Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Exp Hematol ; 67: 60-64.e2, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30125603

RESUMO

Increased levels of fetal hemoglobin (HbF) lessen the severity of symptoms and increase the life span of patients with sickle cell disease (SCD). More effective strategies to increase HbF are needed because the current standard of care, hydroxyurea, is not effective in a significant proportion of patients. Treatment of the millions of patients projected worldwide would best be accomplished with an orally administered drug therapy that increased HbF. LSD1 is a component of corepressor complexes that repress γ-globin gene expression and are a therapeutic target for HbF reactivation. We have shown that subcutaneous administration of RN-1, a pharmacological LSD1 inhibitor, increased γ-globin expression in SCD mice and baboons, which are widely acknowledged as the best animal model in which to test the activity of HbF-inducing drugs. The objective of this investigation was to test the effect of oral administration of a new LSD1 inhibitor, ORY-3001. Oral administration of ORY-3001 to SCD mice (n = 3 groups) increased γ-globin expression, Fetal Hemoglobin (HbF)-containing (F) cells, and F reticulocytes (retics). In normal baboons (n = 7 experiments) treated with ORY-3001, increased F retics, γ-globin chain synthesis, and γ-globin mRNA were observed. Experiments in anemic baboons (n = 2) showed that ORY-3001 increased F retics (PA8695, predose = 24%, postdose = 66.8%; PA8698: predose = 13%, postdose = 93.6%), γ-globin chain synthesis (PA8695: predose = 0.07 γ/γ+ß, postdose = 0.20 γ/γ+ß; PA8698: predose = 0.02 γ/γ+ß, postdose = 0.44 γ/γ+ß), and γ-globin mRNA (PA8695: predose = 0.06 γ/γ+ß, postdose = 0.18 γ/γ+ß; PA8698: predose = 0.03 γ/γ+ß, postdose = 0.33 γ/γ+ß). We conclude that oral administration of ORY-3001 increases F retics, γ-globin chain synthesis, and γ-globin mRNA in baboons and SCD mice, supporting further efforts toward the development of this drug for SCD therapy.


Assuntos
Anemia Falciforme/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Hemoglobina Fetal/biossíntese , Histona Desmetilases/antagonistas & inibidores , gama-Globinas/biossíntese , Administração Oral , Anemia/sangue , Anemia/tratamento farmacológico , Anemia Falciforme/sangue , Animais , Contagem de Células Sanguíneas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/administração & dosagem , Feminino , Hemoglobina Fetal/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Papio , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reticulócitos/metabolismo , gama-Globinas/genética
2.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R840-R847, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30067082

RESUMO

Sickle cell disease (SCD) is caused by a mutation of the ß-globin gene (Ingram VM. Nature 180: 326-328, 1957), which triggers the polymerization of deoxygenated sickle hemoglobin (HbS). Approximately 100,000 SCD patients in the United States and millions worldwide (Piel FB, et al. PLoS Med 10: e1001484, 2013) suffer from chronic hemolytic anemia, painful crises, multisystem organ damage, and reduced life expectancy (Rees DC, et al. Lancet 376: 2018-2031, 2010; Serjeant GR. Cold Spring Harb Perspect Med 3: a011783, 2013). Hematopoietic stem cell transplantation can be curative, but the majority of patients do not have a suitable donor (Talano JA, Cairo MS. Eur J Haematol 94: 391-399, 2015). Advanced gene-editing technologies also offer the possibility of a cure (Goodman MA, Malik P. Ther Adv Hematol 7: 302-315, 2016; Lettre G, Bauer DE. Lancet 387: 2554-2564, 2016), but the likelihood that these strategies can be mobilized to treat the large numbers of patients residing in developing countries is remote. A pharmacological treatment to increase fetal hemoglobin (HbF) as a therapy for SCD has been a long-sought goal, because increased levels of HbF (α2γ2) inhibit the polymerization of HbS (Poillin WN, et al. Proc Natl Acad Sci USA 90: 5039-5043, 1993; Sunshine HR, et al. J Mol Biol 133: 435-467, 1979) and are associated with reduced symptoms and increased lifespan of SCD patients (Platt OS, et al. N Engl J Med 330: 1639-1644, 1994; Platt OS, et al. N Engl J Med 325: 11-16, 1991). Only two drugs, hydroxyurea and l-glutamine, are approved by the US Food and Drug Administration for treatment of SCD. Hydroxyurea is ineffective at HbF induction in ~50% of patients (Charache S, et al. N Engl J Med 332: 1317-1322, 1995). While polymerization of HbS has been traditionally considered the driving force in the hemolysis of SCD, the excessive reactive oxygen species generated from red blood cells, with further amplification by intravascular hemolysis, also are a major contributor to SCD pathology. This review highlights a new class of drugs, lysine-specific demethylase (LSD1) inhibitors, that induce HbF and reduce reactive oxygen species.


Assuntos
Anemia Falciforme/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Eritrócitos/efeitos dos fármacos , Histona Desmetilases/antagonistas & inibidores , Anemia Falciforme/sangue , Anemia Falciforme/enzimologia , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Desenho de Fármacos , Eritrócitos/enzimologia , Hemoglobina Fetal/metabolismo , Histona Desmetilases/metabolismo , Humanos , Camundongos , Terapia de Alvo Molecular , Papio , Espécies Reativas de Oxigênio/sangue , Medição de Risco
3.
Hematology Am Soc Hematol Educ Program ; 2017(1): 440-446, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29222291

RESUMO

Sickle cell disease (SCD) is an inheritable hemoglobinopathy characterized by polymerization of hemoglobin S in red blood cells resulting in chronic hemolytic anemia, vaso-occlusive painful crisis, and multiorgan damage. In SCD, an increased reactive oxygen species (ROS) generation occurs both inside the red blood cells and inside the vascular lumen, which augment hemolysis and cellular adhesion. This review discusses the evolving body of literature on the role of ROS in the pathophysiology of SCD as well as some emerging therapeutic approaches to SCD with a focus on the reduction of ROS.


Assuntos
Anemia Falciforme/sangue , Anemia Falciforme/terapia , Eritrócitos Anormais/metabolismo , Hemoglobina Falciforme/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Anemia Falciforme/patologia , Adesão Celular , Eritrócitos Anormais/patologia , Hemólise , Humanos
4.
Exp Hematol ; 50: 46-52, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28238805

RESUMO

Sickle cell disease (SCD), an inherited blood disorder caused by a point mutation that renders hemoglobin susceptible to polymerization when deoxygenated, affects millions of people worldwide. Manifestations of SCD include chronic hemolytic anemia, inflammation, painful vaso-occlusive crises, multisystem organ damage, and reduced life expectancy. Part of SCD pathophysiology is the excessive formation of intracellular reactive oxygen species (ROS) in SCD red blood cells (RBCs), which accelerates their hemolysis. Normal RBC precursors eliminate their mitochondria during the terminal differentiation process. Strikingly, we observed an increased percentage of RBCs retaining mitochondria in SCD patient blood samples compared with healthy individuals. In addition, using an experimental SCD mouse model, we demonstrate that excessive levels of ROS in SCD are associated with this abnormal mitochondrial retention. Interestingly, the LSD1 inhibitor, RN-1, and the mitophagy-inducing agent mammalian target of rapamycin (mTOR) inhibitor, sirolimus, increased RBC lifespan and reduced ROS accumulation in parallel with reducing mitochondria-retaining RBCs in the SCD mouse model. Furthermore, gene expression analysis of SCD mice treated with RN-1 showed increased expression of mitophagy genes. Our findings suggest that reduction of mitochondria-retaining RBCs may provide a new therapeutic approach to preventing excessive ROS in SCD.


Assuntos
Anemia Falciforme/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Histona Desmetilases/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Modelos Biológicos , Rodaminas/farmacologia , Sirolimo/farmacologia , Compostos de Espiro/farmacologia , Tiofenos/farmacologia
5.
Haematologica ; 101(6): 688-97, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26858356

RESUMO

Increased fetal hemoglobin levels lessen the severity of symptoms and increase the lifespan of patients with sickle cell disease. Hydroxyurea, the only drug currently approved for the treatment of sickle cell disease, is not effective in a large proportion of patients and therefore new pharmacological agents that increase fetal hemoglobin levels have long been sought. Recent studies identifying LSD-1 as a repressor of γ-globin expression led to experiments demonstrating that the LSD-1 inhibitor RN-1 increased γ-globin expression in the sickle cell mouse model. Because the arrangement and developmental stage-specific expression pattern of the ß-like globin genes is highly conserved between man and baboon, the baboon model remains the best predictor of activity of fetal hemoglobin-inducing agents in man. In this report, we demonstrate that RN-1 increases γ-globin synthesis, fetal hemoglobin, and F cells to high levels in both anemic and non-anemic baboons with activity comparable to decitabine, the most potent fetal hemoglobin-inducing agent known. RN-1 not only restores high levels of fetal hemoglobin but causes the individual 5' Iγ- and 3' Vγ-globin chains to be synthesized in the ratio characteristic of fetal development. Increased fetal hemoglobin was associated with increased levels of acetylated Histone H3, H3K4Me2, H3K4Me3, and RNA polymerase II at the γ-globin gene, and diminished γ-globin promoter DNA methylation. RN-1 is likely to induce clinically relevant levels of fetal hemoglobin in patients with sickle cell disease, although careful titration of the dose may be required to minimize myelotoxicity.


Assuntos
Inibidores Enzimáticos/farmacologia , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Histona Desmetilases/antagonistas & inibidores , Anemia/sangue , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Contagem de Células Sanguíneas , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Papio , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , gama-Globinas/biossíntese , gama-Globinas/genética
6.
PLoS One ; 10(7): e0134003, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26225425

RESUMO

OBJECTIVE: We have shown previously that preterm infants are at risk of necrotizing enterocolitis (NEC), an inflammatory bowel necrosis typically seen in infants born prior to 32 weeks' gestation, because of the developmental deficiency of transforming growth factor (TGF)-ß2 in the intestine. The present study was designed to investigate all-trans retinoic acid (atRA) as an inducer of TGF-ß2 in intestinal epithelial cells (IECs) and to elucidate the involved signaling mechanisms. METHODS: AtRA effects on intestinal epithelium were investigated using IEC6 cells. TGF-ß2 expression was measured using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blots. Signaling pathways were investigated using Western blots, transiently-transfected/transduced cells, kinase arrays, chromatin immunoprecipitation, and selective small molecule inhibitors. RESULTS: AtRA-treatment of IEC6 cells selectively increased TGF-ß2 mRNA and protein expression in a time- and dose-dependent fashion, and increased the activity of the TGF-ß2 promoter. AtRA effects were mediated via RhoA GTPase, Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1), p38α MAPK, and activating transcription factor (ATF)-2. AtRA increased phospho-ATF2 binding to the TGF-ß2 promoter and increased histone H2B acetylation in the TGF-ß2 nucleosome, which is typically associated with transcriptional activation. CONCLUSIONS: AtRA induces TGF-ß2 expression in IECs via RhoA- and p38α MAPK-mediated activation of the transcription factor ATF2. Further studies are needed to investigate the role of atRA as a protective/therapeutic agent in gut mucosal inflammation.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Mucosa Intestinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Tretinoína/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Humanos , Mucosa Intestinal/citologia , Fosforilação , Proteínas Smad/metabolismo
7.
Exp Hematol ; 43(7): 546-53.e1-3, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25931013

RESUMO

Increased levels of fetal hemoglobin are associated with decreased symptoms and increased lifespan in patients with sickle cell disease (SCD). Hydroxyurea, the only drug currently approved for SCD, is not effective in a large fraction of patients, and therefore, new agents are urgently needed. Recently it was found that lysine demethylase 1, an enzyme that removes monomethyl and dimethyl residues from the lysine 4 residue of histone H3, is a repressor of γ-globin gene expression. In this article, we have compared the ability of tranylcypromine (TCP) and a more potent TCP derivative, RN-1, to increase γ-globin expression in cultured baboon erythroid progenitor cells and in the SCD mouse model. The results indicate that the ability of RN-1 to induce F cells and γ-globin mRNA in SCD mice is similar to that of decitabine, the most powerful fetal hemoglobin-inducing drug known, and greater than that of either TCP or hydroxyurea. We conclude that RN-1 and other lysine demethylase 1 inhibitors may be promising new γ-globin-inducing agents for the treatment of SCD that warrant further studies in other preclinical models, such as nonhuman primates.


Assuntos
Anemia Falciforme/genética , Hemoglobina Fetal/biossíntese , Histona Desmetilases/antagonistas & inibidores , Reticulócitos/efeitos dos fármacos , Tranilcipromina/farmacologia , gama-Globinas/biossíntese , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/enzimologia , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Hemoglobina Fetal/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Histonas/metabolismo , Humanos , Hidroxiureia/farmacologia , Metilação , Camundongos , Camundongos Transgênicos , Papio , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transgenes/efeitos dos fármacos , Tranilcipromina/análogos & derivados , Tretinoína/farmacologia , Células U937 , gama-Globinas/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 306(5): G361-9, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24407592

RESUMO

Fetal swallowing of amniotic fluid, which contains numerous cytokines and growth factors, plays a key role in gut mucosal development. Preterm birth interrupts this exposure to amniotic fluid-borne growth factors, possibly contributing to the increased risk of necrotizing enterocolitis (NEC) in premature infants. We hypothesized that supplementation of formula feeds with amniotic fluid can provide amniotic fluid-borne growth factors and prevent experimental NEC in rat pups. We compared NEC-like injury in rat pups fed with infant formula vs. formula supplemented either with 30% amniotic fluid or recombinant hepatocyte growth factor (HGF). Cytokines/growth factors in amniotic fluid were measured by immunoassays. Amniotic fluid and HGF effects on enterocyte migration, proliferation, and survival were measured in cultured IEC6 intestinal epithelial cells. Finally, we used an antibody array to investigate receptor tyrosine kinase (RTK) activation and immunoblots to measure phosphoinositide 3-kinase (PI3K) signaling. Amniotic fluid supplementation in oral feeds protected rat pups against NEC-like injury. HGF was the most abundant growth factor in rat amniotic fluid in our panel of analytes. Amniotic fluid increased cell migration, proliferation, and cell survival in vitro. These effects were reproduced by HGF and blocked by anti-HGF antibody or a PI3K inhibitor. HGF transactivated several RTKs in IEC6 cells, indicating that its effects extended to multiple signaling pathways. Finally, similar to amniotic fluid, recombinant HGF also reduced the frequency and severity of NEC-like injury in rat pups. Amniotic fluid supplementation protects rat pups against experimental NEC, which is mediated, at least in part, by HGF.


Assuntos
Líquido Amniótico/metabolismo , Enterocolite Necrosante/prevenção & controle , Fator de Crescimento de Hepatócito/administração & dosagem , Líquido Amniótico/química , Ração Animal , Animais , Células Cultivadas , Citocinas/metabolismo , Suplementos Nutricionais , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica , Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Lactente , Fórmulas Infantis , Mucosa Intestinal/citologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo
9.
Lab Invest ; 94(2): 150-60, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24365747

RESUMO

Neonates and young infants exposed to extracorporeal circulation during extracorporeal membrane oxygenation (ECMO) and cardiopulmonary bypass are at risk of developing a systemic inflammatory response syndrome with multi-organ dysfunction. We used a piglet model of ECMO to investigate the hypothesis that epithelial apoptosis is an early event that precedes villous damage during ECMO-related bowel injury. Healthy 3-week-old piglets were subjected to ECMO for up to 8 h. Epithelial apoptosis was measured in histopathological analysis, nuclear imaging, and terminal deoxynucleotidyl transferase dUTP nick end labeling. Plasma intestinal fatty acid-binding protein (I-FABP) levels were measured by enzyme immunoassay. Intestinal mast cells were isolated by fluorescence-assisted cell sorting. Cleaved caspase-8, caspase-9, phospho-p38 MAPK, and fas ligand expression were investigated by immunohistochemistry, western blots, and reverse transcriptase-quantitative PCR. Piglet ECMO was associated with increased gut epithelial apoptosis. Extensive apoptotic changes were noted on villus tips and in scattered crypt cells after 2 h of ECMO. After 8 h, the villi were denuded and apoptotic changes were evident in a majority of crypt cells. Increased circulating I-FABP levels, a marker of gut epithelial injury, showed that epithelial injury occurred during ECMO. We detected increased cleaved caspase-8, but not cleaved caspase-9, in epithelial cells indicating that the extrinsic apoptotic pathway was active. ECMO was associated with increased fas ligand expression in intestinal mast cells, which was induced through activation of the p38 mitogen-activated protein kinase. We conclude that epithelial apoptosis is an early event that initiates gut mucosal injury in a piglet model of ECMO.


Assuntos
Apoptose/fisiologia , Oxigenação por Membrana Extracorpórea/efeitos adversos , Mucosa Intestinal/lesões , Mucosa Intestinal/fisiopatologia , Animais , Animais Recém-Nascidos , Western Blotting , Caspase 8/metabolismo , Caspase 9/metabolismo , Núcleo Celular/ultraestrutura , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas/metabolismo , Proteínas de Ligação a Ácido Graxo/sangue , Citometria de Fluxo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Mucosa Intestinal/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Suínos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Am J Physiol Gastrointest Liver Physiol ; 304(12): G1055-65, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23558011

RESUMO

Human milk contains substantial amounts of transforming growth factor (TGF)-ß, particularly the isoform TGF-ß2. We previously showed in preclinical models that enterally administered TGF-ß2 can protect against necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of premature infants. In this study we hypothesized that premature infants remain at higher risk of NEC than full-term infants, even when they receive their own mother's milk, because preterm human milk contains less bioactive TGF-ß than full-term milk. Our objective was to compare TGF-ß bioactivity in preterm vs. full-term milk and identify factors that activate milk-borne TGF-ß. Mothers who delivered between 23 0/7 and 31 6/7 wk or at ≥37 wk of gestation provided milk samples at serial time points. TGF-ß bioactivity and NF-κB signaling were measured using specific reporter cells and in murine intestinal tissue explants. TGF-ß1, TGF-ß2, TGF-ß3, and various TGF-ß activators were measured by real-time PCR, enzyme immunoassays, or established enzymatic activity assays. Preterm human milk showed minimal TGF-ß bioactivity in the native state but contained a large pool of latent TGF-ß. TGF-ß2 was the predominant isoform of TGF-ß in preterm milk. Using a combination of several in vitro and ex vivo models, we show that neuraminidase is a key regulator of TGF-ß bioactivity in human milk. Finally, we show that addition of bacterial neuraminidase to preterm human milk increased TGF-ß bioactivity. Preterm milk contains large quantities of TGF-ß, but most of it is in an inactive state. Addition of neuraminidase can increase TGF-ß bioactivity in preterm milk and enhance its anti-inflammatory effects.


Assuntos
Leite Humano/metabolismo , Neuraminidase/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Expressão Gênica , Humanos , Lactação/metabolismo , Camundongos , Leite Humano/enzimologia , NF-kappa B/genética , NF-kappa B/metabolismo , Neuraminidase/genética , Nascimento Prematuro/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Nascimento a Termo/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/genética
11.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G167-80, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23154975

RESUMO

Preterm infants may be at risk of necrotizing enterocolitis (NEC) due to deficiency of transforming growth factor-ß 2 (TGF-ß(2)) in the developing intestine. We hypothesized that low epithelial TGF-ß(2) expression in preterm intestine and during NEC results from diminished autocrine induction of TGF-ß(2) in these cells. Premature baboons delivered at 67% gestation were treated per current norms for human preterm infants. NEC was diagnosed by clinical and radiological findings. Inflammatory cytokines, TGF-ß(2), Smad7, Ski, and strawberry notch N (SnoN)/Ski-like oncoprotein (SKIL) was measured using quantitative reverse transcriptase-polymerase chain reaction, immunoblots, and immunohistochemistry. Smad7 effects were examined in transfected IEC6 intestinal epithelial cells in vitro. Findings were validated in archived human tissue samples of NEC. NEC was recorded in seven premature baboons. Consistent with existing human data, premature baboon intestine expressed less TGF-ß(2) than term intestine. TGF-ß(2) expression was regulated in epithelial cells in an autocrine fashion, which was interrupted in the premature intestine and during NEC due to increased expression of Smad7. LPS increased Smad7 binding to the TGF-ß(2) promoter and was associated with dimethylation of the lysine H3K9, a marker of transcriptional silencing, on the nucleosome of TGF-ß(2). Increased Smad7 expression in preterm intestine was correlated with the deficiency of SnoN/SKIL, a repressor of the Smad7 promoter. Smad7 inhibits autocrine expression of TGF-ß(2) in intestinal epithelial cells in the normal premature intestine and during NEC. Increased Smad7 expression in the developing intestine may be due to a developmental deficiency of the SnoN/SKIL oncoprotein.


Assuntos
Comunicação Autócrina , Colo/metabolismo , Enterocolite Necrosante/metabolismo , Mucosa Intestinal/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Western Blotting , Linhagem Celular , Colo/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Enterocolite Necrosante/genética , Enterocolite Necrosante/patologia , Idade Gestacional , Humanos , Imuno-Histoquímica , Mucosa Intestinal/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Papio anubis , Papio cynocephalus , Nascimento Prematuro , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad7/genética , Transfecção , Fator de Crescimento Transformador beta2/genética
12.
Am J Physiol Gastrointest Liver Physiol ; 303(1): G93-102, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22538401

RESUMO

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.


Assuntos
Mucosa Intestinal/patologia , Macrófagos/fisiologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting , Quimiocina CXCL5/fisiologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Eletroforese em Gel de Gradiente Desnaturante , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Recém-Nascido , Recém-Nascido Prematuro , Inflamação/patologia , Enteropatias/induzido quimicamente , Enteropatias/patologia , Mucosa Intestinal/citologia , Camundongos , Infiltração de Neutrófilos/fisiologia , Reação em Cadeia da Polimerase , Fatores de Risco , Ácido Trinitrobenzenossulfônico
13.
J Biol Chem ; 286(48): 41425-41433, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21979956

RESUMO

Using mass spectrometric analysis we found that oncogenic transcription factor FOXM1 that is overexpressed in a majority of human cancers interacts with multifunctional protein NPM, which is also overexpressed in a variety of human tumors. Coimmunoprecipitation and glutathione S-transferase pull-down experiments demonstrated that NPM forms a complex with FOXM1 and also identified the regions responsible for their interaction. Immunofluorescence microscopy confirmed the interaction between FOXM1 and NPM in cancer and immortal cells. Furthermore, knockdown of NPM in immortal and cancer cells led to significant down-regulation of FOXM1 similar to its levels in normal cells, suggesting that NPM might modulate FOXM1 level. In addition, in OCI/AML3 leukemia cells where mutant NPM is localized in the cytoplasm we found that typically nuclear FOXM1 was predominantly co-localized with NPM in the cytoplasm, while NPM knockdown led to the disappearance of FOXM1 from the cytoplasm, suggesting that NPM may also determine intracellular localization of FOXM1. Knockdown of FOXM1 or NPM in MIA PaCa-2 pancreatic cancer cells inhibited anchorage-dependent and independent growth in cell culture, and tumor growth in nude mice. In addition, over-expression of FOXM1 reversed the effect of NPM knockdown in vitro. Our data suggest that in cancer cells NPM interacts with FOXM1 and their interaction is required for sustaining the level and localization of FOXM1. Targeting the interaction between FOXM1 and NPM by peptides or small molecules may represent a novel therapeutic strategy against cancer.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Mutação , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/terapia , Proteínas Nucleares/genética , Nucleofosmina , Transporte Proteico/genética
14.
Cancer Res ; 69(7): 3021-31, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318576

RESUMO

Recurrent/metastatic head and neck cancer remains a devastating disease with insufficient treatment options. We investigated the MET receptor tyrosine kinase as a novel target for the treatment of head and neck squamous cell carcinoma (HNSCC). MET/phosphorylated MET and HGF expression was analyzed in 121 tissues (HNSCC/normal) by immunohistochemistry, and in 20 HNSCC cell lines by immunoblotting. The effects of MET inhibition using small interfering RNA/two small-molecule inhibitors (SU11274/PF-2341066) on signaling, migration, viability, and angiogenesis were determined. The complete MET gene was sequenced in 66 head and neck cancer tissue samples and eight cell lines. MET gene copy number was determined in 14 cell lines and 23 tumor tissues. Drug combinations of SU11274 with cisplatin or erlotinib were tested in SCC35/HN5 cell lines. Eighty-four percent of the HNSCC samples showed MET overexpression, whereas 18 of 20 HNSCC cell lines (90%) expressed MET. HGF overexpression was present in 45% of HNSCC. MET inhibition with SU11274/PF-2341066 abrogated MET signaling, cell viability, motility/migration in vitro, and tumor angiogenesis in vivo. Mutational analysis of 66 tumor tissues and 8 cell lines identified novel mutations in the semaphorin (T230M/E168D/N375S), juxtamembrane (T1010I/R988C), and tyrosine kinase (T1275I/V1333I) domains (incidence: 13.5%). Increased MET gene copy number was present with >10 copies in 3 of 23 (13%) tumor tissues. A greater-than-additive inhibition of cell growth was observed when combining a MET inhibitor with cisplatin or erlotinib and synergy may be mediated via erbB3/AKT signaling. MET is functionally important in HNSCC with prominent overexpression, increased gene copy number, and mutations. MET inhibition abrogated MET functions, including proliferation, migration/motility, and angiogenesis. MET is a promising, novel target for HNSCC and combination approaches with cisplatin or EGFR inhibitors should be explored.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Neoplasias de Cabeça e Pescoço/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Receptores de Fatores de Crescimento/antagonistas & inibidores , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Receptores ErbB/antagonistas & inibidores , Dosagem de Genes , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Imuno-Histoquímica , Indóis/administração & dosagem , Indóis/farmacologia , Camundongos , Camundongos Nus , Mutação , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/administração & dosagem , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , RNA Interferente Pequeno/genética , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacologia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
15.
BJU Int ; 103(2): 171-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18990162

RESUMO

OBJECTIVE: To test the hypothesis that FYN, a member of the SRC family of kinases (SFKs), is up-regulated in prostate cancer, as FYN is functionally distinct from other SFKs, and interacts with FAK and paxillin (PXN), regulators of cell morphology and motility. MATERIALS AND METHODS: Through data-mining in Oncomine (http://www.oncomine.org), cell-line profiling with immunoblotting, quantitative reverse transcription and polymerase chain reaction (RT-PCR) and immunohistochemical analysis, we described FYN expression in prostate cancer. The analysis included 32 cases of prostate cancer, nine of prostatic intraepithelial neoplasia (PIN) and 19 normal prostates. Samples were scored for the percentage of stained glands and intensity of staining (from 0 to 3). Each sample was assigned a composite score generated by multiplying percentage and intensity. RESULTS: Data-mining showed an eight times greater FYN expression in prostate cancer than in normal tissue; this was specific to FYN and not present for other SFKs. Expression of FYN in prostate cancer cell lines (LNCaP, 22Rv1, PC3, DuPro) was detected using quantitative RT-PCR and immunoblotting. Expression of FYN and its signalling partners FAK and PXN was detected in human tissue. Comparing normal with cancer samples, there was a 2.1-fold increase in median composite score for FYN (P < 0.001) 1.7-fold increase in FAK (P < 0.001), and a doubling in PXN (P < 0.05). There was a 1.7-fold increase in FYN (P < 0.05) and a 1.6-fold increase in FAK (P < 0.01) in cancer compared with PIN. CONCLUSIONS: These studies support the hypothesis that FYN and its related signalling partners are up-regulated in prostate cancer, and support further investigation into the role of the FYN as a therapeutic target.


Assuntos
Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Adulto , Idoso , Western Blotting , Estudos de Casos e Controles , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Paxilina/metabolismo , Neoplasia Prostática Intraepitelial/genética , Neoplasias da Próstata/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
16.
Genes Chromosomes Cancer ; 47(12): 1025-37, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18709663

RESUMO

MET receptor tyrosine kinase and its ligand hepatocyte growth factor (HGF) regulate a variety of cellular functions, many of which can be dysregulated in human cancers. Activated MET signaling can lead to cell motility and scattering, angiogenesis, proliferation, branching morphogenesis, invasion, and eventual metastasis. We performed systematic analysis of the expression of the MET receptor and its ligand HGF in tumor tissue microarrays (TMA) from human solid cancers. Standard immunohistochemistry (IHC) and a computerized automated scoring system were used. DNA sequencing for MET mutations in both nonkinase and kinase domains was also performed. MET was differentially overexpressed in human solid cancers. The ligand HGF was widely expressed in both tumors, primarily intratumoral, and nonmalignant tissues. The MET/HGF likely is functional and may be activated in autocrine fashion in vivo. MET and stem cell factor (SCF) were found to be positively stained in the bronchioalevolar junctions of lung tumors. A number of novel mutations of MET were identified, particularly in the extracellular semaphorin domain and the juxtamembrane domain. MET-HGF pathway can be assayed in TMAs and is often overexpressed in a wide variety of human solid cancers. MET can be activated through overexpression, mutation, or autocrine signaling in malignant cells. Mutations in the nonkinase regions of MET might play an important role in tumorigenesis and tumor progression. MET would be an important therapeutic antitumor target to be inhibited, and in lung cancer, MET may represent a cancer early progenitor cell marker.


Assuntos
Mutação , Neoplasias/genética , Proteínas Proto-Oncogênicas/genética , Receptores de Fatores de Crescimento/genética , Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias/metabolismo , Neovascularização Patológica , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-met , Receptores de Fatores de Crescimento/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Células Tumorais Cultivadas
17.
Cancer Biol Ther ; 7(6): 856-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18340114

RESUMO

Cancers typically harbour several mutant forms of key cellular genes that contribute to its complex phenotype. Our lab has previously identified gain-of-function mutations in some of the receptor tyrosine kinases such as c-Met in lung cancer. In order to investigate the mutant gene in the context of a whole organism, the current choice of in vivo model is limited to the mouse. To rapidly screen the functional aspects of mutant forms of c-Met detected in lung cancer, we used the nematode C. elegans as the model organism. Transgenic worms were generated that harbour wild type or the frequently seen mutant forms of c-Met in lung cancer (c-MetR988C and c-MetT1010I). Expression of the mutant human c-Met forms in C. elegans consistently resulted in significantly low fecundity and abnormal vulval development characterized by hyperplasia. Interestingly, exposure of c-Met mutant transgenic worms to nicotine resulted in enhanced abnormal vulval development, fecundity and locomotion. Our studies provide first evidence that human c-Met mutations can be studied in C. elegans, and that carcinogens can enhance mutant c-Met function expressed in C. elegans transgenic animals. We therefore propose the use of C. elegans as a model to rapidly assess the role of cancer specific gene mutations in the context of a whole organism.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-met/biossíntese , Proteínas Proto-Oncogênicas c-met/genética , Vulva/efeitos dos fármacos , Vulva/patologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Feminino , Humanos , Mutação , Nicotina/química , Fenótipo , Estrutura Terciária de Proteína , Interferência de RNA
18.
Cancer Res ; 68(1): 132-42, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18172305

RESUMO

Lung cancer is characterized by abnormal cell growth and invasion, and the actin cytoskeleton plays a major role in these processes. The focal adhesion protein paxillin is a target of a number of oncogenes involved in key signal transduction and important in cell motility and migration. In lung cancer tissues, we have found that paxillin was highly expressed (compared with normal lung), amplified (12.1%, 8 of 66) and correlated with increased MET and epidermal growth factor receptor (EGFR) gene copy numbers, or mutated (somatic mutation rate of 9.4%, 18 of 191). Paxillin mutations (19 of 21) were clustered between LD motifs 1 and 2 and the LIM domains. The most frequent point mutation (A127T) enhanced lung cancer cell growth, colony formation, focal adhesion formation, and colocalized with Bcl-2 in vitro. Gene silencing from RNA interference of mutant paxillin led to reduction of cell viability. A murine in vivo xenograft model of A127T paxillin showed an increase in tumor growth, cell proliferation, and invasion. These results establish an important role for paxillin in lung cancer.


Assuntos
Neoplasias Pulmonares/patologia , Paxilina/metabolismo , Animais , Proliferação de Células , Dosagem de Genes , Genes erbB-1 , Humanos , Neoplasias Pulmonares/etnologia , Neoplasias Pulmonares/genética , Camundongos , Camundongos Endogâmicos , Mutação , Invasividade Neoplásica , Paxilina/análise , Paxilina/genética , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Interferência de RNA
19.
Clin Cancer Res ; 13(7): 2246-53, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17404109

RESUMO

PURPOSE: c-Met is a receptor tyrosine kinase involved in cell growth, invasion, metastases, and angiogenesis. In this study, we investigated the role of c-Met in melanoma biology using a novel small-molecule tyrosine kinase inhibitor SU11274 and small interfering (si) RNA against the receptor. EXPERIMENTAL DESIGN: The effects of SU11274 and c-Met siRNA were studied on proliferation, apoptosis, differentiation, reactive oxygen species, and intracellular signaling. c-Met mutations were examined, and the expression of c-Met and activated c-Met was studied in nevi, primary, and metastatic melanoma. RESULTS: c-Met was expressed in 6:7 melanoma cell lines by immunoblotting. SU11274 inhibited cell growth in all melanoma cell lines by 85% to 98% with an IC(50) between 1 and 2.5 mumol/L and caused apoptosis (12-58%) in five out of six cell lines. siRNA against c-Met inhibited proliferation of melanoma cells by 60%. This is the first study that shows that SU11274 and siRNA induced microphthalmia-associated transcription factor (MITF) and several other melanoma differentiation proteins and a morphologically differentiated phenotype. SU11274 also inhibited reactive oxygen species formation and phosphorylation of c-Met receptor, AKT and S-6 kinase by the hepatocyte growth factor. A new missense c-Met mutation N948S was identified in cell lines and R988C in tumor tissue in the juxtamembrane domain of c-Met. It was found that c-Met was expressed in 88% of melanomas and 15% of nevi, and that c-Met (pY1003) was activated in 21% of human melanomas. CONCLUSION: These results support the role of c-Met in proliferation, apoptosis, differentiation, and tumor progression of melanoma. SU11274 could be used in the therapeutic inhibition of melanoma.


Assuntos
Indóis/farmacologia , Melanoma/tratamento farmacológico , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Sulfonamidas/farmacologia , Sequência de Aminoácidos , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Imunofluorescência , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Imuno-Histoquímica , Melanoma/metabolismo , Fator de Transcrição Associado à Microftalmia , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-met/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/genética , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/metabolismo , Transfecção
20.
Cancer Res ; 67(8): 3529-34, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17440059

RESUMO

The c-Met receptor tyrosine kinase is emerging as a novel target in many solid tumors, including lung cancer. PHA-665752 was identified as a small molecule, ATP competitive inhibitor of the catalytic activity of the c-Met kinase. Here, we show that treatment with PHA665752 reduced NCI-H69 (small cell lung cancer) and NCI-H441 (non-small cell lung cancer) tumorigenicity in mouse xenografts by 99% and 75%, respectively. Reduction in tumor size was also observed by magnetic resonance imaging of tumors in mice. PHA665752 inhibited c-Met phosphorylation at the autophosphorylation and c-Cbl binding sites in mouse xenografts derived from non-small cell lung cancer cell lines (NCI-H441 and A549) and small cell lung cancer cell line (NCI-H69). PHA665752 also inhibited angiogenesis by >85% in all the abovementioned cell lines and caused an angiogenic switch which resulted in a decreased production of vascular endothelial growth factor and an increase in the production of the angiogenesis inhibitor thrombospondin-1. These studies show the feasibility of selectively targeting c-Met with ATP competitive small molecule inhibitors and suggest that PHA665752 may provide a novel therapeutic approach to lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma de Células Pequenas/tratamento farmacológico , Indóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Sulfonas/farmacologia , Animais , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Pequenas/irrigação sanguínea , Carcinoma de Células Pequenas/enzimologia , Carcinoma de Células Pequenas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Trombospondina 1/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...