Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Cancer ; 124(2): 494-505, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33028955

RESUMO

BACKGROUND: Glutamine (Gln) is an abundant nutrient used by cancer cells. Breast cancers cells and particularly triple-receptor negative breast cancer (TNBC) are reported to be dependent on Gln to produce the energy required for survival and proliferation. Despite intense research on the role of the intracellular Gln pathway, few reports have focussed on Gln transporters in breast cancer and TNBC. METHODS: The role and localisation of the Gln transporter SLC38A2/SNAT2 in response to Gln deprivation or pharmacological stresses was examined in a panel of breast cancer cell lines. Subsequently, the effect of SLC38A2 knockdown in Gln-sensitive cell lines was analysed. The prognostic value of SLC38A2 in a cohort of breast cancer was determined by immunohistochemistry. RESULTS: SLC38A2 was identified as a strongly expressed amino acid transporter in six breast cancer cell lines. We confirmed an autophagic route of degradation for SLC38A2. SLC38A2 knockdown decreased Gln consumption, inhibited cell growth, induced autophagy and led to ROS production in a subgroup of Gln-sensitive cell lines. High expression of SLC38A2 protein was associated with poor breast cancer specific survival in a large cohort of patients (p = 0.004), particularly in TNBC (p = 0.02). CONCLUSIONS: These results position SLC38A2 as a selective target for inhibiting growth of Gln-dependent breast cancer cell lines.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Glutamina/metabolismo , Estresse Oxidativo/fisiologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Adulto , Idoso , Linhagem Celular Tumoral , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico
2.
Proc Natl Acad Sci U S A ; 116(25): 12452-12461, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31152137

RESUMO

Tumor hypoxia is associated with poor patient outcomes in estrogen receptor-α-positive (ERα+) breast cancer. Hypoxia is known to affect tumor growth by reprogramming metabolism and regulating amino acid (AA) uptake. Here, we show that the glutamine transporter, SNAT2, is the AA transporter most frequently induced by hypoxia in breast cancer, and is regulated by hypoxia both in vitro and in vivo in xenografts. SNAT2 induction in MCF7 cells was also regulated by ERα, but it became predominantly a hypoxia-inducible factor 1α (HIF-1α)-dependent gene under hypoxia. Relevant to this, binding sites for both HIF-1α and ERα overlap in SNAT2's cis-regulatory elements. In addition, the down-regulation of SNAT2 by the ER antagonist fulvestrant was reverted in hypoxia. Overexpression of SNAT2 in vitro to recapitulate the levels induced by hypoxia caused enhanced growth, particularly after ERα inhibition, in hypoxia, or when glutamine levels were low. SNAT2 up-regulation in vivo caused complete resistance to antiestrogen and, partially, anti-VEGF therapies. Finally, high SNAT2 expression levels correlated with hypoxia profiles and worse outcome in patients given antiestrogen therapies. Our findings show a switch in the regulation of SNAT2 between ERα and HIF-1α, leading to endocrine resistance in hypoxia. Development of drugs targeting SNAT2 may be of value for a subset of hormone-resistant breast cancer.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/patologia , Hipóxia Celular , Resistencia a Medicamentos Antineoplásicos , Moduladores de Receptor Estrogênico/uso terapêutico , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/metabolismo , Feminino , Xenoenxertos , Humanos , Camundongos , Microambiente Tumoral
3.
Mol Cancer Res ; 17(7): 1531-1544, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30885992

RESUMO

Hypoxia-inducible factor 1α is a key regulator of the hypoxia response in normal and cancer tissues. It is well recognized to regulate glycolysis and is a target for therapy. However, how tumor cells adapt to grow in the absence of HIF1α is poorly understood and an important concept to understand for developing targeted therapies is the flexibility of the metabolic response to hypoxia via alternative pathways. We analyzed pathways that allow cells to survive hypoxic stress in the absence of HIF1α, using the HCT116 colon cancer cell line with deleted HIF1α versus control. Spheroids were used to provide a 3D model of metabolic gradients. We conducted a metabolomic, transcriptomic, and proteomic analysis and integrated the results. These showed surprisingly that in three-dimensional growth, a key regulatory step of glycolysis is Aldolase A rather than phosphofructokinase. Furthermore, glucose uptake could be maintained in hypoxia through upregulation of GLUT14, not previously recognized in this role. Finally, there was a marked adaptation and change of phosphocreatine energy pathways, which made the cells susceptible to inhibition of creatine metabolism in hypoxic conditions. Overall, our studies show a complex adaptation to hypoxia that can bypass HIF1α, but it is targetable and it provides new insight into the key metabolic pathways involved in cancer growth. IMPLICATIONS: Under hypoxia and HIF1 blockade, cancer cells adapt their energy metabolism via upregulation of the GLUT14 glucose transporter and creatine metabolism providing new avenues for drug targeting.


Assuntos
Neoplasias do Colo/genética , Metabolismo Energético/genética , Proteínas Facilitadoras de Transporte de Glucose/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias do Colo/patologia , Creatina/genética , Creatina/metabolismo , Frutose-Bifosfato Aldolase/genética , Glucose/metabolismo , Glicólise/genética , Células HCT116 , Humanos , Esferoides Celulares/metabolismo , Hipóxia Tumoral/genética
4.
Mol Cancer Ther ; 18(2): 376-388, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30478149

RESUMO

Tumor cells exhibit altered lipid metabolism compared with normal cells. Cell signaling kinases are important for regulating lipid synthesis and energy storage. How upstream kinases regulate lipid content, versus direct targeting of lipid-metabolizing enzymes, is currently unexplored. We evaluated intracellular lipid concentrations in prostate and breast tumor spheroids, treated with drugs directly inhibiting metabolic enzymes fatty acid synthase (FASN), acetyl-CoA carboxylase (ACC), diacylglyceride acyltransferase (DGAT), and pyruvate dehydrogenase kinase (PDHK), or cell signaling kinase enzymes PI3K, AKT, and mTOR with lipidomic analysis. We assessed whether baseline lipid profiles corresponded to inhibitors' effectiveness in modulating lipid profiles in three-dimensional (3D) growth and their relationship to therapeutic activity. Inhibitors against PI3K, AKT, and mTOR significantly inhibited MDA-MB-468 and PC3 cell growth in two-dimensional (2D) and 3D spheroid growth, while moderately altering lipid content. Conversely, metabolism inhibitors against FASN and DGAT altered lipid content most effectively, while only moderately inhibiting growth compared with kinase inhibitors. The FASN and ACC inhibitors' effectiveness in MDA-MB-468, versus PC3, suggested the former depended more on synthesis, whereas the latter may salvage lipids. Although baseline lipid profiles did not predict growth effects, lipid changes on therapy matched the growth effects of FASN and DGAT inhibitors. Several phospholipids, including phosphatidylcholine, were also upregulated following treatment, possibly via the Kennedy pathway. As this promotes tumor growth, combination studies should include drugs targeting it. Two-dimensional drug screening may miss important metabolism inhibitors or underestimate their potency. Clinical studies should consider serial measurements of tumor lipids to prove target modulation. Pretherapy tumor classification by de novo lipid synthesis versus uptake may help demonstrate efficacy.


Assuntos
Neoplasias da Mama/metabolismo , Técnicas de Cultura de Células/métodos , Inibidores Enzimáticos/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Acetil-CoA Carboxilase/antagonistas & inibidores , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Ácido Graxo Sintase Tipo I/antagonistas & inibidores , Feminino , Humanos , Masculino , Fosfolipídeos/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
5.
Sci Rep ; 8(1): 8539, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29867102

RESUMO

Ischaemia and reperfusion injury (IRI) is the leading cause of acute kidney injury (AKI), which contributes to high morbidity and mortality rates in a wide range of injuries as well as the development of chronic kidney disease. The cellular and molecular responses of the kidney to IRI are complex and not fully understood. Here, we used an integrated proteomic and metabolomic approach to investigate the effects of IRI on protein abundance and metabolite levels. Rat kidneys were subjected to 45 min of warm ischaemia followed by 4 h and 24 h reperfusion, with contralateral and separate healthy kidneys serving as controls. Kidney tissue proteomics after IRI revealed elevated proteins belonging to the acute phase response, coagulation and complement pathways, and fatty acid (FA) signalling. Metabolic changes were already evident after 4 h reperfusion and showed increased level of glycolysis, lipids and FAs, whilst mitochondrial function and ATP production was impaired after 24 h. This deficit was partially compensated for by the contralateral kidney. Such a metabolic balance counteracts for the developing energy deficit due to reduced mitochondrial function in the injured kidney.


Assuntos
Nefropatias/metabolismo , Rim/metabolismo , Metabolômica , Proteômica , Traumatismo por Reperfusão/metabolismo , Animais , Ácidos Graxos/metabolismo , Glicólise , Rim/patologia , Nefropatias/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteoma/metabolismo , Ratos , Ratos Endogâmicos F344 , Traumatismo por Reperfusão/patologia , Transdução de Sinais
6.
Proc Natl Acad Sci U S A ; 115(24): 6225-6230, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29844165

RESUMO

Cancer cells are reprogrammed to consume large amounts of glucose to support anabolic biosynthetic pathways. However, blood perfusion and consequently the supply with glucose are frequently inadequate in solid cancers. PEPCK-M (PCK2), the mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK), has been shown by us and others to be functionally expressed and to mediate gluconeogenesis, the reverse pathway of glycolysis, in different cancer cells. Serine and ribose synthesis have been identified as downstream pathways fed by PEPCK in cancer cells. Here, we report that PEPCK-M-dependent glycerol phosphate formation from noncarbohydrate precursors (glyceroneogenesis) occurs in starved lung cancer cells and supports de novo glycerophospholipid synthesis. Using stable isotope-labeled glutamine and lactate, we show that PEPCK-M generates phosphoenolpyruvate and 3-phosphoglycerate, which are at least partially converted to glycerol phosphate and incorporated into glycerophospholipids (GPL) under glucose and serum starvation. This pathway is required to maintain levels of GPL, especially phosphatidylethanolamine (PE), as shown by stable shRNA-mediated silencing of PEPCK-M in H23 lung cancer cells. PEPCK-M shRNA led to reduced colony formation after starvation, and the effect was partially reversed by the addition of dioleyl-PE. Furthermore, PEPCK-M silencing abrogated cancer growth in a lung cancer cell xenograft model. In conclusion, glycerol phosphate formation for de novo GPL synthesis via glyceroneogenesis is a newly characterized anabolic pathway in cancer cells mediated by PEPCK-M under conditions of severe nutrient deprivation.


Assuntos
Glicerol/metabolismo , Neoplasias/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Fosfolipídeos/metabolismo , Células A549 , Animais , Glucose/metabolismo , Glutamina/metabolismo , Xenoenxertos , Humanos , Ácido Láctico/metabolismo , Masculino , Camundongos , Camundongos Nus , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , Fosfolipídeos/química
7.
J Biol Chem ; 293(11): 3913-3924, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29279329

RESUMO

Expression of the cell-surface glycoprotein MHC class I polypeptide-related sequence A (MICA) is induced in dangerous, abnormal, or "stressed" cells, including cancer cells, virus-infected cells, and rapidly proliferating cells. MICA is recognized by the activating immune cell receptor natural killer group 2D (NKG2D), providing a mechanism by which immune cells can identify and potentially eliminate pathological cells. Immune recognition through NKG2D is implicated in cancer, atherosclerosis, transplant rejection, and inflammatory diseases, such as rheumatoid arthritis. Despite the wide range of potential therapeutic applications of MICA manipulation, the factors that control MICA expression are unclear. Here we use metabolic interventions and metabolomic analyses to show that the transition from quiescent cellular metabolism to a "Warburg" or biosynthetic metabolic state induces MICA expression. Specifically, we show that glucose transport into the cell and active glycolytic metabolism are necessary to up-regulate MICA expression. Active purine synthesis is necessary to support this effect of glucose, and increases in purine nucleotide levels are sufficient to induce MICA expression. Metabolic induction of MICA expression directly influences NKG2D-dependent cytotoxicity by immune cells. These findings support a model of MICA regulation whereby the purine metabolic activity of individual cells is reflected by cell-surface MICA expression and is the subject of surveillance by NKG2D receptor-expressing immune cells.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Metaboloma/efeitos dos fármacos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Nucleotídeos de Purina/farmacologia , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Ligantes , Células MCF-7 , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética
8.
Clin Proteomics ; 14: 22, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28642677

RESUMO

BACKGROUND: Atherosclerotic plaque rupture is the culprit event which underpins most acute vascular syndromes such as acute myocardial infarction. Novel biomarkers of plaque rupture could improve biological understanding and clinical management of patients presenting with possible acute vascular syndromes but such biomarker(s) remain elusive. Investigation of biomarkers in the context of de novo plaque rupture in humans is confounded by the inability to attribute the plaque rupture as the source of biomarker release, as plaque ruptures are typically associated with prompt down-stream events of myocardial necrosis and systemic inflammation. METHODS: We developed a novel approach to identify potential biomarkers of plaque rupture by integrating plaque imaging, using optical coherence tomography, with both plaque and plasma proteomic analysis in a human model of angioplasty-induced plaque disruption. RESULTS: We compared two pairs of coronary plaque debris, captured by a FilterWire Device, and their corresponding control samples and found matrix metalloproteinase 9 (MMP9) to be significantly enriched in plaque. Plaque contents, as defined by optical coherence tomography, affect the systemic changes of MMP9. Disruption of lipid-rich plaque led to prompt elevation of plasma MMP9, whereas disruption of non-lipid-rich plaque resulted in delayed elevation of plasma MMP9. Systemic MMP9 elevation is independent of the associated myocardial necrosis and systemic inflammation (measured by Troponin I and C-reactive protein, respectively). This information guided the selection of a subset of subjects of for further label free proteomics analysis by liquid chromatography tandem mass spectrometry (LC-MS/MS). We discovered five novel, plaque-enriched proteins (lipopolysaccharide binding protein, Annexin A5, eukaryotic translocation initiation factor, syntaxin 11, cytochrome B5 reductase 3) to be significantly elevated in systemic circulation at 5 min after plaque disruption. CONCLUSION: This novel approach for biomarker discovery in human coronary artery plaque disruption can identify new biomarkers related to human coronary artery plaque composition and disruption.

10.
Oncotarget ; 6(4): 1920-41, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25605240

RESUMO

The biochemistry of cancer cells diverges significantly from normal cells as a result of a comprehensive reprogramming of metabolic pathways. A major factor influencing cancer metabolism is hypoxia, which is mediated by HIF1α and HIF2α. HIF1α represents one of the principal regulators of metabolism and energetic balance in cancer cells through its regulation of glycolysis, glycogen synthesis, Krebs cycle and the pentose phosphate shunt. However, less is known about the role of HIF1α in modulating lipid metabolism. Lipids serve cancer cells to provide molecules acting as oncogenic signals, energetic reserve, precursors for new membrane synthesis and to balance redox biological reactions. To study the role of HIF1α in these processes, we used HCT116 colorectal cancer cells expressing endogenous HIF1α and cells in which the hif1α gene was deleted to characterize HIF1α-dependent and independent effects on hypoxia regulated lipid metabolites. Untargeted metabolomics integrated with proteomics revealed that hypoxia induced many changes in lipids metabolites. Enzymatic steps in fatty acid synthesis and the Kennedy pathway were modified in a HIF1α-dependent fashion. Palmitate, stearate, PLD3 and PAFC16 were regulated in a HIF-independent manner. Our results demonstrate the impact of hypoxia on lipid metabolites, of which a distinct subset is regulated by HIF1α.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metabolismo dos Lipídeos , Lipídeos/biossíntese , Transdução de Sinais , Acetil-CoA C-Aciltransferase/genética , Acetil-CoA C-Aciltransferase/metabolismo , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Idoso , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Hipóxia Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ácidos Graxos/biossíntese , Feminino , Genômica/métodos , Células HCT116 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Metabolômica/métodos , Pessoa de Meia-Idade , Fator de Ativação de Plaquetas/genética , Fator de Ativação de Plaquetas/metabolismo , Proteômica/métodos , Interferência de RNA
11.
Antioxid Redox Signal ; 19(12): 1321-36, 2013 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-23350603

RESUMO

AIMS: To define the consequences of loss of cysteine dioxygenase (CDO) on cysteine metabolism at the tissue level, we determined levels of relevant metabolites and enzymes and evidence of H2S/HS(-) (gaseous hydrogen sulfide and its conjugate base) toxicity in liver, pancreas, kidney, and lung of CDO(-/-) mice that were fed either a taurine-free or taurine-supplemented diet. RESULTS: CDO(-/-) mice had low tissue and serum taurine and hypotaurine levels and high tissue levels of cysteine, consistent with the loss of CDO. CDO(-/-) mice had elevated urinary excretion of thiosulfate, high tissue and serum cystathionine and lanthionine levels, and evidence of inhibition and destabilization of cytochrome c oxidase, which is consistent with excess production of H2S/HS(-). Accumulation of cystathionine and lanthionine appeared to result from cystathionine ß-synthase (CBS)-mediated cysteine desulfhydration. Very high levels of hypotaurine in pancreas of wild-type mice and very high levels of cystathionine and lanthionine in pancreas of CDO(-/-) mice were observed, suggesting a unique cysteine metabolism in the pancreas. INNOVATION: The CDO(-/-) mouse model provides new insights into tissue-specific cysteine metabolism, particularly the role of pancreas in metabolism of excess cysteine by CBS-catalyzed reactions, and will be a useful model for studying the effects of excess endogenous production of H2S/HS(-). CONCLUSION: The CDO(-/-) mouse clearly demonstrates that H2S/HS(-) production in tissues can exceed the capacity of the animal to oxidize sulfide to sulfate and demonstrates that pancreas and lung are more susceptible to toxicity from endogenous H2S/HS(-)production than are liver and kidney.


Assuntos
Cisteína Dioxigenase/genética , Cisteína/metabolismo , Sulfeto de Hidrogênio/metabolismo , Pulmão/metabolismo , Pâncreas/metabolismo , Alanina/análogos & derivados , Alanina/metabolismo , Animais , Cistationina/metabolismo , Cisteína Dioxigenase/deficiência , Dieta , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Estabilidade Enzimática , Feminino , Glutationa/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Pâncreas/patologia , Sulfetos/metabolismo , Taurina/análogos & derivados , Taurina/metabolismo
12.
Am J Physiol Endocrinol Metab ; 301(4): E668-84, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21693692

RESUMO

Cysteine homeostasis is dependent on the regulation of cysteine dioxygenase (CDO) in response to changes in sulfur amino acid intake. CDO oxidizes cysteine to cysteinesulfinate, which is further metabolized to either taurine or to pyruvate plus sulfate. To gain insight into the physiological function of CDO and the consequence of a loss of CDO activity, mice carrying a null CDO allele (CDO(+/-) mice) were crossed to generate CDO(-/-), CDO(+/-), and CDO(+/+) mice. CDO(-/-) mice exhibited postnatal mortality, growth deficit, and connective tissue pathology. CDO(-/-) mice had extremely low taurine levels and somewhat elevated cysteine levels, consistent with the lack of flux through CDO-dependent catabolic pathways. However, plasma sulfate levels were slightly higher in CDO(-/-) mice than in CDO(+/-) or CDO(+/+) mice, and tissue levels of acid-labile sulfide were elevated, indicating an increase in cysteine catabolism by cysteine desulfhydration pathways. Null mice had lower hepatic cytochrome c oxidase levels, suggesting impaired electron transport capacity. Supplementation of mice with taurine improved survival of male pups but otherwise had little effect on the phenotype of the CDO(-/-) mice. H(2)S has been identified as an important gaseous signaling molecule as well as a toxicant, and pathology may be due to dysregulation of H(2)S production. Control of cysteine levels by regulation of CDO may be necessary to maintain low H(2)S/sulfane sulfur levels and facilitate the use of H(2)S as a signaling molecule.


Assuntos
Cisteína Dioxigenase/fisiologia , Cisteína/metabolismo , Sulfeto de Hidrogênio/metabolismo , Taurina/biossíntese , Animais , Cisteína Dioxigenase/genética , Transporte de Elétrons/fisiologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout
13.
Reprod Biomed Online ; 19(6): 838-46, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20031026

RESUMO

Although dialysis and transplantation are widely applied therapies for renal failure, drawbacks such as morbidity, shortage of compatible organs and high cost are limiting factors. Recently, interest has increased in the potential use of stem cells for the repair of kidney injury, which has been considered as an alternative therapeutic strategy. Due to their high proliferation rates, their pluripotent differentiation potential, the finding that they do not induce tumour formation and the fact that they do not raise the ethical concerns connected with human embryonic stem cells, human amniotic fluid stem cells are considered to be a very promising cell source. This study demonstrates that the expression of the mesenchymal markers CD29 and CD44, the epithelial markers CD51 and ZO-1 and the podocyte markers CD2AP and NPHS2 can be induced in these cells via incubation with epidermal growth factor/platelet-derived growth factor BB and fibroblast growth factor 4/hepatocyte growth factor, respectively. Since podocytes are visceral epithelial cells in the kidneys, which form the essential part of the glomerular filtration barrier, these findings warrant further investigation of the potential use of human amniotic fluid stem cells for cell-based kidney therapies.


Assuntos
Biomarcadores/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Líquido Amniótico/citologia , Becaplermina , Diferenciação Celular , Proteínas do Citoesqueleto/biossíntese , Células-Tronco Embrionárias/efeitos dos fármacos , Fator 4 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/biossíntese , Integrina alfaV/biossíntese , Integrina beta1/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Rim/citologia , Proteínas de Membrana/biossíntese , Fator 3 de Transcrição de Octâmero/biossíntese , Fosfoproteínas/biossíntese , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Proteína da Zônula de Oclusão-1
14.
Hum Mol Genet ; 18(17): 3298-310, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19505958

RESUMO

Dysregulation of the mammalian target of rapamycin (mTOR) kinase pathway is centrally involved in a wide variety of cancers and human genetic diseases. In mammalian cells, mTOR is part of two different kinase complexes: mTORC1 composed of mTOR, raptor and mLST8, and mTORC2 containing mTOR, rictor, sin1 and mLST8. Whereas, mTORC1 is known to be a pivotal regulator of cell size and cell cycle control, the question whether the recently discovered mTORC2 complex is involved in these processes remains elusive. We report here that the mTORC1-mediated consequences on cell cycle and cell size are separable and do not involve effects on mTORC2 activity. However, we show that mTORC2 itself is a potent regulator of mammalian cell size and cell cycle via a mechanism involving the Akt/TSC2/Rheb cascade. Our data are of relevance for the understanding of the molecular development of the many human diseases caused by deregulation of upstream and downstream effectors of mTOR.


Assuntos
Ciclo Celular , Tamanho Celular , Complexos Multiproteicos/metabolismo , Proteínas Quinases/metabolismo , Linhagem Celular , Humanos , Complexos Multiproteicos/genética , Proteínas Quinases/genética , Serina-Treonina Quinases TOR
15.
Int J Mol Med ; 23(6): 779-84, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19424604

RESUMO

The discovery of human amniotic fluid stem cells initiated a new and promising stem cell research field. These cells harbor a high proliferative capacity and the potential to differentiate into cells of all three embryonic germ layers. The facts that they do not form tumors in vivo and do not raise the ethical concerns associated with human embryonic stem cells support their role as an optimal tool to study the underlying molecular mechanisms of cell differentiation processes and of their deregulation in human genetic diseases. Deregulation of the protein kinase mammalian target of rapamycin (mTOR) pathway is a hallmark of a wide variety of human genetic diseases. Here we report the establishment of an amniotic fluid stem cell line. We analysed the endogenous expression of the mTOR pathway proteins tuberin, mTOR, raptor, rictor, sin1, mLST8, Akt and p70S6K in human amniotic fluid stem cells. In addition, we studied the endogenous activity of the kinase p70S6K, one of the major targets of the mTOR complex 1 kinase, by analysing the p70S6K T389 phosphorylation status. The activity of the Akt kinase, the major mTOR complex 2 target, was studied by analysing its phosphorylation at S473. In addition, the mTOR inhibitor rapamycin was found to affect the phosphorylation status of p70S6K in amniotic fluid stem cells. Taken together, we provide evidence that the mTOR pathway is fully active in human amniotic fluid stem cells. These data demonstrate that amniotic fluid stem cell lines can be used as new tools to study the molecular and cell biological consequences of natural occurring alterations of the mTOR pathway being responsible for a wide variety of different human genetic diseases.


Assuntos
Líquido Amniótico/citologia , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Humanos , Immunoblotting , Imunossupressores/farmacologia , Fosforilação , Proteínas/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Proteína Regulatória Associada a mTOR , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/genética , Sirolimo/farmacologia , Células-Tronco/efeitos dos fármacos , Serina-Treonina Quinases TOR , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
16.
Kidney Int ; 76(3): 293-6, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19357721

RESUMO

S-adenosylhomocysteine (SAH), the metabolic precursor of homocysteine in the body, is a potent inhibitor of methylation reactions. Several methylation reactions play a major role in epigenetic regulation of protein expression, atherosclerosis, and cancer development. Here we studied the mechanisms responsible for the maintenance of circulating SAH levels by measurement of the arterio-venous differences across the kidney, splanchnic organs, and the lung in humans. The lungs did not remove or add any circulating SAH, whereas the liver released it into the hepatic veins. The kidney extracted 40% of SAH and the SAH arterio-venous difference across the kidney was directly and significantly related to its arterial levels. Thus, the kidney plays a major role in maintaining SAH levels and may, indirectly, control tissue transmethylation reactions. Our findings of a pivotal role for the human kidney in sulfur amino acid metabolism may also account for the increased plasma levels of SAH in patients with chronic kidney diseases.


Assuntos
Rim/metabolismo , S-Adenosil-Homocisteína/sangue , Idoso , Feminino , Humanos , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , Circulação Esplâncnica
17.
Leuk Res ; 33(7): 883-5, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19286253

RESUMO

The genes TSC1 and TSC2, encoding hamartin and tuberin, respectively, have been shown to be involved in the development of the autosomal dominantly inherited tumor syndrome tuberous sclerosis (TSC). However, inactivation of these genes has also been demonstrated to be associated with sporadic bladder cancer, ovarian and gall bladder carcinoma, non-small-cell carcinoma of the lung, breast cancer, pancreatic cancer, astrocytoma, xanthoastrocytoma, ependymomas, oral squamous cell carcinoma and endometrial cancer. The hamartin/tuberin protein complex plays a central role in the regulation of the mammalian target of rapamycin (mTOR) signalling network. A wide variety of components of the mTOR cascade have been demonstrated to be involved in many different human cancers. Mutations in several mTOR pathway component genes are known to cause specific monogenic human genetic diseases and this signalling cascade has been shown to be of relevance for Alzheimer's disease, type 2 diabetes, obesity and hypertrophy. Consequently, e.g. clinical trials for the treatment with rapamycin, a negative regulator of mTOR, of hamartomas in TSC have already been initiated. Now the first evidence is provided for an involvement of the TSC genes in acute leukemia.


Assuntos
Genes Supressores de Tumor , Leucemia/genética , Proteínas Supressoras de Tumor/genética , Humanos , Leucemia/metabolismo , Leucemia/patologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa
18.
Mutat Res ; 659(3): 284-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18598780

RESUMO

The signalling components upstream and downstream of the protein kinase mammalian target of rapamycin (mTOR) are frequently altered in a wide variety of human diseases. Upstream of mTOR key signalling molecules are the small GTPase Ras, the lipid kinase PI3K, the Akt kinase, and the GTPase Rheb, which are known to be deregulated in many human cancers. Mutations in the mTOR pathway component genes TSC1, TSC2, LKB1, PTEN, VHL, NF1 and PKD1 trigger the development of the syndromes tuberous sclerosis, Peutz-Jeghers syndrome, Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Lhermitte-Duclos disease, Proteus syndrome, von Hippel-Lindau disease, Neurofibromatosis type 1, and Polycystic kidney disease, respectively. In addition, the tuberous sclerosis proteins have been implicated in the development of several sporadic tumors and in the control of the cyclin-dependent kinase inhibitor p27, known to be of relevance for several cancers. Recently, it has been recognized that mTOR is regulated by TNF-alpha and Wnt, both of which have been shown to play critical roles in the development of many human neoplasias. In addition to all these human diseases, the role of mTOR in Alzheimer's disease, cardiac hypertrophy, obesity and type 2 diabetes is discussed.


Assuntos
Doenças Genéticas Inatas/genética , Neoplasias/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Doença de Alzheimer/metabolismo , Diabetes Mellitus Tipo 2/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Quinases/fisiologia , Serina-Treonina Quinases TOR
19.
Kidney Int ; 74(7): 937-45, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18633341

RESUMO

Resistance to the anabolic action of growth hormone may contribute to the loss of strength and muscle mass in adult patients with chronic kidney disease. We tested this hypothesis by infusing growth hormone in patients to levels necessary to saturate hormone receptors. This led to a significant decrease of plasma potassium and amino acid levels in control and hyperkalemic patients with chronic kidney disease. These effects were completely or partially blunted in patients with elevated C-reactive protein levels. In forearm perfusion studies, growth hormone caused a further decrease in the negative potassium and protein balance of hemodialysis patients without inflammation but no effect was seen in patients with inflammation. Only IL-6 levels and age were found to be independent correlates in these growth hormone-induced variations in plasma potassium and blood amino acids. This shows that although a resistance to pharmacologic doses of growth hormone is not a general feature of patients with chronic kidney disease, there is a subgroup characterized by blunted growth hormone action. Our results support the hypothesis that uremia with inflammation, but not uremia per se, inhibits downstream growth hormone signaling contributing to muscle atrophy.


Assuntos
Resistência a Medicamentos , Hormônio do Crescimento Humano/farmacocinética , Inflamação , Nefropatias/patologia , Uremia , Fatores Etários , Idoso , Aminoácidos/sangue , Estudos de Casos e Controles , Doença Crônica , Feminino , Hormônio do Crescimento Humano/administração & dosagem , Humanos , Interleucina-6/sangue , Nefropatias/complicações , Masculino , Pessoa de Meia-Idade , Atrofia Muscular/etiologia , Potássio/sangue
20.
Clin Chim Acta ; 395(1-2): 106-10, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18565329

RESUMO

OBJECTIVE: The putative role of sulfur amino acids such as homocysteine (tHcy) as cardiovascular risk factors is controversial in chronic kidney disease (CKD). Although, S-adenosylhomocysteine (SAH) levels have been linked to CVD in non-renal populations, such relationship has not been evaluated in CKD. DESIGN: Serum concentrations of S-adenosylmethionine (SAM), SAH and total homocysteine (tHcy) were determined by HPLC in 124 CKD stage 5 patients (GFR range 1-11 m/min) and 47 control subjects, and related to renal function, presence of CVD, inflammation and protein-energy wasting (PEW). RESULTS: The levels of SAM and SAH were higher in CKD patients than in controls. Both SAM (rho=-0.19; P<0.05) and SAH (rho=-0.37, P<0.001) were inversely related to GFR. The concentrations of SAH were significantly higher (P<0.001) in patients with CVD than in non-CVD patients, (683 (201-3057) vs 485 (259-2620) nmol/L; median (range)) as opposed to tHcy levels, which were lower in CVD patients. While SAH was not associated with the presence of inflammation or PEW, it was a significant contributor (OR; 4.9 (CI 1.8-12.8), P<0.001) to CVD in a multinomial logistic regression model (pseudo r(2)=0.31). CONCLUSION: Concentrations of serum SAH and SAM in CKD stage 5 patients are associated with renal function, but not with inflammation or PEW. Among the investigated sulfur amino acids, only SAH was independently associated with the presence of clinical signs of CVD. These findings suggest that while tHcy might be influenced by a number of confounding uremic factors, SAH levels may better reflect the putative increased cardiovascular risk of sulfur amino acid alterations in CKD patients.


Assuntos
Doenças Cardiovasculares/sangue , Homocisteína/sangue , Nefropatias/sangue , S-Adenosil-Homocisteína/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Doenças Cardiovasculares/diagnóstico , Cromatografia Líquida de Alta Pressão/métodos , Doença Crônica , Feminino , Humanos , Testes de Função Renal/métodos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , S-Adenosilmetionina/sangue , Sensibilidade e Especificidade , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...