Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Clin Cancer Res ; 25(11): 3384-3391, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30846478

RESUMEN

PURPOSE: Squamous cell carcinoma (SCC) of the skin is the leading cause of death in patients with the severe generalized form of the genetic disease recessive dystrophic epidermolysis bullosa (RDEB). Although emerging data are identifying why patients suffer this fatal complication, therapies for treatment of RDEB SCC are in urgent need.Experimental Design: We previously identified polo-like kinase 1 (PLK1) as a therapeutic target in skin SCC, including RDEB SCC. Here, we undertake a screen of 6 compounds originally designated as PLK1 inhibitors, and detail the efficacy of the lead compound, the multipathway allosteric inhibitor ON-01910, for targeting RDEB SCC in vitro and in vivo. RESULTS: ON-01910 (or rigosertib) exhibited significant specificity for RDEB SCC: in culture rigosertib induced apoptosis in 10 of 10 RDEB SCC keratinocyte populations while only slowing the growth of normal primary skin cells at doses 2 orders of magnitude higher. Furthermore, rigosertib significantly inhibited the growth of two RDEB SCC in murine xenograft studies with no apparent toxicity. Mechanistically, rigosertib has been shown to inhibit multiple signaling pathways. Comparison of PLK1 siRNA with MEK inhibition, AKT inhibition, and the microtubule-disrupting agent vinblastine in RDEB SCC shows that only PLK1 reduction exhibits a similar sensitivity profile to rigosertib. CONCLUSIONS: These data support a "first in RDEB" phase II clinical trial of rigosertib to assess tumor targeting in patients with late stage, metastatic, and/or unresectable SCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/etiología , Epidermólisis Ampollosa Distrófica/complicaciones , Epidermólisis Ampollosa Distrófica/genética , Glicina/análogos & derivados , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/etiología , Sulfonas/uso terapéutico , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células Escamosas/diagnóstico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Genes Recesivos , Glicina/farmacología , Glicina/uso terapéutico , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero , ARN Interferente Pequeño , Neoplasias Cutáneas/diagnóstico , Sulfonas/farmacología , Quinasa Tipo Polo 1
2.
Sci Transl Med ; 10(455)2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135250

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare inherited skin and mucous membrane fragility disorder complicated by early-onset, highly malignant cutaneous squamous cell carcinomas (SCCs). The molecular etiology of RDEB SCC, which arises at sites of sustained tissue damage, is unknown. We performed detailed molecular analysis using whole-exome, whole-genome, and RNA sequencing of 27 RDEB SCC tumors, including multiple tumors from the same patient and multiple regions from five individual tumors. We report that driver mutations were shared with spontaneous, ultraviolet (UV) light-induced cutaneous SCC (UV SCC) and head and neck SCC (HNSCC) and did not explain the early presentation or aggressive nature of RDEB SCC. Instead, endogenous mutation processes associated with apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) deaminases dominated RDEB SCC. APOBEC mutation signatures were enhanced throughout RDEB SCC tumor evolution, relative to spontaneous UV SCC and HNSCC mutation profiles. Sixty-seven percent of RDEB SCC driver mutations was found to emerge as a result of APOBEC and other endogenous mutational processes previously associated with age, potentially explaining a >1000-fold increased incidence and the early onset of these SCCs. Human papillomavirus-negative basal and mesenchymal subtypes of HNSCC harbored enhanced APOBEC mutational signatures and transcriptomes similar to those of RDEB SCC, suggesting that APOBEC deaminases drive other subtypes of SCC. Collectively, these data establish specific mutagenic mechanisms associated with chronic tissue damage. Our findings reveal a cause for cancers arising at sites of persistent inflammation and identify potential therapeutic avenues to treat RDEB SCC.


Asunto(s)
Desaminasas APOBEC/genética , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Citosina Desaminasa/genética , Epidermólisis Ampollosa Distrófica/enzimología , Epidermólisis Ampollosa Distrófica/genética , Mutación/genética , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Variaciones en el Número de Copia de ADN/genética , Reparación del ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Mutagénesis/genética , Tasa de Mutación , Transcriptoma/genética
3.
Oncotarget ; 8(47): 81754-81775, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29137220

RESUMEN

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that drives cell autonomous cell cycle progression and proliferation. Herein we show cyclin D1 abundance is increased >30-fold in the stromal fibroblasts of patients with invasive breast cancer, associated with poor outcome. Cyclin D1 transformed hTERT human fibroblast to a cancer-associated fibroblast phenotype. Stromal fibroblast expression of cyclin D1 (cyclin D1Stroma) in vivo, enhanced breast epithelial cancer tumor growth, restrained apoptosis, and increased autophagy. Cyclin D1Stroma had profound effects on the breast tumor microenvironment increasing the recruitment of F4/80+ and CD11b+ macrophages and increasing angiogenesis. Cyclin D1Stroma induced secretion of factors that promoted expansion of stem cells (breast stem-like cells, embryonic stem cells and bone marrow derived stem cells). Cyclin D1Stroma resulted in increased secretion of proinflammatory cytokines (CCL2, CCL7, CCL11, CXCL1, CXCL5, CXCL9, CXCL12), CSF (CSF1, GM-CSF1) and osteopontin (OPN) (30-fold). OPN was induced by cyclin D1 in fibroblasts, breast epithelial cells and in the murine transgenic mammary gland and OPN was sufficient to induce stem cell expansion. These results demonstrate that cyclin D1Stroma drives tumor microenvironment heterocellular signaling, promoting several key hallmarks of cancer.

4.
Am J Transl Res ; 8(5): 2432-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27347352

RESUMEN

Transforming growth factor-beta (TGFß) signaling in cancer is context dependent and acts either as a tumor suppressor or a tumor promoter. Loss of function mutation in TGFß type II receptor (TßRII) is a frequent event in oral cavity squamous cell carcinoma (SCC). Recently, heterogeneity of TGFß response has been described at the leading edge of SCC and this heterogeneity has been shown to influence stem cell renewal and drug resistance. Because exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways we investigated whether exosomes contain components of the TGFß signaling pathway and whether exosome transfer between stromal fibroblasts and tumor cells can influence TGFß signaling in SCC. We demonstrate that exosomes purified from stromal fibroblasts isolated from patients with oral SCC contains TßRII. We also demonstrate that transfer of fibroblast exosomes increases TGFß signaling in SCC keratinocytes devoid of TßRII which remain non-responsive to TGFß ligand in the absence of exosome transfer. Overall our data show that stromal communication with tumor cells can direct TGFß signaling in SCC.

5.
J Foot Ankle Surg ; 55(2): 230-4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26620421

RESUMEN

The purpose of the present retrospective study was to evaluate the outcomes (ie, ulcer recurrence, major amputation, death) in diabetic patients undergoing Chopart amputation because of deep infection or gangrene extending to the midfoot. From 2009 to 2011, 83 patients, aged 71.4 ± 9.3 years, underwent a midtarsal amputation and were followed up until December 31, 2012 (mean follow-up 2.8 ± 0.8 years). Of the 83 patients, 26 were female, 61 required insulin, 47 had renal insufficiency, 19 underwent hemodialysis, 65 had hypertension, 34 had a history of cardiac disease, and 4 had a history of stroke. Chopart amputation was performed in 38 patients (45.8%) with gangrene, 31 (37.4%) with abscess, and 14 (16.9%) with osteomyelitis. Urgent surgery was performed in 56 patients (67.5%). Effective revascularization was performed in 64 patients (77.1%) patients. Of the 83 patients, 47 had healed at a mean period of 164.7 (range 11 to 698) days. Ulcer recurrence developed in 15 patients (31.9%). A major amputation was necessary in 23 patients (27.7%), with an annual incidence of 13.0%. None of the included variables on logistic regression analysis was significantly associated with proximal amputation. Of the 83 patients, 38 (45.8%) died, with an annual incidence of 25.8%. On logistic regression analysis, age (odds ratio [OR] 1.11, 95% confidence interval [CI] 1.01 to 1.16), history of stroke (OR 9.94, 95% CI 3.16 to 31.24), and urgent surgery (OR 2.60, 95% CI 1.14 to 5.93) were associated with mortality. Chopart amputation represents the last chance to avoid major amputation for diabetic patients with serious foot complications. Our success rate was great enough to consider Chopart amputation a viable option for limb salvage in this high-risk population.


Asunto(s)
Amputación Quirúrgica , Pie Diabético/cirugía , Absceso/etiología , Absceso/cirugía , Anciano , Anciano de 80 o más Años , Amputación Quirúrgica/efectos adversos , Pie Diabético/complicaciones , Pie Diabético/fisiopatología , Femenino , Gangrena/etiología , Gangrena/cirugía , Humanos , Recuperación del Miembro , Masculino , Persona de Mediana Edad , Osteomielitis/etiología , Osteomielitis/cirugía , Estudios Retrospectivos , Resultado del Tratamiento , Cicatrización de Heridas
6.
PLoS One ; 10(9): e0137639, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26380979

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by mutations in COL7A1 resulting in reduced or absent type VII collagen, aberrant anchoring fibril formation and subsequent dermal-epidermal fragility. Here, we identify a significant decrease in PLOD3 expression and its encoded protein, the collagen modifying enzyme lysyl hydroxylase 3 (LH3), in RDEB. We show abundant LH3 localising to the basement membrane in normal skin which is severely depleted in RDEB patient skin. We demonstrate expression is in-part regulated by endogenous type VII collagen and that, in agreement with previous studies, even small reductions in LH3 expression lead to significantly less secreted LH3 protein. Exogenous type VII collagen did not alter LH3 expression in cultured RDEB keratinocytes and we show that RDEB patients receiving bone marrow transplantation who demonstrate significant increase in type VII collagen do not show increased levels of LH3 at the basement membrane. Our data report a direct link between LH3 and endogenous type VII collagen expression concluding that reduction of LH3 at the basement membrane in patients with RDEB will likely have significant implications for disease progression and therapeutic intervention.


Asunto(s)
Membrana Basal/enzimología , Membrana Basal/patología , Epidermólisis Ampollosa Distrófica/enzimología , Epidermólisis Ampollosa Distrófica/patología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/análisis , Membrana Basal/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Colágeno Tipo VII/análisis , Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/metabolismo , Epidermólisis Ampollosa Distrófica/terapia , Humanos , Queratinocitos/enzimología , Queratinocitos/metabolismo , Queratinocitos/patología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Mapas de Interacción de Proteínas , Piel/enzimología , Piel/metabolismo , Piel/patología
7.
Ann Vasc Surg ; 28(7): 1729-36, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24952297

RESUMEN

BACKGROUND: To compare demographic and clinical characteristics, revascularization, major amputation, and mortality among patients admitted to a diabetic foot center because of critical limb ischemia (CLI) during 1999-2003 (cohort 1) and 2009 (cohort 2). METHODS: During 1999-2003, 564 diabetic patients with CLI (cohort 1) were admitted to our center, and 344 patients (360 affected limbs) were admitted during 2009 (cohort 2). Data on demographic and clinical characteristics, revascularization by peripheral angioplasty (PTA) or bypass graft (BPG), major amputation, and mortality were recorded. RESULTS: Patients belonging to cohort 2 were older than patients of cohort 1 (P = 0.001). In cohort 2, there were more subjects requiring insulin (P = 0.008) and duration of diabetes was longer (P = 0.001); moreover, there were more patients requiring dialysis (P = 0.001), patients with history of stroke (P = 0.004), or foot ulcer (P = 0.001). No significant difference between the 2 groups was found concerning gender, metabolic control, hypertension, lipid values, neuropathy, and retinopathy. Occlusion was more frequent than stenosis in the posterior tibial (P < 0.001) and peroneal (P = 0.016) arteries. However, the revascularization rate did not differ (P = 0.318) between the 2 groups. Restenosis after PTA was not significantly different (P = 0.627), whereas BPG failure was significantly more frequent (P = 0.010) in cohort 2 (2009). Major amputation (P = 0.222) and mortality rate (P = 0.727) did not differ between the 2 groups. CONCLUSIONS: The severity of either foot lesions or patients comorbidities should be concomitantly assessed and taken into proper consideration when evaluating changes in the amputation rate among different studies or in different temporal settings.


Asunto(s)
Amputación Quirúrgica , Pie Diabético/mortalidad , Pie Diabético/cirugía , Isquemia/mortalidad , Isquemia/cirugía , Pierna/irrigación sanguínea , Anciano , Angioplastia , Implantación de Prótesis Vascular , Estudios de Cohortes , Comorbilidad , Femenino , Humanos , Recuperación del Miembro , Masculino , Factores de Riesgo , Índice de Severidad de la Enfermedad , Tasa de Supervivencia , Resultado del Tratamiento
8.
Cell Cycle ; 10(8): 1271-86, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21512313

RESUMEN

Previously, we showed that high-energy metabolites (lactate and ketones) "fuel" tumor growth and experimental metastasis in an in vivo xenograft model, most likely by driving oxidative mitochondrial metabolism in breast cancer cells. To mechanistically understand how these metabolites affect tumor cell behavior, here we used genome-wide transcriptional profiling. Briefly, human breast cancer cells (MCF7) were cultured with lactate or ketones, and then subjected to transcriptional analysis (exon-array). Interestingly, our results show that treatment with these high-energy metabolites increases the transcriptional expression of gene profiles normally associated with "stemness," including genes upregulated in embryonic stem (ES) cells. Similarly, we observe that lactate and ketones promote the growth of bonafide ES cells, providing functional validation. The lactate- and ketone-induced "gene signatures" were able to predict poor clinical outcome (including recurrence and metastasis) in a cohort of human breast cancer patients. Taken together, our results are consistent with the idea that lactate and ketone utilization in cancer cells promotes the "cancer stem cell" phenotype, resulting in significant decreases in patient survival. One possible mechanism by which these high-energy metabolites might induce stemness is by increasing the pool of Acetyl-CoA, leading to increased histone acetylation, and elevated gene expression. Thus, our results mechanistically imply that clinical outcome in breast cancer could simply be determined by epigenetics and energy metabolism, rather than by the accumulation of specific "classical" gene mutations. We also suggest that high-risk cancer patients (identified by the lactate/ketone gene signatures) could be treated with new therapeutics that target oxidative mitochondrial metabolism, such as the anti-oxidant and "mitochondrial poison" metformin. Finally, we propose that this new approach to personalized cancer medicine be termed "Metabolo-Genomics," which incorporates features of both 1) cell metabolism and 2) gene transcriptional profiling. Importantly, this powerful new approach directly links cancer cell metabolism with clinical outcome, and new therapeutic strategies for inhibiting the TCA cycle and mitochondrial oxidative phosphorylation in cancer cells.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cetonas/metabolismo , Ácido Láctico/metabolismo , Células Madre/metabolismo , Acetilcoenzima A/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclo del Ácido Cítrico , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genómica , Glucólisis , Humanos , Metabolómica , Mitocondrias/metabolismo , Metástasis de la Neoplasia , Fosforilación Oxidativa , Medicina de Precisión/métodos , Recurrencia , Células Madre/patología , Resultado del Tratamiento
9.
Cancer Res ; 70(20): 7949-59, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20924107

RESUMEN

Ectopic expression of CAAT/enhancer binding protein α (C/EBPα) in p210BCR/ABL-expressing cells induces granulocytic differentiation, inhibits proliferation, and suppresses leukemogenesis. To dissect the molecular mechanisms underlying these biological effects, C/EBPα-regulated genes were identified by microarray analysis in 32D-p210BCR/ABL cells. One of the genes whose expression was activated by C/EBPα in a DNA binding-dependent manner in BCR/ABL-expressing cells is the transcriptional repressor Gfi-1. We show here that C/EBPα interacts with a functional C/EBP binding site in the Gfi-1 5'-flanking region and enhances the promoter activity of Gfi-1. Moreover, in K562 cells, RNA interference-mediated downregulation of Gfi-1 expression partially rescued the proliferation-inhibitory but not the differentiation-inducing effect of C/EBPα. Ectopic expression of wild-type Gfi-1, but not of a transcriptional repressor mutant (Gfi-1P2A), inhibited proliferation and markedly suppressed colony formation but did not induce granulocytic differentiation of BCR/ABL-expressing cells. By contrast, Gfi-1 short hairpin RNA-tranduced CD34(+) chronic myeloid leukemia cells were markedly more clonogenic than the scramble-transduced counterpart. Together, these studies indicate that Gfi-1 is a direct target of C/EBPα required for its proliferation and survival-inhibitory effects in BCR/ABL-expressing cells.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Fusión bcr-abl/genética , Factores de Transcripción/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , División Celular , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Regulación hacia Abajo , Amplificación de Genes , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Secuencias Invertidas Repetidas/genética , Células K562 , Luciferasas/genética , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/fisiología , Transcripción Genética , Transfección
10.
Blood ; 112(5): 1942-50, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18550858

RESUMEN

Ectopic C/EBPalpha expression in p210(BCR/ABL)-expressing hematopoietic cells induces granulocytic differentiation, inhibits proliferation, and suppresses leukemogenesis. To assess the underlying mechanisms, C/EBPalpha targets were identified by microarray analyses. Upon C/EBPalpha activation, expression of c-Myb and GATA-2 was repressed in 32D-BCR/ABL, K562, and chronic myelogenous leukemia (CML) blast crisis (BC) primary cells but only c-Myb levels decreased slightly in CD34(+) normal progenitors. The role of these 2 genes for the effects of C/EBPalpha was assessed by perturbing their expression in K562 cells. Ectopic c-Myb expression blocked the proliferation inhibition- and differentiation-inducing effects of C/EBPalpha, whereas c-Myb siRNA treatment enhanced C/EBPalpha-mediated proliferation inhibition and induced changes in gene expression indicative of monocytic differentiation. Ectopic GATA-2 expression suppressed the proliferation inhibitory effect of C/EBPalpha but blocked in part the effect on differentiation; GATA-2 siRNA treatment had no effects on C/EBPalpha induction of differentiation but inhibited proliferation of K562 cells, alone or upon C/EBPalpha activation. In summary, the effects of C/EBPalpha in p210(BCR/ABL)-expressing cells depend, in part, on transcriptional repression of c-Myb and GATA-2. Since perturbation of c-Myb and GATA-2 expression has nonidentical consequences for proliferation and differentiation of K562 cells, the effects of C/EBPalpha appear to involve dif-ferent transcription-regulated targets.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/farmacología , Proteínas de Fusión bcr-abl/biosíntesis , Factor de Transcripción GATA2/genética , Genes myb/efectos de los fármacos , Secuencia de Bases , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cartilla de ADN/genética , Proteínas de Fusión bcr-abl/genética , Genes abl , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , ARN Interferente Pequeño/genética , Transcripción Genética/efectos de los fármacos , Transfección
11.
J Biol Chem ; 282(45): 32582-90, 2007 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-17827156

RESUMEN

The insulin receptor substrate-1 (IRS-1), a docking protein for both the type 1 insulin-like growth factor receptor (IGF-IR) and the insulin receptor, is known to send a mitogenic, anti-apoptotic, and anti-differentiation signal. Several micro RNAs (miRs) are suggested by the data base as possible candidates for targeting IRS-1. We show here that one of the miRs predicted by the data base, miR145, whether transfected as a synthetic oligonucleotide or expressed from a plasmid, causes down-regulation of IRS-1 in human colon cancer cells. IRS-1 mRNA is not decreased by miR145, while it is down-regulated by an siRNA targeting IRS-1. Targeting of the IRS-1 3'-untranslated region (UTR) by miR145 was confirmed using a reporter gene (luciferase) expressing the miR145 binding sites of the IRS-1 3'-UTR. In agreement with the role of IRS-1 in cell proliferation, we show that treatment of human colon cancer cells with miR145 causes growth arrest comparable to the use of an siRNA against IRS-1. Taken together, these results identify miR145 as a micro RNA that down-regulates the IRS-1 protein, and inhibits the growth of human cancer cells.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , MicroARNs/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Genes Reporteros/genética , Humanos , Proteínas Sustrato del Receptor de Insulina , ARN Mensajero/genética
12.
J Cell Physiol ; 213(2): 445-53, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17620314

RESUMEN

Mouse embryonic stem (mES) cells are pluripotent cells that can be propagated in vitro with leukemia inhibitory factor (LIF) and serum. Intracellular signaling by LIF is principally mediated by activation of STAT-3, although additional pathways for self-renewal have been described. Here, we identified a novel role for Insulin receptor substrate-1 (IRS-1) as a critical factor in mES cells self-renewal and differentiation. IRS-1 is expressed and tyrosyl phosphorylated during mES cells self-renewal. Differentiation of mES cells, by LIF withdrawal, is associated with a marked reduction in IRS-1 expression. Targeting of IRS-1 by si-IRS-1 results in a severe reduction of Oct-4 protein expression and alkaline phosphatase activity, markers of undifferentiated mES cells. IRS-1 targeting does not interfere with LIF-induced STAT-3 phosphorylation, but negatively affects protein kinase B (PKB/AKT) and glycogen synthase kinase-3 (GSK-3beta) phosphorylation, which are downstream effectors of the LIF-mediated PI3K signaling cascade. Targeting of IRS-1 also results in a marked down regulation of Id-1 and Id-2 proteins expression, which are important components for self-renewal of ES cells. Conversely, over expression of IRS-1 inhibits mES cell differentiation. Taken together, these results suggest that expression and activity of IRS-1 are critical to the maintenance of the self-renewal program in mES cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Fosfoproteínas/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología , Animales , Biomarcadores/metabolismo , Ciclo Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Proteínas Sustrato del Receptor de Insulina , Factor Inhibidor de Leucemia/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
13.
J Cell Physiol ; 207(3): 706-10, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16482530

RESUMEN

32D cells are murine myeloid cells that grow indefinitely in Interleukin-3 (IL-3). In these cells, the type 1 insulin-like growth factor (IGF-I) and granulocytic-colony stimulating factor (G-CSF) induce differentiation to granulocytes. 32D cells do not express insulin receptor substrate-1 (IRS-1) or IRS-2, docking proteins of the IGF-I receptor. Ectopic expression of IRS-1 in these cells inhibits differentiation, the cells become IL-3 independent and IGF-1 dependent and can form tumors in mice. 32D and 32D-derived cells offer a good model in which to study the expression profiles of Micro Rna (miR) related to sustained proliferation or differentiation. We present here the data obtained with miR micro-arrays and identify the miR that are regulated by IGF-1 or G-CSF and are associated with either differentiation or indefinite cell proliferation of 32D murine myeloid cells.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica , MicroARNs/análisis , MicroARNs/genética , Células Mieloides/citología , Células Mieloides/metabolismo , Animales , Línea Celular , Proliferación Celular , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Proteínas Sustrato del Receptor de Insulina , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Fosfoproteínas/metabolismo , Receptor IGF Tipo 1/genética
14.
J Cell Physiol ; 205(2): 302-9, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15895393

RESUMEN

24p3 is a secreted lipocalin that has been variously related to apoptosis, proliferation, and the neutrophil lineage of blood cells. We have investigated the expression of 24p3 mRNA and protein in myeloid cell lines induced to differentiate by insulin-like growth factor 1 (IGF-1) and the granulocytic-colony simulating factor (G-CSF). Both these growth factors, which cause myeloid cells to differentiate into granulocytes, induced a marked increase in the expression of both 24p3 protein and mRNA. The mRNA especially appeared early after the cells were induced with either IGF-1 or G-CSF, at a time when the cells were still proliferating and are morphologically undifferentiated. 24p3 can be considered an early marker of granulocytic differentiation.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Diferenciación Celular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Células Mieloides/metabolismo , Proteínas Oncogénicas/metabolismo , Biomarcadores , Técnicas de Cultivo de Célula , Línea Celular , Granulocitos/fisiología , Humanos , Lipocalina 2 , Lipocalinas , Células Mieloides/citología , ARN Mensajero/metabolismo
15.
J Biol Chem ; 280(4): 2863-72, 2005 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-15533945

RESUMEN

The upstream binding factor 1 (UBF1) is one of the proteins in a complex that regulates the activity of RNA polymerase I, which controls the rate of ribosomal RNA (rRNA) synthesis. We have shown previously that insulin receptor substrate-1 (IRS-1) can translocate to the nuclei and nucleoli of cells and bind UBF1. We report here that activation of the type I insulin-like growth factor receptor (IGF-IR) by IGF-I increases transcription from the ribosomal DNA (rDNA) promoter in both myeloid cells and mouse fibroblasts. The increased activity of the rDNA promoter is accompanied by increased phosphorylation of UBF1, a requirement for UBF1 activation. Phosphorylation occurs on a number of UBF1 peptides, most prominently on the highly acidic, serine-rich C terminus. In myeloid cells (but not in mouse embryo fibroblasts) IRS-1 signaling stabilizes the levels of UBF1 protein. These findings demonstrate that IGF-IR signaling can increase the activity of UBF1 and transcription from the rDNA promoter, providing one explanation for the reported effects of the IGF/IRS-1 axis on cell and body size in animals and cells in culture.


Asunto(s)
Regulación de la Expresión Génica , Proteínas del Complejo de Iniciación de Transcripción Pol1/biosíntesis , Receptor IGF Tipo 1/metabolismo , Células 3T3 , Animales , Northern Blotting , Western Blotting , Diferenciación Celular , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , ADN Ribosómico/química , ADN Ribosómico/metabolismo , Exones , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Mutación , Péptidos/química , Fosforilación , Fosfotirosina/química , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , Ribosomas/química , Ribosomas/metabolismo , Factores de Tiempo , Transcripción Genética , Tripsina/farmacología
16.
Rev. venez. urol ; 50(2): 70-74, jul.-dic. 2004. ilus
Artículo en Español | LILACS | ID: lil-431604

RESUMEN

El objetivo de este trabajo es investigar la utilidad del uso de Cianocrilato en la síntesis de heridas de fimosectomías realizadas en el Hospital "Miguel Pérez Carreño" durante el período de enero a diciembre de 2003. Se realizó estudio prospectivo, procediéndose a la síntesis de heridas de fimosectomías con Cianocrilato en 10 pacientes y comparándose en 10 pacientes en quienes la síntesis se realizó con Catgut Crómico, tomando en cuenta el tiempo utilizado en la síntesis, dolor, complicaciones y el tiempo para reinicio de relaciones sexules. Con el uso de Cianocrilato no se evidenciaron cicatrices hipertróficas. El tiempo utilizado en la síntesis con Cianocrilato fue significativamente menor que con la sutura (P<0,0001), al igual que el dolor a las dos semanas de postoperatorio (p<0,000092) y el tiempo de reinicio de relaciones sexuales (P<0,00033) con un valor de 4.87 y 11.03 días más corto


Asunto(s)
Masculino , Humanos , Fimosis , Circuncisión Masculina , Cianoacrilatos , Cicatrización de Heridas , Urología , Venezuela
17.
Oncogene ; 23(46): 7701-11, 2004 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-15361847

RESUMEN

Id1 is a helix-loop-helix transcriptional factor that controls growth and survival of neuronal cells. Downregulation of Id1 expression is required to initiate differentiation and cell-cycle withdrawal in primary neuronal culture as well as in PC12 cells. The HIV-1 transactivating factor, Tat, has been suspected of causing neuronal dysfunction that often leads to the development of HIV-associated dementia in AIDS patients. We found that the expression of Tat in PC12 cells promotes serum-independent growth, formation of large colonies in soft agar, and the acceleration of tumor growth in nude mice. In addition, Tat showed the ability to inhibit the nerve growth factor (NGF)-induced neuronal differentiation of PC12 cells. Our results show that the Tat-mediated signaling events, which lead to serum-independent growth and the inhibition of NGF-induced differentiation, have a common cellular target: the upregulation of Id1 expression. In the absence of NGF, expression of Id1 is required to promote serum-independent proliferation of PC12/Tat cells, as the inhibition of Id1 by antisense DNA restored the serum-dependent growth of PC12/Tat cells. In the presence of NGF, Tat utilizes an additional pathway that involves phosphorylation of Stat5a, to upregulate Id1 expression and block neuronal cell differentiation. Suppression of Stat5a by use of its dominant-negative mutant reversed the transient expression of Id1 and the blockage of NGF-mediated differentiation in PC12/Tat cells. Finally, the treatment of PC12 cells with recombinant Tat also enhanced the NGF-induced Id1 expression, further pointing to Id1 as a target for Tat. Taken together, these studies suggest additional targets for Tat action in neuronal cells and provide new insights into the mechanisms involved in the dysregulation of neuronal functions.


Asunto(s)
Productos del Gen tat/farmacología , Factor de Crecimiento Nervioso/farmacología , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular , VIH-1/fisiología , Proteína 1 Inhibidora de la Diferenciación , Neuronas/citología , Neuronas/fisiología , Células PC12 , Feocromocitoma , Ratas , Proteínas Recombinantes/farmacología , Proteínas Represoras/genética , Factores de Transcripción/genética , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
18.
Mol Cell Biol ; 24(12): 5421-33, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15169904

RESUMEN

Pescadillo (PES1) and the upstream binding factor (UBF1) play a role in ribosome biogenesis, which regulates cell size, an important component of cell proliferation. We have investigated the effects of PES1 and UBF1 on the growth and differentiation of cell lines derived from 32D cells, an interleukin-3 (IL-3)-dependent murine myeloid cell line. Parental 32D cells and 32D IGF-IR cells (expressing increased levels of the type 1 insulin-like growth factor I [IGF-I] receptor [IGF-IR]) do not express insulin receptor substrate 1 (IRS-1) or IRS-2. 32D IGF-IR cells differentiate when the cells are shifted from IL-3 to IGF-I. Ectopic expression of IRS-1 inhibits differentiation and transforms 32D IGF-IR cells into a tumor-forming cell line. We found that PES1 and UBF1 increased cell size and/or altered the cell cycle distribution of 32D-derived cells but failed to make them IL-3 independent. PES1 and UBF1 also failed to inhibit the differentiation program initiated by the activation of the IGF-IR, which is blocked by IRS-1. 32D IGF-IR cells expressing PES1 or UBF1 differentiate into granulocytes like their parental cells. In contrast, PES1 and UBF1 can transform mouse embryo fibroblasts that have high levels of endogenous IRS-1 and are not prone to differentiation. Our results provide a model for one of the theories of myeloid leukemia, in which both a stimulus of proliferation and a block of differentiation are required for leukemia development.


Asunto(s)
Células Mieloides/citología , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/fisiología , Proteínas/genética , Proteínas/fisiología , Animales , Secuencia de Bases , Ciclo Celular , Proteínas de Ciclo Celular , Diferenciación Celular/genética , Diferenciación Celular/fisiología , División Celular/genética , División Celular/fisiología , Línea Celular , ADN Complementario/genética , Proteínas Sustrato del Receptor de Insulina , Factor I del Crecimiento Similar a la Insulina/farmacología , Leucemia Mieloide/etiología , Ratones , Modelos Biológicos , Células Mieloides/efectos de los fármacos , Células Mieloides/fisiología , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Proteínas de Unión al ARN , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/fisiología , Transducción Genética
19.
Exp Cell Res ; 288(1): 119-30, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12878164

RESUMEN

We have investigated the intracellular localization of four proteins in murine hemopoietic 32D and 32D-derived cells during exponential growth and after induction of differentiation. The four proteins studied were the insulin receptor substrate-1 (IRS-1), the ID2 protein, nucleolin, and the upstream binding factor (UBF), all of which are involved directly or indirectly in the differentiation program. These four proteins were found to be predominantly nuclear (and/or nucleolar) during exponential growth, as expected. In three models of induced differentiation along the granulocytic pathway, IRS-1, ID2, and nucleolin shifted in part to the cytoplasm, where their levels eventually decreased. UBF also disappeared during differentiation, but we could not detect a cytoplasmic shift in this protein. These experiments indicate that induction of granulocytic differentiation in 32D and 32D-derived cells is accompanied by intracellular redistribution of proteins. This nucleo-cytoplasmic shuttle may play a significant role in the changes in gene expression that occur during differentiation.


Asunto(s)
Núcleo Celular/metabolismo , Células Madre Hematopoyéticas/citología , Proteínas Nucleares/metabolismo , Proteínas Represoras , Transporte Activo de Núcleo Celular , Animales , Diferenciación Celular , Línea Celular , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Proteína 2 Inhibidora de la Diferenciación , Proteínas Sustrato del Receptor de Insulina , Ratones , Fosfoproteínas/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Transporte de Proteínas , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Nucleolina
20.
Endocrinology ; 144(6): 2650-8, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12746329

RESUMEN

The A isoform of the insulin receptor (IR) is frequently overexpressed in cancer cells and is activated by IGF-II as well as by insulin, whereas the B isoform is predominant in differentiated tissues and responds poorly to IGF-II. The IR substrate-1 (IRS-1), a docking protein for the IR, is known to send a mitogenic signal and to be a powerful inhibitor of cell differentiation. We have investigated the biological effects of the two IR isoforms in parental 32D hemopoietic cells, which do not express IRS-1, and in 32D-derived cells in which IRS-1 is ectopically expressed. The effects of the two isoforms on cell survival, differentiation markers and nuclear translocation of IRS-1 were compared. The results confirm that the A isoform responds to IGF-II and preferentially sends mitogenic, antiapoptotic signals, whereas the B form, poorly responsive to IGF-II, tends to send differentiation signals.


Asunto(s)
Fosfoproteínas/metabolismo , Receptor de Insulina/metabolismo , Proteínas Represoras , Transducción de Señal/fisiología , Proteínas de Fase Aguda/genética , Animales , Antígenos CD , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Núcleo Celular/metabolismo , Supervivencia Celular , Proteínas de Unión al ADN/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Proteína 2 Inhibidora de la Diferenciación , Proteínas Sustrato del Receptor de Insulina , Interleucina-3/farmacología , Isomerismo , Lipocalina 2 , Lipocalinas , Ratones , Proteínas Oncogénicas/genética , Peroxidasa/genética , Fosfoproteínas/farmacología , Proteínas Proto-Oncogénicas , Receptor de Insulina/química , Receptor de Insulina/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA