR function. Here we show that Notch receptor binds and inactivates ATM kinase and that this mechanism is evolutionarily conserved in Caenorhabditis elegans, Xenopus laevis and humans. In C. elegans, the Notch pathway impairs DDR signaling in gonad germ cells. In mammalian cells, activation of human Notch1 leads to reduced ATM signaling in a manner independent of Notch1 transcriptional activity. Notch1 binds directly to the regulatory FATC domain of ATM and inhibits ATM kinase activity. Notch1 and ATM activation are inversely correlated in human breast cancers, and inactivation of ATM by Notch1 contributes to the survival of Notch1-driven leukemia cells upon DNA damage.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Caenorhabditis elegans/metabolismo , Reparo do DNA/genética , Receptor Notch1/metabolismo , Xenopus laevis/metabolismo , Animais , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Sítios de Ligação , Linhagem Celular Tumoral , Dano ao DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/genética , Células HEK293 , Células HeLa , Humanos , Complexos Multiproteicos , Neoplasias/genética , Ligação Proteica , Estrutura Terciária de Proteína , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Transdução de Sinais/genética , Transcrição Gênica/genética
10.
Biochimie ; 112: 85-95, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25742740

RESUMO

BACKGROUND: The modest efficacy of available therapies for Hepatocellular carcinoma (HCC) indicates the need to develop novel therapeutic approaches. For the proteasome inhibitor Bortezomib (BZB), potentially attractive for HCC treatment, the mechanism of action is largely unknown. The BZB effect on E2Fs and the E2Fs control on the peptidylproline cis-trans isomerase (Pin1), prompted us to explore the BZB effect on the Pin1-E2F1 axis. METHODS: The tumorigenic cell line HuH7 together with the non-tumorigenic cells IHH and the human pluripotent stem cell derived hepatocytes (hPSC-H), were used as cellular models of HCC and normal liver cells, respectively. RESULTS: BZB reduces HuH7 growth as shown by cell counting, cell vitality test and cell cycle analysis; this is paralleled by the decrease of Pin1, E2F1, cyclin A2 and of the hyper-phosphorylated pRB. Pin1-E2F1 axis impairment justifies the anti-proliferative effect since Pin-E2F1 depletion decreases HuH7 growth while the over-expression rescues BZB-induced inhibition of proliferation. Moreover, Pin1-E2F1 promote HuH7 growth via the up-regulation of cyclin D1, cyclin E, cyclin A2, E2F2 and in part E2F3. Finally, in the control cells IHH and hPSC-H, BZB effect on cell vitality is not irrelevant, a fact correlated to the cellular proliferation rate. Thus, BZB effect on healthy liver tissue may not be entirely negligible hence caution should be exercised in its use in liver regeneration processes. CONCLUSION: For the first time we prove the functional involvement of the Pin1-E2F1 axis in the anti-proliferative effect of BZB indicating Pin1-E2F as an attractive target to control HCC cell growth.


Assuntos
Bortezomib/farmacologia , Carcinoma Hepatocelular/metabolismo , Fator de Transcrição E2F1/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Peptidilprolil Isomerase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Fator de Transcrição E2F1/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Peptidilprolil Isomerase de Interação com NIMA , Proteínas de Neoplasias/genética , Peptidilprolil Isomerase/genética
11.
Biochim Biophys Acta ; 1850(10): 2048-60, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25641576

RESUMO

BACKGROUND: The p53 protein family, comprising p53, p63 and p73, is primarily involved in preserving genome integrity and preventing tumor onset, and also affects a range of physiological processes. Signal-dependent modifications of its members and of other pathway components provide cells with a sophisticated code to transduce a variety of stress signaling into appropriate responses. TP53 mutations are highly frequent in cancer and lead to the expression of mutant p53 proteins that are endowed with oncogenic activities and sensitive to stress signaling. SCOPE OF REVIEW: p53 family proteins have unique structural and functional plasticity, and here we discuss the relevance of prolyl-isomerization to actively shape these features. MAJOR CONCLUSIONS: The anti-proliferative functions of the p53 family are carefully activated upon severe stress and this involves the interaction with prolyl-isomerases. In particular, stress-induced stabilization of p53, activation of its transcriptional control over arrest- and cell death-related target genes and of its mitochondrial apoptotic function, as well as certain p63 and p73 functions, all require phosphorylation of specific S/T-P motifs and their subsequent isomerization by the prolyl-isomerase Pin1. While these functions of p53 counteract tumorigenesis, under some circumstances their activation by prolyl-isomerases may have negative repercussions (e.g. tissue damage induced by anticancer therapies and ischemia-reperfusion, neurodegeneration). Moreover, elevated Pin1 levels in tumor cells may transduce deregulated phosphorylation signaling into activation of mutant p53 oncogenic functions. GENERAL SIGNIFICANCE: The complex repertoire of biological outcomes induced by p53 finds mechanistic explanations, at least in part, in the association between prolyl-isomerases and the p53 pathway. This article is part of a Special Issue entitled Proline-directed foldases: Cell signaling catalysts and drug targets.


Assuntos
Apoptose , Proliferação de Células , Peptidilprolil Isomerase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peptidilprolil Isomerase/química , Peptidilprolil Isomerase/genética , Fosforilação , Estabilidade Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
12.
Eur Heart J ; 36(13): 817-28, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24801072

RESUMO

AIM: Diabetes is a major driver of cardiovascular disease, but the underlying mechanisms remain elusive. Prolyl-isomerase Pin1 recognizes specific peptide bonds and modulates function of proteins altering cellular homoeostasis. The present study investigates Pin1 role in diabetes-induced vascular disease. METHODS AND RESULTS: In human aortic endothelial cells (HAECs) exposed to high glucose, up-regulation of Pin1-induced mitochondrial translocation of pro-oxidant adaptor p66(Shc) and subsequent organelle disruption. In this setting, Pin1 recognizes Ser-116 inhibitory phosphorylation of endothelial nitric oxide synthase (eNOS) leading to eNOS-caveolin-1 interaction and reduced NO availability. Pin1 also mediates hyperglycaemia-induced nuclear translocation of NF-κB p65, triggering VCAM-1, ICAM-1, and MCP-1 expression. Indeed, gene silencing of Pin1 in HAECs suppressed p66(Shc)-dependent ROS production, restored NO release and blunted NF-kB p65 nuclear translocation. Consistently, diabetic Pin1(-/-) mice were protected against mitochondrial oxidative stress, endothelial dysfunction, and vascular inflammation. Increased expression and activity of Pin1 were also found in peripheral blood monocytes isolated from diabetic patients when compared with age-matched healthy controls. Interestingly, enough, Pin1 up-regulation was associated with impaired flow-mediated dilation, increased urinary 8-iso-prostaglandin F2α and plasma levels of adhesion molecules. CONCLUSIONS: Pin1 drives diabetic vascular disease by causing mitochondrial oxidative stress, eNOS dysregulation as well as NF-kB-induced inflammation. These findings provide molecular insights for novel mechanism-based therapeutic strategies in patients with diabetes.


Assuntos
Angiopatias Diabéticas/prevenção & controle , Doenças Mitocondriais/prevenção & controle , Estresse Oxidativo/fisiologia , Peptidilprolil Isomerase/fisiologia , Análise de Variância , Animais , Aorta/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CCL2/metabolismo , Citocromos c/biossíntese , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Humanos , Hiperglicemia/fisiopatologia , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Peptidilprolil Isomerase de Interação com NIMA , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Regulação para Cima/fisiologia , Molécula 1 de Adesão de Célula Vascular/metabolismo , Vasculite/fisiopatologia
13.
Hum Mol Genet ; 23(14): 3830-42, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24569166

RESUMO

Ectrodactyly, or Split-Hand/Foot Malformation (SHFM), is a congenital condition characterized by the loss of central rays of hands and feet. The p63 and the DLX5;DLX6 transcription factors, expressed in the embryonic limb buds and ectoderm, are disease genes for these conditions. Mutations of p63 also cause the ectodermal dysplasia-ectrodactyly-cleft lip/palate (EEC) syndrome, comprising SHFM. Ectrodactyly is linked to defects of the apical ectodermal ridge (AER) of the developing limb buds. FGF8 is the key signaling molecule in this process, able to direct proximo-distal growth and patterning of the skeletal primordial of the limbs. In the limb buds of both p63 and Dlx5;Dlx6 murine models of SHFM, the AER is poorly stratified and FGF8 expression is severely reduced. We show here that the FGF8 locus is a downstream target of DLX5 and that FGF8 counteracts Pin1-ΔNp63α interaction. In vivo, lack of Pin1 leads to accumulation of the p63 protein in the embryonic limbs and ectoderm. We show also that ΔNp63α protein stability is negatively regulated by the interaction with the prolyl-isomerase Pin1, via proteasome-mediated degradation; p63 mutant proteins associated with SHFM or EEC syndromes are resistant to Pin1 action. Thus, DLX5, p63, Pin1 and FGF8 participate to the same time- and location-restricted regulatory loop essential for AER stratification, hence for normal patterning and skeletal morphogenesis of the limb buds. These results shed new light on the molecular mechanisms at the basis of the SHFM and EEC limb malformations.


Assuntos
Ectoderma/embriologia , Fator 8 de Crescimento de Fibroblasto/metabolismo , Proteínas de Homeodomínio/metabolismo , Deformidades Congênitas dos Membros/metabolismo , Peptidilprolil Isomerase/metabolismo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Animais , Padronização Corporal , Linhagem Celular , Modelos Animais de Doenças , Ectoderma/metabolismo , Técnicas de Inativação de Genes , Proteínas de Homeodomínio/genética , Humanos , Botões de Extremidades/embriologia , Deformidades Congênitas dos Membros/patologia , Camundongos , Peptidilprolil Isomerase de Interação com NIMA , Fosfoproteínas/genética , Estabilidade Proteica , Transativadores/genética
14.
EMBO Mol Med ; 6(1): 99-119, 2014 01.
Artigo em Inglês | MEDLINE | ID: mdl-24357640

RESUMO

Mammary epithelial stem cells are fundamental to maintain tissue integrity. Cancer stem cells (CSCs) are implicated in both treatment resistance and disease relapse, and the molecular bases of their malignant properties are still poorly understood. Here we show that both normal stem cells and CSCs of the breast are controlled by the prolyl-isomerase Pin1. Mechanistically, following interaction with Pin1, Notch1 and Notch4, key regulators of cell fate, escape from proteasomal degradation by their major ubiquitin-ligase Fbxw7α. Functionally, we show that Fbxw7α acts as an essential negative regulator of breast CSCs' expansion by restraining Notch activity, but the establishment of a Notch/Pin1 active circuitry opposes this effect, thus promoting breast CSCs self-renewal, tumor growth and metastasis in vivo. In human breast cancers, despite Fbxw7α expression, high levels of Pin1 sustain Notch signaling, which correlates with poor prognosis. Suppression of Pin1 holds promise in reverting aggressive phenotypes, through CSC exhaustion as well as recovered drug sensitivity carrying relevant implications for therapy of breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Peptidilprolil Isomerase/metabolismo , Células-Tronco/metabolismo , Animais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 com Repetições F-Box-WD , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Camundongos , Camundongos Knockout , Camundongos SCID , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/antagonistas & inibidores , Peptidilprolil Isomerase/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptor Notch4 , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Transplante Heterólogo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
J Toxicol Pathol ; 26(4): 423-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24526816

RESUMO

A nephroblastoma is a tumor arising from metanephric blastema occurring in childhood. Among laboratory rodents, nephroblastoma has been frequently reported in rats, but it remains exceedingly rare in mice. The present work describes a nephroblastoma in a young mouse homozygous for the specific Trp53 R172H point mutation coupled with targeted deletion of the Pin1 gene. The affected kidney was effaced by a biphasic tumor with an epithelial component arranged in tubules surrounded by nests of blastemal cells. Immunohistochemically, the neoplasm was diffusely positive for Wilms' tumor antigen. The epithelial component expressed markers of renal tubular differentiation including wide-spectrum cytokeratin, E-cadherin and folate-binding protein. Furthermore, the neoplasm exhibited a high proliferative index and diffuse nucleocytoplasmic ß-catenin expression. Based on histological and immunohistochemical features, a diagnosis of nephroblastoma potentially associated with Trp53 loss and oncogenic ß-catenin activation has been proposed.

16.
Toxicol Pathol ; 40(4): 675-81, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22328409

RESUMO

Giant cell tumor of bone (GCTB) is a common primary neoplasm of bone characterized by distinctive clinicopathological features. GCTB is exceedingly rare in nonhuman species, and it has been sporadically reported in cats, dogs, rats, and birds. This report describes a primary murine bone tumor that shares striking clinicopathological similarities with human GCTB. The neoplasm occurred in a 71-week-old C57BL/6 mouse heterozygous for the specific Trp53 R172H point mutation. Grossly, the tumor presented as a mono-ostotic nodular mass arising from the distal metaphysis of the right femur. Microscopically, the affected bone was effaced by an osteolytic neoplasm with focal infiltrations into the surrounding tissues. Similarly to what was reported for human GCTB, the murine neoplasm consisted of 3 main cell populations: (1) bundles of pleomorphic spindle-shaped mononuclear cells displaying an indefinite mesenchymal histogenesis with immunohistochemical expression of vimentin and smooth muscle actin, (2) scattered multinucleated giant cells exhibiting osteoclast differentiation with prominent tartrate-resistant acid phosphatase activity and immunoreactivity for monocyte/macrophage markers including CD45 and lysozyme, and (3) scattered round mononuclear cells consistent with activated macrophages and expressing CD45, lysozyme, and F4/80. Based on these morphological and immunohistological results, the murine bone tumor described in this study has been putatively classified as GCTB.


Assuntos
Neoplasias Ósseas/genética , Tumor de Células Gigantes do Osso/genética , Proteína Supressora de Tumor p53/genética , Animais , Neoplasias Ósseas/diagnóstico , Neoplasias Ósseas/patologia , Feminino , Tumor de Células Gigantes do Osso/diagnóstico , Tumor de Células Gigantes do Osso/patologia , Células Gigantes , Imuno-Histoquímica , Leucócitos Mononucleares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Mutação Puntual
17.
Cancer Cell ; 20(1): 79-91, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21741598

RESUMO

TP53 missense mutations dramatically influence tumor progression, however, their mechanism of action is still poorly understood. Here we demonstrate the fundamental role of the prolyl isomerase Pin1 in mutant p53 oncogenic functions. Pin1 enhances tumorigenesis in a Li-Fraumeni mouse model and cooperates with mutant p53 in Ras-dependent transformation. In breast cancer cells, Pin1 promotes mutant p53 dependent inhibition of the antimetastatic factor p63 and induction of a mutant p53 transcriptional program to increase aggressiveness. Furthermore, we identified a transcriptional signature associated with poor prognosis in breast cancer and, in a cohort of patients, Pin1 overexpression influenced the prognostic value of p53 mutation. These results define a Pin1/mutant p53 axis that conveys oncogenic signals to promote aggressiveness in human cancers.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Proteínas Mutantes/metabolismo , Peptidilprolil Isomerase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Camundongos , Modelos Biológicos , Proteínas Mutantes/genética , Peptidilprolil Isomerase de Interação com NIMA , Invasividade Neoplásica , Transcrição Gênica , Resultado do Tratamento , Proteína Supressora de Tumor p53/genética
18.
J Biol Chem ; 285(45): 34518-27, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20801874

RESUMO

Reversible proline-directed phosphorylation at Ser/Thr-Pro motifs has an essential role in myogenesis, a multistep process strictly regulated by several signaling pathways that impinge on two families of myogenic effectors, the basic helix-loop-helix myogenic transcription factors and the MEF2 (myocyte enhancer factor 2) proteins. The question of how these signals are deciphered by the myogenic effectors remains largely unaddressed. In this study, we show that the peptidyl-prolyl isomerase Pin1, which catalyzes the isomerization of phosphorylated Ser/Thr-Pro peptide bonds to induce conformational changes of its target proteins, acts as an inhibitor of muscle differentiation because its knockdown in myoblasts promotes myotube formation. With the aim of clarifying the mechanism of Pin1 function in skeletal myogenesis, we investigated whether MEF2C, a critical regulator of the myogenic program that is the end point of several signaling pathways, might serve as a/the target for the inhibitory effects of Pin1 on muscle differentiation. We show that Pin1 interacts selectively with phosphorylated MEF2C in skeletal muscle cells, both in vitro and in vivo. The interaction with Pin1 requires two novel critical phospho-Ser/Thr-Pro motifs in MEF2C, Ser(98) and Ser(110), which are phosphorylated in vivo. Overexpression of Pin1 decreases MEF2C stability and activity and its ability to cooperate with MyoD to activate myogenic conversion. Collectively, these findings reveal a novel role for Pin1 as a regulator of muscle terminal differentiation and suggest that Pin1-mediated repression of MEF2C function could contribute to this function.


Assuntos
Proliferação de Células , Desenvolvimento Muscular/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Fatores de Regulação Miogênica/metabolismo , Peptidilprolil Isomerase/metabolismo , Transdução de Sinais/fisiologia , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Humanos , Fatores de Transcrição MEF2 , Camundongos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fatores de Regulação Miogênica/genética , Peptidilprolil Isomerase de Interação com NIMA , Peptídeos/genética , Peptídeos/metabolismo , Peptidilprolil Isomerase/genética , Fosforilação/fisiologia , Estabilidade Proteica
19.
Cancer Res ; 69(3): 1016-26, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19155306

RESUMO

The peptidyl-prolyl-isomerase Pin1 interacts with phosphorylated proteins, altering their conformation. The retinoic acid receptor RARalpha and the acute-promyelocytic-leukemia-specific counterpart PML-RARalpha directly interact with Pin1. Overexpression of Pin1 inhibits ligand-dependent activation of RARalpha and PML-RARalpha. Inhibition is relieved by Pin1-targeted short interfering RNAs and by pharmacologic inhibition of the catalytic activity of the protein. Mutants of Pin1 catalytically inactive or defective for client-protein-binding activity are incapable of inhibiting ligand-dependent RARalpha transcriptional activity. Functional inhibition of RARalpha and PML-RARalpha by Pin1 correlates with degradation of the nuclear receptors via the proteasome-dependent pathway. In the acute myelogenous leukemia cell lines HL-60 and NB4, Pin1 interacts with RARalpha in a constitutive fashion. Suppression of Pin1 by a specific short hairpin RNA in HL-60 or NB4 cells stabilizes RARalpha and PML-RARalpha, resulting in increased sensitivity to the cytodifferentiating and antiproliferative activities of all-trans retinoic acid. Treatment of the two cell lines and freshly isolated acute myelogenous leukemia blasts (M1 to M4) with ATRA and a pharmacologic inhibitor of Pin1 causes similar effects. Our results add a further layer of complexity to the regulation of nuclear retinoic acid receptors and suggest that Pin1 represents an important target for strategies aimed at increasing the therapeutic index of retinoids.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Peptidilprolil Isomerase/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Doença Aguda , Animais , Células COS , Chlorocebus aethiops , Células HL-60 , Humanos , Leucemia Mieloide/enzimologia , Leucemia Mieloide/genética , Peptidilprolil Isomerase de Interação com NIMA , Proteínas de Fusão Oncogênica/genética , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Receptores do Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Ativação Transcricional , Transfecção
20.
Nat Cell Biol ; 11(2): 133-42, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19151708

RESUMO

Signalling through Notch receptors requires ligand-induced cleavage to release the intracellular domain, which acts as a transcriptional activator in the nucleus. Deregulated Notch1 signalling has been implicated in mammary tumorigenesis; however the mechanisms underlying Notch activation in breast cancer remain unclear. Here, we demonstrate that the prolyl-isomerase Pin1 interacts with Notch1 and affects Notch1 activation. Pin1 potentiates Notch1 cleavage by gamma-secretase, leading to an increased release of the active intracellular domain and ultimately enhancing Notch1 transcriptional and tumorigenic activity. We found that Notch1 directly induces transcription of Pin1, thereby generating a positive loop. In human breast cancers, we observed a strong correlation between Pin1 overexpression and high levels of activated Notch1. Thus, the molecular circuitry established by Notch1 and Pin1 may have a key role in cancer.


Assuntos
Neoplasias/enzimologia , Neoplasias/genética , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ativação Enzimática/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Peptidilprolil Isomerase de Interação com NIMA , Neoplasias/fisiopatologia , Estrutura Terciária de Proteína/genética , Ativação Transcricional/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...