Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Biochim Biophys Acta ; 1834(10): 2219-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23777859

RESUMO

Anaplastic lymphoma kinase is essential in early development, differentiation, and maintenance of cell survival; nevertheless, the mechanism to activate ALK has remained elusive. ALK has remained an "Orphan Receptor." The studies cited below describe a unique mechanism termed "Ligand Independent Activation." It is shown that activation of ALK results when the cytokine pleiotrophin (PTN) interacts with its receptor, the receptor protein tyrosine phosphatase ß/ζ (RPTPß/ζ). Pleiotrophin inactivates the catalytic activity of RPTPß/ζ, which, when not inactivated, dephosphorylates phosphotyrosine sites in the activation domain of ALK; as a consequence of the inactivation of RPTPß/ζ by PTN, autophosphorylation and autoactivation of ALK rapidly follow. The PTN/RPTPß/ζ signaling pathway thus regulates the catalytic activity of ALK and tyrosine phosphorylation levels of ALK downstream target proteins. Furthermore, since ALK is only one of the key ALK phosphoproteins targeted by the PTN/RPTPß/ζ signaling pathway, the PTN/RPTPß/ζ signaling pathway has the potential to coordinately regulate tyrosine phosphorylation of other different key proteins in multiple cellular compartments. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.


Assuntos
Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Neurônios/metabolismo , Fosfotirosina/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/metabolismo , Quinase do Linfoma Anaplásico , Proteínas de Transporte/genética , Citocinas/genética , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Ligantes , Neurônios/citologia , Fosforilação , Estrutura Terciária de Proteína , Receptores Proteína Tirosina Quinases/genética , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/genética , Transdução de Sinais , Tirosina/metabolismo
2.
Blood ; 113(9): 1992-2002, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19060246

RESUMO

Enhanced angiogenesis is a hallmark of cancer. Pleiotrophin (PTN) is an angiogenic factor that is produced by many different human cancers and stimulates tumor blood vessel formation when it is expressed in malignant cancer cells. Recent studies show that monocytes may give rise to vascular endothelium. In these studies, we show that PTN combined with macrophage colony-stimulating factor (M-CSF) induces expression of vascular endothelial cell (VEC) genes and proteins in human monocyte cell lines and monocytes from human peripheral blood (PB). Monocytes induce VEC gene expression and develop tube-like structures when they are exposed to serum or cultured with bone marrow (BM) from patients with multiple myeloma (MM) that express PTN, effects specifically blocked with antiPTN antibodies. When coinjected with human MM cells into severe combined immunodeficient (SCID) mice, green fluorescent protein (GFP)-marked human monocytes were found incorporated into tumor blood vessels and expressed human VEC protein markers and genes that were blocked by anti-PTN antibody. Our results suggest that vasculogenesis in human MM may develop from tumoral production of PTN, which orchestrates the transdifferentiation of monocytes into VECs.


Assuntos
Proteínas de Transporte/farmacologia , Transdiferenciação Celular/efeitos dos fármacos , Citocinas/farmacologia , Células Endoteliais/fisiologia , Monócitos/efeitos dos fármacos , Mieloma Múltiplo/metabolismo , Neovascularização Patológica/etiologia , Animais , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/metabolismo , Células Cultivadas , Citocinas/administração & dosagem , Citocinas/metabolismo , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Humanos , Fator Estimulador de Colônias de Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Camundongos , Camundongos SCID , Camundongos Transgênicos , Monócitos/patologia , Monócitos/fisiologia , Mieloma Múltiplo/patologia , Neovascularização Patológica/induzido quimicamente , Transplante Heterólogo , Células U937
3.
Addict Biol ; 15(4): 403-12, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20192945

RESUMO

Pleiotrophin (PTN), a neurotrophic factor with important roles in survival and differentiation of dopaminergic neurons, is up-regulated in the nucleus accumbens after amphetamine administration suggesting that PTN could modulate amphetamine-induced pharmacological or neuroadaptative effects. To test this hypothesis, we have studied the effects of amphetamine administration in PTN genetically deficient (PTN -/-) and wild type (WT, +/+) mice. In conditioning studies, we found that amphetamine induces conditioned place preference in both PTN -/- and WT (+/+) mice. When these mice were re-evaluated after a 5-day period without amphetamine administration, we found that WT (+/+) mice did not exhibit amphetamine-seeking behaviour, whereas, PTN -/- mice still showed a robust drug-seeking behaviour. In immunohystochemistry studies, we found that amphetamine (10 mg/kg, four times, every 2 hours) causes a significant increase of glial fibrillary acidic protein positive cells in the striatum of amphetamine-treated PTN -/- mice compared with WT mice 4 days after last administration of the drug, suggesting an enhanced amphetamine-induced astrocytosis in the absence of endogenous PTN. Interestingly, we found in concomitant in vitro studies that PTN (3 µM) limits amphetamine (1 mM)-induced loss of viability of PC12 cell cultures, effect that could be related to the ability of PTN to induce the phosphorylation of Akt and ERK1/2. To test this possibility, we used specific Akt and ERK1/2 inhibitors uncovering for the first time that PTN-induced protective effects against amphetamine-induced toxicity in PC12 cells are mediated by the ERK1/2 signalling pathway. The data suggest an important role of PTN to limit amphetamine-induced neurotoxic and rewarding effects.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/genética , Anfetamina/toxicidade , Proteínas de Transporte/genética , Sobrevivência Celular/genética , Condicionamento Clássico/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Citocinas/genética , Dopamina/metabolismo , Motivação/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Condicionamento Clássico/fisiologia , Corpo Estriado/fisiopatologia , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/induzido quimicamente , Gliose/fisiopatologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Motivação/fisiologia , Núcleo Accumbens/fisiopatologia , Células PC12 , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
4.
Proc Natl Acad Sci U S A ; 104(26): 10888-93, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17578909

RESUMO

Pleiotrophin (PTN, Ptn) is an 18-kDa secretory cytokine expressed in many breast cancers; however, the significance of Ptn expression in breast cancer has not been established. We have now tested three models to determine the role of inappropriate expression of Ptn in breast cancer. Mouse mammary tumor virus (MMTV) promoter-driven Ptn expressed in MMTV-polyoma virus middle T antigen (PyMT)-Ptn mouse breast cancers was first shown to induce rapid growth of morphologically identified foci of "scirrhous" carcinoma and to extensively remodel the microenvironment, including increased tumor angiogenesis and striking increases in mouse protocollagens Ialpha2, IValpha5, and XIalpha1, and elastin. Ectopic Ptn expression in MCF-7 (human breast cancer)-Ptn cell xenografts also was shown to markedly increase MCF-7-Ptn cell xenograft growth in nude mice; furthermore, it induced extensive remodeling of the microenvironment and tumor angiogenesis. In a coculture model of equal numbers of NIH 3T3 stromal fibroblasts and MCF-7-Ptn cells, PTN secreted from MCF-7-Ptn cells was then shown to induce a more malignant MCF-7-Ptn breast cancer cell phenotype and extensive remodeling of the MCF-7-Ptn/NIH 3T3 cell microenvironment; it up-regulated expression of markers of aggressive breast cancers, including PKCdelta and matrix metalloproteinase-9 in both MCF-7-Ptn and NIH 3T3 cells. The morphological phenotypes of MCF-7-Ptn cell xenografts and MCF-7-Ptn cell/NIH 3T3 cell cocultures closely resembled breast cancers in MMTV-PyMT-Ptn mice. Inappropriate expression of Ptn thus promotes breast cancer progression in mice; the data suggest that secretion of PTN through stimulation of the stromal cell microenvironment alone may be sufficient to account for significant features of breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Processos Neoplásicos , Comunicação Parácrina , Animais , Neoplasias da Mama/etiologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Progressão da Doença , Feminino , Substâncias de Crescimento , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Células Estromais , Transplante Heterólogo
5.
Growth Factors ; 27(3): 189-94, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19384682

RESUMO

Pleiotrophin (PTN) is a growth factor that has been shown to be involved in hippocampal synaptic plasticity and learning. To further understand the involvement of PTN in memory processes, we performed in vitro electrophysiological studies in PTN-stimulated CA1 from rat hippocampal slices combined with the behavioural testing of PTN deficient (PTN - / - ) mice. We found that PTN inhibited hippocampal long-term potentiation (LTP) induced by high-frequency stimulation (HFS) consisted in three trains of 100 Hz separated by 20 s. To test the possibility that PTN might be involved in behavioural memory processes, we tested the learning behaviour of PTN - / - mice using the Y-maze test. We did not observe significant differences in recognition memory between PTN - / - and Wild Type (WT) mice when a 30 min-interval intertrial (ITI) was used in the Y-maze test. However, whereas WT mice showed disruption of recognition memory using a 60 min-ITI, PTN - / - mice maintained the recognition memory. The data demonstrate that PTN inhibits hippocampal LTP in vitro and might play a role in memory processes in vivo.


Assuntos
Proteínas de Transporte/fisiologia , Citocinas/fisiologia , Hipocampo/fisiologia , Aprendizagem/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Animais , Proteínas de Transporte/genética , Citocinas/genética , Estimulação Elétrica , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley
6.
Eur J Pharmacol ; 557(2-3): 147-50, 2007 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17157293

RESUMO

Pleiotrophin and midkine are two recently discovered growth factors that promote survival and differentiation of catecholaminergic neurons. Chronic opioid stimulation has been reported to induce marked alterations of the locus coeruleus-hippocampus noradrenergic pathway, an effect that is prevented when opioids are coadministered with the alpha2-adrenoceptor antagonist yohimbine. The present work tries to examine a possible link between yohimbine reversal of morphine effects and pleiotrophin/midkine activation in the rat hippocampus by studying the levels of expression of pleiotrophin and midkine in response to acute and chronic administration of morphine, yohimbine and combinations of both drugs. Pleiotrophin gene expression was not altered by any treatment; however midkine mRNA levels were increased after chronic treatment with morphine. Chronic administration of yohimbine alone also increased midkine expression levels, whereas yohimbine and morphine administered together exhibited summatory effects on the upregulation of midkine expression levels. The data suggest that midkine could play a role in the prevention of opioid-induced neuroadaptations in hippocampus by yohimbine.


Assuntos
Analgésicos Opioides/farmacologia , Citocinas/metabolismo , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Morfina/farmacologia , Ioimbina/farmacologia , Animais , Citocinas/genética , Hipocampo/metabolismo , Masculino , Midkina , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Neurosci Lett ; 412(2): 163-7, 2007 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-17123717

RESUMO

The alpha(2)-adrenoceptor antagonist yohimbine is known to oppose to several pharmacological effects of opioid drugs, but the consequences and the mechanisms involved remain to be clearly established. In the present study we have checked the effects of yohimbine on morphine-induced alterations of the expression of key proteins (glial fibrillary acidic protein, GFAP) and genes (alpha(2)-adrenoceptors) in rat brain areas known to be relevant in opioid dependence, addiction and individual vulnerability to drug abuse. Rats were treated with morphine in the presence or absence of yohimbine. The effects of the treatments on GFAP expression were studied by immunohistochemical staining in Locus Coeruleus (LC) and Nucleus of the Solitary Tract (NST), two important noradrenergic nuclei. In addition, drug effects on alpha(2)-adrenoceptor gene expression were determined by real time RT-PCR in the hippocampus, a brain area that receives noradrenergic input from the brainstem. Morphine administration increased GFAP expression both in LC and NST as it was previously reported in other brain areas. Yohimbine was found to efficiently prevent morphine-induced GFAP upregulation. Chronic (but not acute) morphine downregulated mRNA levels of alpha(2A)- and alpha(2C)-adrenoceptors in the hippocampus, while simultaneously increased the expression of the alpha(2B)-adrenoceptor gene. Again, yohimbine was able to prevent morphine-induced changes in the levels of expression of the three alpha(2)-adrenoceptor genes. These results correlate the well-established reduction of opioid dependence and addiction by yohimbine and suggest that this drug could interfere with the neural plasticity induced by chronic morphine in central noradrenergic pathways.


Assuntos
Tronco Encefálico/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/efeitos dos fármacos , Morfina/farmacologia , Receptores Adrenérgicos alfa 2/genética , Ioimbina/farmacologia , Antagonistas Adrenérgicos alfa/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Vias Autônomas/efeitos dos fármacos , Vias Autônomas/metabolismo , Biomarcadores/metabolismo , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Morfina/antagonistas & inibidores , Dependência de Morfina/tratamento farmacológico , Dependência de Morfina/metabolismo , Entorpecentes/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Life Sci ; 79(11): 1049-55, 2006 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-16643958

RESUMO

To discover regulatory pathways dependent on midkine (Mk the gene, MK the protein) signaling, we compared the transcriptional profiles of aortae obtained from Mk -/- and wild type (WT, +/+) mice; the comparison demonstrated an extraordinary high level expression of tyrosine hydroxylase (12-fold), the rate-limiting enzyme in catecholamine biosynthesis, DOPA decarboxylase (73-fold), and dopamine beta-hydroxylase (75-fold) in aortae of Mk -/- mice compared with aortae of WT (+/+) mice. Phenylethanolamine-N-methyltransferase, the enzyme catalyzing the conversion of norepinephrine into epinephrine, was not detected in either Mk -/- and WT (+/+) mouse aorta. The protein levels of tyrosine hydroxylase, DOPA decarboxylase and dopamine beta-hydroxylase confirmed the analysis of the transcriptional profiles. Surprisingly, MK failed to regulate the enzymes of the catecholamine biosynthesis pathway in 10 other tissues studied. Furthermore, the expression levels of the enzymes of catecholamine biosynthesis in aortae of Mk -/- mice were effectively the same as those in aortae of Pleiotrophin (Ptn the gene, PTN the protein) genetically deficient (Ptn -/-) mice when compared with WT (+/+) mice. The remarkable increases in levels of expression of tyrosine hydroxylase, DOPA decarboxylase and dopamine beta-hydroxylase suggest that MK together with PTN are very important regulators of the catecholamine pathway in mouse aorta and may critically regulate catecholamine biosynthesis and function in inflammatory and the other pathological conditions in which Mk or Ptn are upregulated. The data also establish that norepinephrine is effectively the only catecholamine synthesized in mouse aorta.


Assuntos
Catecolaminas/biossíntese , Citocinas/fisiologia , Dopa Descarboxilase/genética , Regulação Enzimológica da Expressão Gênica , Oxigenases de Função Mista/genética , Animais , Aorta/enzimologia , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Catecolaminas/genética , Citocinas/genética , Dopa Descarboxilase/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Midkina , Oxigenases de Função Mista/metabolismo , Norepinefrina/biossíntese , Norepinefrina/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo
9.
Anticancer Res ; 23(5A): 3735-40, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14666671

RESUMO

The retinoblastoma (Rb) suppressor associated protein 46 (RbAp46) is a subunit of chromatin modifying and remodeling complexes. Previously, we found that RbAp46 functions as a potent growth inhibitor. It is also a downstream effector of the Wilms' tumor suppressor, WT1. The findings that expression levels of WT1 were down-regulated in breast cancer cell lines and in subsets of primary breast tumors led us to investigate the possible role of RbAp46 in breast cancer tumorigenesis. Here, we found that RbAp46 expression levels were decreased in five established breast cancer cell lines compared to a normal mammary gland epithelial cell line. To investigate the effect of constitutive expression of RbAp46 on the transformed phenotypes of breast cancer cells, we established stable cell lines that constitutively express exogenous RbAp46 using three breast cancer cell lines, MCF-7, MDA-MB-231 and MDA-MB-436. We have found that RbAp46 expression suppressed colony formation of these breast cancer cells in soft-agar, and inhibited tumor formation of these cells in nude mice. Our data demonstrated that constitutive RbAp46 expression suppresses the transformed phenotypes of breast cancer cells, and suggested that dysregulation of RbAp46 expression may contribute to breast cancer tumorigenesis.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Transporte/fisiologia , Transformação Celular Neoplásica/patologia , Proteínas Nucleares/fisiologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteína 7 de Ligação ao Retinoblastoma , Transfecção
10.
J Histochem Cytochem ; 60(5): 366-75, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22382872

RESUMO

Pleiotrophin (PTN) is an extracellular matrix-associated growth factor and chemokine expressed in mesodermal and ectodermal cells. It plays an important role in osteoblast recruitment and differentiation. There is limited information currently available about PTN expression during odontoblast differentiation and tooth formation, and thus the authors aimed to establish the spatiotemporal expression pattern of PTN during mouse odontogenesis. Immortalized mouse dental pulp (MD10-D3, MD10-A11) and odontoblast-like (M06-G3) and ameloblast-like (EOE-3M) cell lines were grown and samples prepared for immunocytochemistry, Western blot, and conventional and quantitative PCR analysis. Effects of BMP2, BMP4, and BMP7 treatment on PTN expression in odontoblast-like M06-G3 cells were tested by quantitative PCR. Finally, immunohistochemistry of sectioned mice mandibles and maxillaries at developmental stages E16, E18, P1, P6, P10, and P28 was performed. The experiments showed that PTN, at both the mRNA and protein level, was expressed in all tested epithelial and mesenchymal dental cell lines and that the level of PTN mRNA was influenced differentially by the bone morphogenetic proteins. The authors observed initial expression of PTN in the inner enamel epithelium with prolonged expression in the ameloblasts and odontoblasts throughout their stages of maturation and strong expression in the terminally differentiated and enamel matrix-secreting ameloblasts and odontoblasts of the adult mouse incisors and molars.


Assuntos
Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Dente/metabolismo , Ameloblastos/metabolismo , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 4/farmacologia , Proteína Morfogenética Óssea 7/farmacologia , Proteínas de Transporte/genética , Linhagem Celular , Citocinas/genética , Polpa Dentária/metabolismo , Imuno-Histoquímica , Incisivo/embriologia , Incisivo/crescimento & desenvolvimento , Incisivo/metabolismo , Mesoderma/embriologia , Mesoderma/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Dente Molar/embriologia , Dente Molar/crescimento & desenvolvimento , Dente Molar/metabolismo , Odontoblastos/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dente/embriologia , Dente/crescimento & desenvolvimento
11.
Curr Opin Hematol ; 15(3): 210-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18391787

RESUMO

PURPOSE OF REVIEW: This study seeks to integrate recent studies that identify new critical mechanisms through which the 136 amino acid secreted heparin-binding cytokine pleiotrophin (PTN, Ptn) stimulates both normal and pathological angiogenesis. RECENT FINDINGS: Pleiotrophin is directly angiogenic; it initiates an angiogenic switch in different cancer models in vivo. It acts as an angiogenic factor through multiple mechanisms that include a unique signaling pathway that activates newly identified downstream tyrosine kinases through a unique mechanism, an interaction with endothelial cells to initiate proliferation, migration, and tube formation, the regulation of basic fibroblast growth factor and vascular endothelial growth factor signaling, the remodeling of the stromal microenvironment, and induction of transdifferentiation of monocytes into endothelial cells. Recently also, domains of PTN that stimulate angiogenesis and peptides that function to inhibit PTN signaling have been identified. SUMMARY: Recent studies have identified new mechanisms dependent on activation of the PTN signaling pathway that regulate angiogenesis and new targets to use PTN to both stimulate angiogenesis and block its activity to control pathological angiogenesis.


Assuntos
Proteínas de Transporte/fisiologia , Citocinas/fisiologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Animais , Células Endoteliais/fisiologia , Regulação da Expressão Gênica , Humanos , Camundongos , Transdução de Sinais/fisiologia
12.
Growth Factors ; 26(1): 44-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18365878

RESUMO

Pleiotrophin (PTN) and midkine (MK) are two growth factors highly redundant in function that exhibit neurotrophic actions and are upregulated at sites of nerve injury, both properties being compatible with a potential involvement in the pathophysiological events that follow nerve damage (i.e. neuropathic pain). We have tested this hypothesis by comparatively studying PTN and MK gene expression in the spinal cord and dorsal root ganglia (DRG) of three rat strains known to differ in their behavioural responses to chronic constriction injury (CCI) of the sciatic nerve: Lewis, Fischer 344 (F344) and Sprague-Dawley (SD). Real time RT-PCR revealed minimal changes in PTN/MK gene expression in the spinal cord after CCI despite the strain considered, but marked changes were detected in DRG. A significant upregulation of PTN gene expression occurred in injured DRG of the F344 strain, the only strain that recovers from CCI-induced mechanical allodynia 28 days after surgery. In contrast, PTN was found to be downregulated in injured DRG of SD rats, the most sensitive strain in behavioural studies. These changes in PTN were not paralleled by concomitant modifications of MK gene expression. The results demonstrate previously unidentified differences between PTN and MK patterns of expression. Furthermore, the data suggest that upregulation of PTN, but not MK, could play an important role in the recovery from CCI.


Assuntos
Proteínas de Transporte/biossíntese , Citocinas/biossíntese , Gânglios Espinais/patologia , Regulação da Expressão Gênica , Neurônios/metabolismo , Traumatismos dos Nervos Periféricos , Animais , Midkina , Dor , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Regulação para Cima
13.
Cell Cycle ; 6(23): 2877-83, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18156802

RESUMO

Pleiotrophin (PTN, Ptn) is a widely expressed, developmentally regulated 136 amino acid secreted heparin-binding cytokine. It signals through a unique signaling pathway; the PTN receptor is the transmembrane receptor protein tyrosine phosphatase (RPTP)beta/zeta. RPTPbeta/zeta is inactivated by PTN, which leads to increased tyrosine phosphorylation of the downstream targets of the PTN/RPTPbeta/zeta signaling pathway. Pleiotrophin gene expression is found in cells in early differentiation during different developmental periods. It is upregulated in cells with an early differentiation phenotype in wound repair. The Ptn gene also is a proto-oncogene; PTN is expressed in human tumor cells, and, in cell lines derived from human tumors that express Ptn, Ptn expression is constitutive and thus "inappropriate". Importantly, properties of different cells induced by PTN in PTN-stimulated cells are strikingly similar to properties of highly malignant cells. Furthermore, transformed cells into which Ptn is introduced undergo "switches" to malignant cells of higher malignancy with properties that are strikingly similar to properties of PTN-stimulated cells. These unique features of PTN support the conclusion that constitutive PTN signaling in malignant cells that inappropriately express Ptn functions as a potent tumor promoter. Recently, in confirmation, Ptn targeted by the mouse mammary tumor virus (MMTV) promoter in a transgenic mouse model was found to promote breast cancers to a more aggressive breast cancer cell phenotype that morphologically closely resembles scirrhous carcinoma in human; in addition, it promoted a striking increase in tumor angiogenesis and a remarkable degree of remodeling of the micro-environment. Pleiotrophin thus regulates both different normal and pathological functions; collectively, the different studies have uncovered the unique ability of a single cytokine PTN, which signals through the unique PTN/RPTPbeta/zeta signaling pathway, to induce the many properties associated with tumor promotion in the malignant cells that constitutively express Ptn and in their microenvironment.


Assuntos
Proteínas de Transporte/fisiologia , Citocinas/fisiologia , Fibroblastos/metabolismo , Neoplasias/patologia , Neovascularização Patológica/patologia , Fibroblastos/patologia , Substâncias de Crescimento/fisiologia , Humanos , Proto-Oncogene Mas , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/patologia
14.
J Biol Chem ; 282(39): 28683-28690, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17681947

RESUMO

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) first discovered as the constitutively active nucleophosmin-ALK oncoprotein in anaplastic large cell lymphomas (ALCL). Full-length ALK has a critical role in normal development and differentiation. Activated full-length ALK also is found in different malignant cancers. Nevertheless, the ligand to activate ALK remained unknown until recently, when ALK was proposed to be the physiological receptor of the cytokine pleiotrophin (PTN, Ptn). However, earlier studies had demonstrated that receptor protein tyrosine phosphatase (RPTP) beta/zeta is a physiological PTN receptor. We now demonstrate that phosphorylation of ALK in PTN-stimulated cells is mediated through the PTN/RPTPbeta/zeta signaling pathway. ALK is phosphorylated independently of a direct interaction of PTN with ALK. The data thus support a unique model of ALK activation. In cells not stimulated by PTN, RPTPbeta/zeta dephosphorylates ALK at the site(s) in ALK that is undergoing autophosphorylation through autoactivation. In contrast, when RPTPbeta/zeta is inactivated in PTN-stimulated cells, the sites that are autophosphorylated in ALK no longer can be dephosphorylated by RPTPbeta/zeta; thus, autoactivation and tyrosine phosphorylation of ALK rapidly increase. The data indicate that the PTN/RPTPbeta/zeta signaling pathway is a critical regulator of the steady state levels of tyrosine phosphorylation and activation of ALK; the data support the conclusion that ALK phosphorylation and activation in PTN-stimulated cells are increased through a unique "alternative mechanism of RTK activation."


Assuntos
Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Quinase do Linfoma Anaplásico , Proteínas de Transporte/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Citocinas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células HeLa , Humanos , Ligantes , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Receptores Proteína Tirosina Quinases , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Transdução de Sinais/efeitos dos fármacos
15.
Blood ; 110(1): 287-95, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17369488

RESUMO

Pleiotrophin (PTN) is an important developmental cytokine that is highly expressed during embryogenesis but shows very limited expression in adult tissues, where it is largely restricted to the brain. High PTN serum levels are associated with a variety of solid tumors. We recently showed that patients with multiple myeloma (MM) also have elevated serum levels of this protein and the amount of PTN correlated with the patients' disease status and response to treatment. In this study, we demonstrate that MM cell lines and the malignant cells from MM patients' bone marrow produced PTN and secreted PTN protein into the supernatants during short-term culture. Moreover, Ptn gene expression correlated with the patients' disease status. Inhibition of PTN with a polyclonal anti-PTN antibody reduced growth and enhanced apoptosis of MM cell lines and freshly isolated bone marrow tumor cells from MM patients in vitro. Importantly, this antibody also markedly suppressed the growth of MM in vivo using a severe combined immunodeficiency (SCID)-hu murine model. This represents the first study showing the importance of PTN in the growth of any hematological disorder. Because the expression of this protein is very limited in normal adult tissues, PTN may represent a new target for the treatment of MM.


Assuntos
Proteínas de Transporte/sangue , Citocinas/sangue , Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/imunologia , Proliferação de Células/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/etiologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/imunologia , Índice de Gravidade de Doença , Distribuição Tecidual , Células Tumorais Cultivadas
16.
Biochem Biophys Res Commun ; 351(2): 336-9, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17067552

RESUMO

Pleiotrophin (PTN, Ptn) is an 18kDa secretory cytokine that is expressed in many human cancers, including glioblastoma. In previous experiments, interruption of the constitutive PTN signaling in human U87MG glioblastoma cells that inappropriately express endogenous Ptn reversed their rapid growth in vitro and their malignant phenotype in vivo. To seek a mechanism for the effect of the dominant-negative PTN, flow cytometry was used to compare the profiles of U87MG cells and four clones of U87MG cells that express the dominant-negative PTN (U87MG/PTN1-40 cells); here, we report that the dominant-negative PTN in U87MG cells induces tetraploidy and aneuploidy and arrests the tetraploid and aneuploid cells in the G1 phase of the cell cycle. The data suggest that PTN signaling may have a critical role in chromosomal segregation and cell cycle progression; the data suggest induction of tetraploidy and aneuploidy in U87MG glioblastoma cells may be an important mechanism that contributes to the loss of the malignant phenotype of U87MG cells.


Assuntos
Aneuploidia , Proteínas de Transporte/fisiologia , Ciclo Celular/fisiologia , Citocinas/fisiologia , Poliploidia , Linhagem Celular Tumoral , Clonagem Molecular , Citometria de Fluxo , Fase G1/fisiologia , Glioblastoma , Humanos , Mutação
17.
Proc Natl Acad Sci U S A ; 103(24): 9063-8, 2006 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-16754886

RESUMO

The status of the 66-kDa human estrogen receptor-alpha (hER-alpha66) is a critical determinant in the assessment of the prognosis and in the design of treatment strategies of human breast cancer. Recently, we cloned the cDNA of an alternatively spliced variant of hER-alpha66, termed hER-alpha36; the predicted protein lacks both transcriptional activation domains of hER-alpha66 but retains its DNA-binding domain, partial dimerization, and ligand-binding domains and three potential myristoylation sites located near the N terminus. These findings thus predict that hER-alpha36 functions very differently from hER-alpha66 in response to estrogen signaling. We now demonstrate that hER-alpha36 inhibits the estrogen-dependent and estrogen-independent transactivation activities of hER-alpha66 and hER-beta. We further demonstrate that hER-alpha36 is predominantly associated with the plasma membrane where it transduces both estrogen- and antiestrogen-dependent activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathway and stimulates cell growth. We conclude that hER-alpha36 is a predominantly membrane-based, unique alternatively spliced variant of hER-alpha66 that acts as a dominant-negative effector of both estrogen-dependent and estrogen-independent transactivation functions signaled through hER-alpha66 and ER-beta; it also transduces membrane-initiated estrogen-dependent activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase mitogenic signaling pathway. The estrogen and antiestrogen signaling pathways mediated by hER-alpha36 provide an alternative explanation for why some human breast cancers are resistant to and others are worsened by antiestrogen therapy; the data suggest that hER-alpha36 also may be an important marker to direct therapy in human breast cancers, and perhaps hER-alpha36 also may transduce estrogen-dependent signaling in other estrogen target tissues.


Assuntos
Processamento Alternativo , Membrana Celular/metabolismo , Moduladores de Receptor Estrogênico/metabolismo , Receptor alfa de Estrogênio/metabolismo , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Neoplasias da Mama/metabolismo , Linhagem Celular , Proliferação de Células , Estradiol/química , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Isoformas de Proteínas/genética
18.
Biochem Biophys Res Commun ; 343(2): 653-8, 2006 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-16554021

RESUMO

Neoplasms progress through genetic and epigenetic mutations that deregulate pathways in the malignant cell that stimulate more aggressive growth of the malignant cell itself and/or remodel the tumor microenvironment to support the developing tumor mass. The appearance of new blood vessels in malignant tumors is known as the "angiogenic switch." The angiogenic switch triggers a stage of rapid tumor growth supported by extensive tumor angiogenesis and a more aggressive tumor phenotype and its onset is a poor prognostic indicator for host survival. Identification of the factors that stimulate the angiogenic switch thus is of high importance. Pleiotrophin (PTN the protein, Ptn the gene) is an angiogenic factor and the Ptn gene has been found to be constitutively expressed in many human tumors of different cell types. These studies use a nude mouse model to test if Ptn constitutively expressed in premalignant cells is sufficient to trigger an angiogenic switch in vivo. We introduced an ectopic Ptn gene into "premalignant" SW-13 cells and analyzed the phenotype of SW-13 Ptn cell tumor implants in the flanks of nude mice. SW-13 Ptn cell subcutaneous tumor implants grew very rapidly and had a striking increase in the density of new blood vessels compared to the SW-13 cell tumor implants, suggesting that constitutive PTN signaling in the premalignant SW-13 cell implants in the nude mouse recapitulates fully the angiogenic switch. It was found also that ectopic expression of the C-terminal domain of PTN in SW-13 cell implants was equally effective in initiating an angiogenic switch as the full-length PTN whereas implants of SW-13 cells in nude mice that express the N-terminal domain of PTN grew rapidly but failed to develop tumor angiogenesis. The data suggest the possibility that mutations that activate Ptn in premalignant cells are sufficient to stimulate an angiogenic switch in vivo and, since these mutations are frequently found in human malignancies, that constitutive PTN signaling may be an important contributor to progression of human tumors. The data also suggest that the C-terminal and the N-terminal domains of PTN equally initiate switches in premalignant cells to cells of a more aggressive tumor phenotype but the separate domains of PTN signal different mechanisms and perhaps signal through activation of a separate receptor-like protein.


Assuntos
Neoplasias das Glândulas Suprarrenais/irrigação sanguínea , Neoplasias das Glândulas Suprarrenais/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Citocinas/química , Citocinas/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Citocinas/genética , Feminino , Camundongos , Camundongos Nus , Estrutura Terciária de Proteína , Transdução de Sinais , Relação Estrutura-Atividade
19.
Biochem Biophys Res Commun ; 335(1): 232-9, 2005 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-16105548

RESUMO

Pleiotrophin (PTN the protein, Ptn the gene) signals through a unique mechanism; it inactivates the tyrosine phosphatase activity of its receptor, the transmembrane receptor protein tyrosine phosphatase (RPTP)beta/zeta, and increases tyrosine phosphorylation of the substrates of RPTPbeta/zeta through the continued activity of a yet to be described protein tyrosine kinase(s) in PTN-stimulated cells. We have now found that the cytoskeletal protein beta-adducin interacts with the intracellular domain of RPTPbeta/zeta in a yeast two-hybrid system, that beta-adducin is a substrate of RPTPbeta/zeta, that beta-adducin is phosphorylated in tyrosine in cells not stimulated by PTN, and that tyrosine phosphorylation of beta-adducin is sharply increased in PTN-stimulated cells, suggesting that beta-adducin is a downstream target of and regulated by the PTN/RPTPbeta/zeta signaling pathway. beta-Catenin was the first downstream target of the PTN/RPTPbeta/zeta signaling pathway to be identified; these data thus also suggest that PTN coordinately regulates steady state levels of tyrosine phosphorylation of the important cytoskeletal proteins beta-adducin and beta-catenin and, through PTN-stimulated tyrosine phosphorylation, beta-adducin may contribute to the disruption of cytoskeletal structure, increased plasticity, and loss of homophilic cell-cell adhesion that are the consequences of PTN stimulation of cells and a characteristic feature of different malignant cells with mutations that activate constitutive expression of the endogenous Ptn gene.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Proteínas de Transporte/farmacologia , Citocinas/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ligação a Calmodulina/química , Proteínas de Ligação a Calmodulina/genética , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Especificidade por Substrato , Técnicas do Sistema de Duplo-Híbrido
20.
Proc Natl Acad Sci U S A ; 102(35): 12407-12, 2005 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-16116087

RESUMO

Pleiotrophin (PTN) was found to regulate tyrosine phosphorylation of beta-adducin through the PTN/receptor protein tyrosine phosphatase (RPTP)beta/zeta signaling pathway. We now demonstrate that PTN stimulates the phosphorylation of serines 713 and 726 in the myristoylated alanine-rich protein kinase (PK) C substrate domain of beta-adducin through activation of either PKC alpha or beta. We also demonstrate that PTN stimulates translocation of phosphoserine 713 and 726 beta-adducin either to nuclei, where it associates with nuclear chromatin and with centrioles of dividing cells, or to a membrane-associated site, depending on the phase of cell growth. Furthermore, we demonstrate that PTN stimulates the degradation of beta-adducin in PTN-stimulated cells. Phosphorylation of serines 713 and 726 in beta-adducin is known to markedly reduce the affinity of beta-adducin for spectrin and actin and to uncouple actin/spectrin/beta-adducin multimeric complexes needed for cytoskeletal stability. The data thus suggest that the PTN-stimulated phosphorylation of serines 713 and 726 in beta-adducin disrupts cytoskeletal protein complexes and integrity, features demonstrated in both PTN-stimulated cells and of highly malignant cells that constitutively express the endogenous Ptn gene. The data also support the important conclusion that PTN determines the cellular location of beta-adducin phosphorylated in serines 713 and 726 and raise the possibility that beta-adducin functions in support of structure of heterochromatin and centrioles during mitosis.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Proteínas de Transporte/farmacologia , Citocinas/farmacologia , Proteína Quinase C/metabolismo , Serina/metabolismo , Transporte Biológico Ativo/efeitos dos fármacos , Proteínas de Ligação a Calmodulina/química , Proteínas de Transporte/metabolismo , Citocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Microscopia Confocal , Modelos Biológicos , Fosforilação , Serina/química , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa