Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Am J Respir Crit Care Med ; 198(7): 850-858, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29746147

RESUMEN

Female sex/gender is an undercharacterized variable in studies related to lung development and disease. Notwithstanding, many aspects of lung and sleep biology and pathobiology are impacted by female sex and female reproductive transitions. These may manifest as differential gene expression or peculiar organ development. Some conditions are more prevalent in women, such as asthma and insomnia, or, in the case of lymphangioleiomyomatosis, are seen almost exclusively in women. In other diseases, presentation differs, such as the higher frequency of exacerbations experienced by women with chronic obstructive pulmonary disease or greater cardiac morbidity among women with sleep-disordered breathing. Recent advances in -omics and behavioral science provide an opportunity to specifically address sex-based differences and explore research needs and opportunities that will elucidate biochemical pathways, thus enabling more targeted/personalized therapies. To explore the status of and opportunities for research in this area, the NHLBI, in partnership with the NIH Office of Research on Women's Health and the Office of Rare Diseases Research, convened a workshop of investigators in Bethesda, Maryland on September 18 and 19, 2017. At the workshop, the participants reviewed the current understanding of the biological, behavioral, and clinical implications of female sex and gender on lung and sleep health and disease, and formulated recommendations that address research gaps, with a view to achieving better health outcomes through more precise management of female patients with nonneoplastic lung disease. This report summarizes those discussions.


Asunto(s)
Enfermedades Pulmonares/epidemiología , Enfermedades Pulmonares/fisiopatología , Trastornos del Sueño-Vigilia/epidemiología , Trastornos del Sueño-Vigilia/fisiopatología , Salud de la Mujer , Adulto , Anciano , Asma/epidemiología , Asma/fisiopatología , Conducta , Comprensión , Manejo de la Enfermedad , Educación , Femenino , Humanos , Persona de Mediana Edad , National Heart, Lung, and Blood Institute (U.S.) , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Medición de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales , Síndromes de la Apnea del Sueño/epidemiología , Síndromes de la Apnea del Sueño/fisiopatología , Estados Unidos
2.
Sarcoidosis Vasc Diffuse Lung Dis ; 35(3): 206-212, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-32476904

RESUMEN

Background: A subset of lymphangioleiomyomatosis (LAM) patients present with normal FEV1 and FVC but with reduced DLCO. Patients with an isolated reduction in DLCO in other diseases appear to be at higher risk for pulmonary hypertension and worse survival but this has not been previously described in LAM patients. Objective: To characterize the prevalence and clinical progression of LAM patients who present with discordantly low DLCO. Methods: This was a retrospective cohort study of LAM patients in two centers in the United States and Brazil. Discordant DLCO was defined as FEV1 >80% predicted, FVC >80% predicted, and DLCO<80% predicted. We compared the rate of decline in pulmonary function, pulmonary artery to aorta (PA-A) ratio, and VEGF-D levels in patients with concordant and discordant DLCO. Results: The overall prevalence of discordant DLCO was 26.0%. Patients with discordant DLCO did not have a higher rate of yearly decline in FEV1 (-1.0±0.6 vs -1.0±0.6, p=0.50), FVC (-1.0±0.7 vs -0.3±0.8, p=0.54), or DLCO (-2.2±0.9 vs -1.6±0.6, p=0.79). They did not have higher rates of PA-A ratio>1 (23.3% vs 20.1%, p=1.00). Patients with discordant DLCO did not have higher levels of VEGF-D (1214±1256 pg/mL vs 1706±1214 pg/mL, p=0.07). Conclusions: LAM patients who present with a discordantly low DLCO do not appear to have different rates of decline in pulmonary function. Additional biological and radiographic markers are needed to more fully characterize this population. (Sarcoidosis Vasc Diffuse Lung Dis 2018; 35: 206-212).

3.
Eur Respir Rev ; 26(143)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28096282

RESUMEN

Lymphangioleiomyomatosis (LAM) is a destructive cystic lung disease. Mammalian target of rapamycin (mTOR) inhibitors are the primary treatment for LAM but it is unknown whether these immunosuppressing medications increase the risk for or the severity of respiratory infections in LAM patients.We searched multiple databases for original articles that reported the rate of respiratory infections in LAM patients treated with mTOR inhibitors or placebo. We calculated incidence rates for respiratory infections in these groups and incidence rate ratios for respiratory infections and severe respiratory infections in mTOR inhibitors treated versus placebo treated patients.11 studies were included. There were 294 patients in the treatment groups and 93 patients in the placebo groups. Among subjects in placebo arms, the incidence rate of respiratory infections was 58.8 per 100 patient-years (95% CI 35.3-82.3 per 100 patient-years). The incidence-rate ratio (IRR) for respiratory infection among treated subjects was 0.71 (95% CI 0.50-1.02; p=0.06 compared to placebo subjects). The IRR for severe respiratory infections among treated subjects was 1.56 (95% CI 0.43-8.55; p=0.52).We found that respiratory infections are common in patients with LAM. Importantly, treatment with mTOR inhibitors does not increase the incidence of these infections and may be protective.


Asunto(s)
Inmunosupresores/uso terapéutico , Linfangioleiomiomatosis/tratamiento farmacológico , Infecciones Oportunistas/epidemiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Infecciones del Sistema Respiratorio/epidemiología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Humanos , Huésped Inmunocomprometido , Inmunosupresores/efectos adversos , Incidencia , Linfangioleiomiomatosis/enzimología , Linfangioleiomiomatosis/epidemiología , Linfangioleiomiomatosis/inmunología , Terapia Molecular Dirigida , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/prevención & control , Factores Protectores , Inhibidores de Proteínas Quinasas/efectos adversos , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Resultado del Tratamiento
4.
Eur Respir J ; 46(3): 783-94, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26113676

RESUMEN

Lymphangioleiomyomatosis is a rare, progressive cystic lung disorder characterised by dysregulated activation of mammalian target of rapamycin (mTOR) signalling.This was a phase IIa, multicentre, open-label study of the mTOR inhibitor everolimus (2.5 mg·day(-1) escalated to 10 mg·day(-1)) in 24 women with lymphangioleiomyomatosis. Primary endpoints were safety, pharmacokinetics and serum vascular endothelial growth factor-D (VEGF-D) levels; secondary endpoints were measures of lung function.Following 26 weeks of everolimus treatment, forced vital capacity exhibited stability, while forced expiration volume in 1 s improved from baseline, with mean changes (95% confidence interval) of 10 mL (-111-132) and 114 mL (11-217), respectively; 6-min walk distance improved by 47 m. Median VEGF-D and collagen IV levels decreased from baseline, from 1730 pg·mL(-1) to 934.5 pg·mL(-1), and 103 ng·mL(-1) to 80.5 ng·mL(-1), respectively. Adverse events were mostly grade 1-2; mouth ulceration, headache, nausea, stomatitis and fatigue were common. Serious adverse events suspected to be treatment related included peripheral oedema, pneumonia, cardiac failure and Pneumocystis jirovecii infection. Everolimus blood levels increased dose proportionally.In this study, everolimus improved some measures of lung function and exercise capacity and reduced serum VEGF-D and collagen IV. Side effects were generally consistent with known toxicities of mTOR inhibitors, although some were severe.


Asunto(s)
Antineoplásicos/uso terapéutico , Everolimus/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Linfangioleiomiomatosis/tratamiento farmacológico , Factor D de Crecimiento Endotelial Vascular/efectos de los fármacos , Adolescente , Adulto , Anciano , Antineoplásicos/efectos adversos , Intervalos de Confianza , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Everolimus/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/patología , Dosis Máxima Tolerada , Persona de Mediana Edad , Medición de Riesgo , Resultado del Tratamiento , Factor D de Crecimiento Endotelial Vascular/sangre , Capacidad Vital/efectos de los fármacos , Capacidad Vital/fisiología , Adulto Joven
5.
J Natl Cancer Inst ; 106(8)2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25063326

RESUMEN

Myopericytoma (MPC) is a rare tumor with perivascular proliferation of pluripotent stem-cell-like pericytes. Although indolent, MPC may be locally aggressive with recurrent disease. The pathogenesis and diagnostic biomarkers of MPC are poorly understood. We discovered that 15% of benign MPCs (thyroid, skin; 3 of 20 samples) harbored BRAF(WT/V600E); 33.3% (1 of 3 samples) of BRAF(WT/V600E)-MPCs were multifocal/infiltrative/recurrent. Patient-MPC and primary MPC cells harbored BRAF(WT/V600E), were clonal and expressed pericytic-differentiation biomarkers crucial for its microenvironment. BRAF(WT/V600E)-positive thyroid MPC primary cells triggered in vitro (8.8-fold increase) and in vivo (3.6-fold increase) angiogenesis. Anti-BRAF(V600E) therapy with vemurafenib disrupted angiogenic and metabolic properties (~3-fold decrease) with down-regulation (~2.2-fold decrease) of some extracellular-matrix (ECM) factors and ECM-associated long non-coding RNA (LincRNA) expression, with no effects in BRAF(WT)-pericytes. Vemurafenib also inhibited (~3-fold decrease) cell viability in vitro and in BRAF(WT/V600E)-positive thyroid MPC patient-derived xenograft (PDX) mice (n = 5 mice per group). We established the first BRAF(WT/V600E)-dependent thyroid MPC cell culture. Our findings identify BRAF(WT/V600E) as a novel genetic aberration in MPC pathogenesis and MPC-associated biomarkers and imply that anti-BRAF(V600E) agents may be useful adjuvant therapy in BRAF(WT/V600E)-MPC patients. Patients with BRAF(WT/V600E)-MPC should be closely followed because of the risk for multifocality/recurrence.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Biomarcadores de Tumor/genética , Hemangiopericitoma/patología , Indoles/farmacología , Mutación , Pericitos/patología , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/farmacología , Neoplasias de la Tiroides/patología , Línea Celular Tumoral , Proliferación Celular , Genotipo , Ácido Glutámico , Hemangiopericitoma/genética , Humanos , Espectrometría de Masas , Recurrencia Local de Neoplasia/genética , Neoplasias de la Tiroides/genética , Valina , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Exp Med ; 211(1): 15-28, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24395886

RESUMEN

Lymphangioleiomyomatosis (LAM) is a progressive neoplastic disorder that leads to lung destruction and respiratory failure primarily in women. LAM is typically caused by tuberous sclerosis complex 2 (TSC2) mutations resulting in mTORC1 activation in proliferative smooth muscle-like cells in the lung. The female predominance of LAM suggests that estradiol contributes to disease development. Metabolomic profiling identified an estradiol-enhanced prostaglandin biosynthesis signature in Tsc2-deficient (TSC(-)) cells, both in vitro and in vivo. Estradiol increased the expression of cyclooxygenase-2 (COX-2), a rate-limiting enzyme in prostaglandin biosynthesis, which was also increased at baseline in TSC-deficient cells and was not affected by rapamycin treatment. However, both Torin 1 treatment and Rictor knockdown led to reduced COX-2 expression and phospho-Akt-S473. Prostaglandin production was also increased in TSC-deficient cells. In preclinical models, both Celecoxib and aspirin reduced tumor development. LAM patients had significantly higher serum prostaglandin levels than healthy women. 15-epi-lipoxin-A4 was identified in exhaled breath condensate from LAM subjects and was increased by aspirin treatment, indicative of functional COX-2 expression in the LAM airway. In vitro, 15-epi-lipoxin-A4 reduced the proliferation of LAM patient-derived cells in a dose-dependent manner. Targeting COX-2 and prostaglandin pathways may have therapeutic value in LAM and TSC-related diseases, and possibly in other conditions associated with mTOR hyperactivation.


Asunto(s)
Carcinogénesis/metabolismo , Estradiol/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Linfangioleiomiomatosis/metabolismo , Complejos Multiproteicos/metabolismo , Prostaglandinas/biosíntesis , Serina-Treonina Quinasas TOR/metabolismo , Análisis de Varianza , Animales , Aspirina/farmacología , Pruebas Respiratorias , Proteínas Portadoras/genética , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Inmunohistoquímica , Lipoxinas/análisis , Diana Mecanicista del Complejo 2 de la Rapamicina , Metabolómica , Ratones , Ratones SCID , Microscopía Confocal , Naftiridinas/farmacología , Prostaglandinas/sangre , Proteína Asociada al mTOR Insensible a la Rapamicina , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia
7.
Proc Natl Acad Sci U S A ; 110(37): 14960-5, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23983265

RESUMEN

Lymphangioleiomyomatosis (LAM) is a destructive lung disease of women associated with the metastasis of tuberin-null cells with hyperactive mammalian target of rapamycin complex 1 (mTORC1) activity. Clinical trials with the mTORC1 inhibitor rapamycin have revealed partial efficacy but are not curative. Pregnancy appears to exacerbate LAM, suggesting that estrogen (E2) may play a role in the unique features of LAM. Using a LAM patient-derived cell line (bearing biallelic Tuberin inactivation), we demonstrate that E2 stimulates a robust and biphasic activation of ERK2 and transcription of the late response-gene Fra1 associated with epithelial-to-mesenchymal transition. In a carefully orchestrated collaboration, activated mTORC1/S6K1 signaling enhances the efficiency of Fra1 translation of Fra1 mRNA transcribed by the E2-ERK2 pathway, through the phosphorylation of the S6K1-dependent eukaryotic translation initiation factor 4B. Our results indicate that targeting the E2-ERK pathway in combination with the mTORC1 pathway may be an effective combination therapy for LAM.


Asunto(s)
Estradiol/metabolismo , Linfangioleiomiomatosis/etiología , Linfangioleiomiomatosis/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Factores Eucarióticos de Iniciación/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Linfangioleiomiomatosis/genética , Sistema de Señalización de MAP Quinasas , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Invasividad Neoplásica , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
9.
Autophagy ; 7(11): 1400-1, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21997371

RESUMEN

Mammalian target of rapamycin (mTOR) complex 1 (mTORC1), which is activated in tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), is a master regulator of cell growth, cellular metabolism, and autophagy. Treatment of TSC and LAM patients with mTORC1 inhibitors partially decreases the size of brain and kidney tumors, and stabilizes pulmonary function. However, the tumors regrow and lung function continues to decline when treatment is discontinued. We hypothesized that dysregulation of autophagy plays a critical role in the pathogenesis of tumors with mTORC1 hyperactivation and in their response to mTORC1-targeted therapy. We found that cells lacking TSC2 have low levels of autophagy under basal and cellular stress conditions. Using genetic and pharmacological approaches, we discovered that the survival of Tsc2-deficient tumor cells is dependent on autophagy induction. Thus, autophagy inhibitors may have therapeutic potential in TSC and LAM, either as single agent therapy or in combination with mTORC1 inhibitors.


Asunto(s)
Autofagia , Transformación Celular Neoplásica/patología , Linfangioleiomiomatosis/patología , Esclerosis Tuberosa/patología , Animales , Transformación Celular Neoplásica/metabolismo , Humanos , Linfangioleiomiomatosis/metabolismo , Ratones , Factores de Transcripción/metabolismo , Investigación Biomédica Traslacional , Esclerosis Tuberosa/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(30): 12455-60, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21746920

RESUMEN

Tuberous sclerosis complex (TSC) is a tumor suppressor syndrome characterized by benign tumors in multiple organs, including the brain and kidney. TSC-associated tumors exhibit hyperactivation of mammalian target of rapamycin complex 1 (mTORC1), a direct inhibitor of autophagy. Autophagy can either promote or inhibit tumorigenesis, depending on the cellular context. The role of autophagy in the pathogenesis and treatment of the multisystem manifestations of TSC is unknown. We found that the combination of mTORC1 and autophagy inhibition was more effective than either treatment alone in inhibiting the survival of tuberin (TSC2)-null cells, growth of TSC2-null xenograft tumors, and development of spontaneous renal tumors in Tsc2(+/-) mice. Down-regulation of Atg5 induced extensive central necrosis in TSC2-null xenograft tumors, and loss of one allele of Beclin1 almost completely blocked macroscopic renal tumor formation in Tsc2(+/-) mice. Surprisingly, given the finding that lowering autophagy blocks TSC tumorigenesis, genetic down-regulation of p62/sequestosome 1 (SQSTM1), the autophagy substrate that accumulates in TSC tumors as a consequence of low autophagy levels, strongly inhibited the growth of TSC2-null xenograft tumors. These data demonstrate that autophagy is a critical component of TSC tumorigenesis, suggest that mTORC1 inhibitors may have autophagy-dependent prosurvival effects in TSC, and reveal two distinct therapeutic targets for TSC: autophagy and the autophagy target p62/SQSTM1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Choque Térmico/metabolismo , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Autofagia/fisiología , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Genes p53 , Proteínas de Choque Térmico/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Complejos Multiproteicos , Proteínas/genética , Proteínas/metabolismo , Proteína Sequestosoma-1 , Serina-Treonina Quinasas TOR , Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
11.
EMBO Mol Med ; 3(4): 189-200, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21412983

RESUMEN

The protein products of the tuberous sclerosis complex (TSC) genes, TSC1 and TSC2, form a complex, which inhibits the small G-protein, Ras homolog enriched in brain (Rheb). The vast majority of research regarding these proteins has focused on mammalian Target of Rapamycin (mTOR), a target of Rheb. Here, we propose that there are clinically relevant functions and targets of TSC1, TSC2 and Rheb, which are independent of mTOR. We present evidence that such non-canonical functions of the TSC-Rheb signalling network exist, propose a standard of evidence for these non-canonical functions, and discuss their potential clinical and therapeutic implications for patients with TSC and lymphangioleiomyomatosis (LAM).


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Transducción de Señal , Esclerosis Tuberosa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Neuropéptidos/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
12.
Lymphat Res Biol ; 8(1): 43-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20235886

RESUMEN

Research interest in lymphangioleiomyomatosis (LAM) has grown dramatically in the past decade, particularly among cancer biologists. There are at least two reasons for this: first, the discovery in the year 2000 that LAM cells carry TSC2 gene mutations, linking LAM with cellular pathways including the PI3K/Akt/mTOR axis, and allowing the Tuberous Sclerosis Complex (TSC)-regulated pathways that are believed to underlie LAM pathogenesis to be studied in cells, yeast, Drosophila, and mice. A second reason for the rising interest in LAM is the discovery that LAM cells can travel to the lung, including repopulating a donor lung after lung transplantation, despite the fact that LAM cells are histologically benign. This "benign metastasis" underpinning suggests that elucidating LAM pathogenesis will unlock a set of fundamental mechanisms that underlie metastatic potential in the context of a cell that has not yet undergone malignant transformation. Here, we will outline the data supporting the metastatic model of LAM, consider the biochemical and cellular mechanisms that may enable LAM cells to metastasize, including both cell autonomous and non-cell autonomous factors, and highlight a mouse model in which estrogen promotes the metastasis and survival of TSC2-deficient cells in a MEK-dependent manner. We propose a multistep model of LAM cell metastasis that highlights multiple opportunities for therapeutic intervention. Taken together, the metastatic behavior of LAM cells and the involvement of tumor-related signaling pathways lead to optimism that cancer-related paradigms for diagnosis, staging, and therapy will lead to therapeutic breakthroughs for women living with LAM.


Asunto(s)
Estrógenos/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfangiogénesis , Linfangioleiomiomatosis/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/metabolismo , Ratones , Metástasis de la Neoplasia , Serina-Treonina Quinasas TOR , Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
13.
Proc Am Thorac Soc ; 7(1): 48-53, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20160148

RESUMEN

The pace of progress in lymphangioleiomyomatosis (LAM) is remarkable. In the year 2000, TSC2 gene mutations were found in LAM cells; in 2001 the tuberous sclerosis complex (TSC) genes were discovered to regulate cell size in Drosophila via the kinase TOR (target of rapamycin); and in 2008 the results were published of a clinical trial of rapamycin, a specific inhibitor of TOR, in patients with TSC and LAM with renal angiomyolipomas. This interval of just 8 years between a genetic discovery for which the relevant signaling pathway was as yet unknown, to the initiation, completion, and publication of a clinical trial, is an almost unparalleled accomplishment in modern biomedical research. This robust foundation of basic, translational, and clinical research in TOR, TSC, and LAM is now poised to optimize and validate effective therapeutic strategies for LAM. An immediate challenge is to deduce the mechanisms underlying the partial response of renal angiomyolipomas to rapamycin, and thereby guide the design of combinatorial approaches. TOR complex 1 (TORC1), which is known to be active in LAM cells, is a key inhibitor of autophagy. One hypothesis, which will be explored here, is that low levels of autophagy in TSC2-null LAM cells limits their survival under conditions of bioenergetic stress. A corollary of this hypothesis is that rapamycin, by inducing autophagy, promotes the survival of LAM cells, while simultaneously arresting their growth. If this hypothesis proves to be correct, then combining TORC1 inhibition with autophagy inhibition may represent an effective clinical strategy for LAM.


Asunto(s)
Angiomiolipoma/tratamiento farmacológico , Autofagia/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Proteínas Serina-Treonina Quinasas/fisiología , Sirolimus/farmacología , Esclerosis Tuberosa/genética , Angiomiolipoma/genética , Angiomiolipoma/patología , Animales , Ensayos Clínicos como Asunto , Femenino , Humanos , Linfangioleiomiomatosis/patología , Pruebas de Función Respiratoria , Transducción de Señal , Serina-Treonina Quinasas TOR , Esclerosis Tuberosa/tratamiento farmacológico , Esclerosis Tuberosa/patología , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
14.
J Clin Invest ; 120(1): 93-102, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20038815

RESUMEN

Mutations in either of the genes encoding the tuberous sclerosis complex (TSC), TSC1 and TSC2, result in a multisystem tumor disorder characterized by lesions with unusual lineage expression patterns. How these unusual cell-fate determination patterns are generated is unclear. We therefore investigated the role of the TSC in the Drosophila external sensory organ (ESO), a classic model of asymmetric cell division. In normal development, the sensory organ precursor cell divides asymmetrically through differential regulation of Notch signaling to produce a pIIa and a pIIb cell. We report here that inactivation of Tsc1 and overexpression of the Ras homolog Rheb each resulted in duplication of the bristle and socket cells, progeny of the pIIa cell, and loss of the neuronal cell, a product of pIIb cell division. Live imaging of ESO development revealed this cell-fate switch occurred at the pIIa-pIIb 2-cell stage. In human angiomyolipomas, benign renal neoplasms often found in tuberous sclerosis patients, we found evidence of Notch receptor cleavage and Notch target gene activation. Further, an angiomyolipoma-derived cell line carrying biallelic TSC2 mutations exhibited TSC2- and Rheb-dependent Notch activation. Finally, inhibition of Notch signaling using a gamma-secretase inhibitor suppressed proliferation of Tsc2-null rat cells in a xenograft model. Together, these data indicate that the TSC and Rheb regulate Notch-dependent cell-fate decision in Drosophila and Notch activity in mammalian cells and that Notch dysregulation may underlie some of the distinctive clinical and pathologic features of TSC.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Drosophila/fisiología , Proteínas de Unión al GTP Monoméricas/fisiología , Neuropéptidos/fisiología , Receptores Notch/fisiología , Órganos de los Sentidos/embriología , Transducción de Señal/fisiología , Angiomiolipoma/metabolismo , Animales , Evolución Biológica , Drosophila , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Riñón/metabolismo , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones SCID , Proteína Homóloga de Ras Enriquecida en el Cerebro , Ratas , Esclerosis Tuberosa/etiología
15.
EMBO J ; 26(23): 4812-23, 2007 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17962806

RESUMEN

Miscoordination of growth and proliferation with the cellular stress response can lead to tumorigenesis. Mammalian target of rapamycin (mTOR), a central cell growth controller, is highly activated in some malignant neoplasms, and its clinical implications are under extensive investigation. We show that constitutive mTOR activity amplifies p53 activation, in vitro and in vivo, by stimulating p53 translation. Thus, loss of TSC1 or TSC2, the negative regulators of mTOR, results in dramatic accumulation of p53 and apoptosis in response to stress conditions. In other words, the inactivation of mTOR prevents cell death by nutrient stress and genomic damage via p53. Consistently, we also show that p53 is elevated in TSC tumors, which rarely become malignant. The coordinated relationship between mTOR and p53 during cellular stress provides a possible explanation for the benign nature of hamartoma syndromes, including TSC. Clinically, this also suggests that the efficacy of mTOR inhibitors in anti-neoplastic therapy may also depend on p53 status, and mTOR inhibitors may antagonize the effects of genotoxic chemotherapeutics.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Genes p53 , Proteínas Quinasas/genética , Proteínas Quinasas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/genética , Animales , Daño del ADN , Privación de Alimentos , Hamartoma/genética , Hamartoma/patología , Ratones , Mutágenos/química , Mutágenos/metabolismo , Fosforilación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa
16.
J Biol Chem ; 282(34): 24583-90, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17556368

RESUMEN

Birt-Hogg-Dube (BHD) is a tumor suppressor gene disorder characterized by skin hamartomas, cystic lung disease, and renal cell carcinoma. The fact that hamartomas, lung cysts, and renal cell carcinoma can also occur in tuberous sclerosis complex (TSC) suggests that the BHD and TSC proteins may function within a common pathway. To evaluate this hypothesis, we deleted the BHD homolog in Schizosaccharomyces pombe. Expression profiling revealed that six permease and transporter genes, known to be down-regulated in Deltatsc1 and Deltatsc2, were up-regulated in Deltabhd, and levels of specific intracellular amino acids known to be low in Deltatsc1 and Deltatsc2 were elevated in Deltabhd. This "opposite" profile was unexpected, given the overlapping clinical phenotypes. The TSC1/2 proteins inhibit Rheb in mammals, and Tsc1/Tsc2 inhibit Rhb1 in S. pombe. Expression of a hypomorphic allele of rhb1(+) dramatically increased permease expression levels in Deltabhd but not in wild-type yeast. Loss of Bhd sensitized yeast to rapamycin-induced increases in permease expression levels, and rapamycin induced lethality in Deltabhd yeast expressing the hypomorphic Rhb1 allele. In S. pombe, it is known that Rhb1 binds Tor2, and Tor2 inhibition leads to up-regulation of permeases including those that are regulated by Bhd. Our data, therefore, suggest that Bhd activates Tor2. If the mammalian BHD protein, folliculin, similarly activates mammalian target of rapamycin, it will be of great interest to determine how mammalian target of rapamycin inhibition in BHD patients and mammalian target of rapamycin activation in TSC patients lead to overlapping clinical phenotypes.


Asunto(s)
Regulación Fúngica de la Expresión Génica , Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/metabolismo , Proteínas Supresoras de Tumor/genética , Alelos , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Canavanina/farmacología , Etionina/farmacología , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Fenotipo , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/fisiología , Homología de Secuencia de Aminoácido , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
18.
Cancer Res ; 66(19): 9461-6, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17018601

RESUMEN

Inhibitors of the mammalian target of rapamycin (mTOR) are currently in clinical trials for the treatment of breast cancer. The mechanisms through which mTOR are activated in breast cancer and the relationship of mTOR activation to steroid hormones, such as estrogen, that are known to influence breast cancer pathogenesis, are not yet understood. Using MCF-7 cells as a model, we found that 17-beta estradiol (E(2)) rapidly increased the phosphorylation of downstream targets of mTOR: p70 ribosomal protein S6 kinase, ribosomal protein S6, and eukaryotic initiation factor 4E-binding protein 1. The phosphoinositide-3-kinase inhibitor, wortmannin, and the mTOR inhibitor, rapamycin, blocked E(2)-induced activation of p70 ribosomal protein S6 kinase. We hypothesized that tuberin and the small GTPase Ras homologue enriched in brain (Rheb), regulators of the mTOR pathway, mediate E(2)-induced activation of mTOR. Consistent with this hypothesis, E(2) rapidly (within 5 minutes) stimulated tuberin phosphorylation at T1462, a site at which Akt phosphorylates and inactivates tuberin. E(2) also rapidly decreased the inactive, GDP-bound form of Rheb. Finally, we found that small interfering RNA down-regulation of endogenous Rheb blocked the E(2)-stimulated proliferation of MCF-7 cells, demonstrating that Rheb is a key determinant of E(2)-dependent cell growth. Taken together, these data reveal that the TSC/Rheb/mTOR pathway plays a critical role in the regulation of E(2)-induced proliferation, and highlight Rheb as a novel molecular target for breast cancer therapy.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Estrógenos , Proteínas de Unión al GTP Monoméricas/fisiología , Neoplasias Hormono-Dependientes/metabolismo , Neuropéptidos/fisiología , Proteínas Quinasas/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/patología , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , División Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Estradiol/farmacología , Femenino , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neoplasias Hormono-Dependientes/patología , Neuropéptidos/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/farmacología , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR , Proteína 2 del Complejo de la Esclerosis Tuberosa
20.
Methods Mol Med ; 126: 185-96, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16930013

RESUMEN

Tuberous sclerosis complex (TSC) is a neurocutaneous syndrome characterized by seizures, mental retardation, and benign tumors of many organs, including the brain, kidneys, skin, retina, and heart. TSC is caused by mutations in the TSC1 and TSC2 tumor suppressor genes. The genes follow the two-hit model for tumorigenesis, with germline mutations inactivating one allele and somatic mutations inactivating the remaining wild-type allele. Allelic loss (also called loss of heterozygosity [LOH]) in the 9q34 and 16p13 regions has been found in many tumor types from TSC patients. Cardiac rhabdomyomas are frequently found in infants with TSC. Because rhabdomyomas often spontaneously regress, access to fresh tissue is limited. In this chapter, we present methodology for detection of genetic inactivation of TSC1 and TSC2 in paraffin-embedded archival tissues. The template DNA is obtained either by direct scraping of tissue or after laser capture microdissection. LOH analysis is performed after polymerase chain reaction amplification of microsatellite markers in the 9q34 and 16p13 regions and denaturing polyacrylamide gel electrophoresis. Mutation detection is performed using single-strand conformation polymorphisms on mutation detection enhancement gels. Finally, variant bands are amplified and analyzed by direct sequencing.


Asunto(s)
Análisis Mutacional de ADN/métodos , Genes Supresores de Tumor , Mutación/genética , Bancos de Tejidos , ADN/genética , ADN/aislamiento & purificación , Humanos , Lactante , Pérdida de Heterocigocidad/genética , Repeticiones de Microsatélite/genética , Adhesión en Parafina , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...