Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
AIDS Res Ther ; 11(1): 33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25320633

RESUMEN

BACKGROUND: Ongoing HIV-1 replication in lymphoid cells is one explanation of the persistence of HIV-1 reservoirs despite highly active antiretroviral therapy (cART). We tested the potential of cART intensification by Maraviroc plus Raltegravir to decrease proviral HIV-1 DNA levels in lymphoid cells during a randomized trial. PATIENTS AND METHODS: We randomly assigned for 48 weeks 22 patients to continue their current first line regimen of Truvada® plus Kaletra® or intensify it with Maraviroc and Raltegravir. The primary objective was to obtain a 50% decrease in proviral HIV-1 DNA levels in lymphoid cells with intensification. Blood samples were drawn at W-2, W0, W2, W4, W12, W24 and W48. Plasma viremia, cellular proviral DNA and cellular RNA, 2-LTR circles and lymphocytes subsets were assayed using validated methods. Patients in the intensified group underwent a gut biopsy at baseline and W48 to measure proviral DNA levels. Statistical analysis used parametric and non-parametric tests. RESULTS: Ten patients in each arm completed the trial. The 2 populations were comparable at baseline. No change in the reservoir size was observed in the intensified arm compared to the control arm measured in peripheral blood mononuclear cells (PBMCs). No change in the reservoir size was observed in gut proviral DNA in the intensified arm. In this group, no increase in 2-LTR circles was observed as early as 2 weeks after intensification and no change was found in residual plasma RNA levels measured by the single copy assay. However, a decrease in CD8(+) T cells activation was observed at 24 and 48 weeks, as well as in PBMCs HIV-1 RNA levels. CONCLUSION: We conclude that the intensification of a Protease Inhibitor regimen with Maraviroc and Raltegravir does not impact the blood proviral DNA reservoir of HIV but can decrease the cell-associated HIV RNA, the CD8 activation and has a possible impact on rectal proviral HIV DNA in some patients. TRIAL REGISTRATION: ClinicalTrials.gov identifier number NCT00935480.

3.
HIV Clin Trials ; 10(5): 324-7, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19906625

RESUMEN

Gut-associated lymphoid tissue is a huge reservoir for HIV-1. Developing new strategies to target "residual" HIV-1 in patients on effective therapy brings the need for an evaluation of tissue reservoirs in the clinic. We measured cell-associated HIV-1 RNA and DNA in blood and rectal biopsies from 23 patients, including 14 with undetectable viremia on HAART, by using an adaptation of commercially available tests. Rectal cell HIV-1 RNA was detected in all viremic patients, with median levels of 4.90 log(10) copies/million CD4. Although plasma viremia was found at a median of 3 copies/mL in "aviremic" patients, rectal cell HIV-1 RNA was detected in 28.5% with median levels of 5.17 log(10) copies/million CD4. Consequently, we propose to use this marker in future clinical trials targeting "residual" HIV-1 in patients with viremia below the detection limit.


Asunto(s)
Biomarcadores , Infecciones por VIH/virología , VIH-1/genética , Tejido Linfoide/virología , ARN Viral/aislamiento & purificación , Recto/virología , Terapia Antirretroviral Altamente Activa , ADN Viral/sangre , ADN Viral/aislamiento & purificación , Reservorios de Enfermedades , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Humanos , Laboratorios de Hospital , Límite de Detección , Persona de Mediana Edad , Proyectos Piloto , ARN Viral/sangre , Recto/citología
4.
Clin Cancer Res ; 13(3): 844-50, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17289876

RESUMEN

PURPOSE: Allelic losses [loss of heterozygosity (LOH)] at the 17p13 locus are frequent (85%) in adrenocortical cancers. The tumor suppressor gene TP53 is located at 17p13. The aim of the study was to determine the frequency of TP53 somatic inactivating mutations in adrenocortical tumors with 17p13 LOH and their clinico-biological correlations. EXPERIMENTAL DESIGN: TP53 somatic mutations, intragenic LOH (VNTR1 marker), and p53 overexpression were studied in 36 adrenocortical tumors with 17p13 LOH determined by Southern blot. RESULTS: TP53 mutations were detected in 33% of the tumors, and VNTR1 LOH was present in 44% of the cases and did not always correlate with the presence of a TP53 mutation. Only the TP53-mutant tumors exhibit a strong nuclear immunoreactivity. TP53-mutant tumors were significantly larger than wild-type TP53 tumors (median tumor weight: 640 versus 185 g; P=0.02), were associated with a more advanced stage of tumor progression (MacFarlane stage IV; P=0.01), and had a shorter disease-free survival (P=0.03). CONCLUSIONS: The finding that only a minority of adrenocortical tumors with 17p13 LOH had either a VNTR1 LOH or a TP53 mutation indicates that TP53 might not be the only or major tumor suppressor gene at 17p13 involved in adrenocortical cancer progression. We suggest that a genetic instability of the 17p13 region, occurring early in adrenocortical cancer development, involves various genes located in this region. TP53 might be only one of them, and its alteration by the occurrence of inactivating mutation is associated with the development of more aggressive tumors.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Cromosomas Humanos Par 17 , Genes p53 , Pérdida de Heterocigocidad , Repeticiones de Minisatélite/genética , Mutación , Adulto , Alelos , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , ARN Neoplásico/química
5.
J Clin Endocrinol Metab ; 91(5): 1943-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16464939

RESUMEN

CONTEXT: Primary pigmented nodular adrenocortical disease (PPNAD), a rare cause of corticotropin-independent Cushing syndrome, can be part of Carney complex (CNC), an autosomal dominant multiple neoplasia syndrome characterized by spotty skin pigmentation, cardiac myxomas, and endocrine tumors or be isolated (i). Germline PRKAR1A-inactivating mutations have been observed in both CNC and iPPNAD, but with no apparent genotype-phenotype correlation. OBJECTIVE: The objectives of the study were a detailed phenotyping for CNC manifestations in 12 kindreds bearing the same PRKAR1A mutation and a study of the consequences of the mutation and a potential founder effect. DESIGN: The study consisted of descriptive case reports. SETTING: The study was conducted at two referral centers. PATIENTS: The patients described in this study were referred for PRKAR1A gene mutation analysis because of a diagnosis of apparently iPPNAD. RESULTS: We describe a 6-bp polypyrimidine tract deletion [exon 7 IVS del (-7-->-2)] in 12 unrelated kindreds that were referred for Cushing syndrome due to PPNAD. Nine of the patients had no family history; in two, there was a family history of iPPNAD. Only one patient met the criteria for CNC. Relatives carrying the same mutation had no manifestations of CNC or PPNAD, suggesting a low penetrance of this PRKAR1A defect. A founder effect was excluded by extensive genotyping of chromosome 17 markers. CONCLUSIONS: In conclusion, a small intronic deletion of the PRKAR1A gene is a low-penetrance cause of mainly iPPNAD; it is the first PRKAR1A genetic defect to have an association with a specific phenotype.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Mutación/genética , Adolescente , Adulto , Células Cultivadas , Síndrome de Cushing/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Cicloheximida/farmacología , ADN/biosíntesis , ADN/genética , Femenino , Efecto Fundador , Genotipo , Haplotipos , Humanos , Intrones/genética , Masculino , Mutación/fisiología , Linaje , Penetrancia , Fenotipo , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética
6.
Horm Res ; 64(3): 132-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16192737

RESUMEN

The Carney complex (CNC) is a dominantly inherited syndrome responsible mainly for spotty skin pigmentation (lentiginosis), endocrine overactivity, and cardiac myxomas. Adrenocorticotropic hormone independent Cushing's syndrome due to primary pigmented nodular adrenocortical disease (PPNAD) is a main characteristic of CNC. PPNAD is a very rare cause of Cushing's syndrome due to a primary bilateral adrenal defect that can be also observed in some patients without other CNC manifestations nor familial history. One of the putative CNC genes, located on 17q22-24, has been identified as the regulatory subunit R1A of protein kinase A (PRKAR1A). Heterozygous inactivating mutations of PRKAR1A have been reported initially in about 45% of the CNC index cases and could be found in about 80% of the CNC families presenting mainly with Cushing's syndrome. PRKAR1A is a key component of the cyclic AMP signaling pathway that has been implicated in endocrine tumorigenesis and could, at least partly, function as a tumor suppressor gene. Interestingly, patients with isolated PPNAD and no familial history of CNC can also present a germline de novo mutation of PRKAR1A. Somatic mutations of PRKAR1A have been found in PPNAD as a mechanism of inactivation of the wild-type allele, in a patient already presenting a germline mutation, and in a subset of sporadic secreting adrenocortical adenomas with clinical, hormonal, and pathological features quite similar to PPNAD. This review will summarize the recent findings on CNC from the perspective of the pathophysiology of adrenal Cushing's syndrome and PPNAD.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/fisiopatología , Glándulas Suprarrenales/fisiopatología , Neoplasia Endocrina Múltiple/fisiopatología , Enfermedades de la Corteza Suprarrenal/genética , Adulto , Preescolar , AMP Cíclico/fisiología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico , Humanos , Neoplasia Endocrina Múltiple/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/etiología , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/fisiopatología , Proteínas/genética , Transducción de Señal
7.
Cancer Res ; 65(17): 7622-7, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16140927

RESUMEN

Adrenocortical cancer is a rare cancer with a very poor prognosis. The genetic alterations identified to date in adrenocortical tumors are limited. Activating mutations of the Wnt signaling pathway have been observed in more frequent cancers, particularly digestive tract tumors. We investigated whether Wnt pathway activation is involved in adrenocortical tumorigenesis. In a series of 39 adrenocortical tumors, immunohistochemistry revealed abnormal cytoplasmic and/or nuclear accumulation of beta-catenin in 10 of 26 adrenocortical adenomas and in 11 of 13 adrenocortical carcinomas. An activating somatic mutation of the beta-catenin gene was shown in 7 of 26 adrenocortical adenomas and in 4 of 13 adrenocortical carcinomas; these mutations were observed only in adrenocortical tumors with abnormal beta-catenin accumulation and most were point mutations altering the Ser45 of exon 3 (in the consensus GSK3-beta/CK1 phosphorylation site). Functional studies showed that the activating Ser45 beta-catenin mutation found in the adrenocortical cancer H295R cell line leads to constitutive activation of T-cell factor-dependent transcription. This is the first molecular defect to be reported with the same prevalence in both benign (27%) and malignant (31%) adrenocortical tumors. beta-Catenin mutations are also the most frequent genetic defect currently known in adrenocortical adenomas. In adrenocortical adenomas, beta-catenin alterations are more frequent in nonfunctioning tumors, suggesting that beta-catenin pathway activation might be mostly involved in the development of nonsecreting adrenocortical adenomas and adrenocortical carcinomas. The very frequent and substantial accumulation of beta-catenin in adrenocortical carcinomas suggests that other alterations might also be involved. This finding may contribute to new therapeutic approaches targeting the Wnt pathway in malignant adrenocortical tumors, for which limited medical therapy is available.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Proteínas del Citoesqueleto/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Transactivadores/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Adulto , Anciano , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación , Transducción de Señal , Transactivadores/metabolismo , Proteínas Wnt , beta Catenina
8.
Cancer Res ; 64(23): 8761-6, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15574788

RESUMEN

Cell-mediated immunity directed against human papillomavirus 16 (HPV-16) antigens was studied in six patients affected with grade 3 vulvar intraepithelial neoplasia (VIN3, also known as bowenoid papulosis). Five of the patients presented with a chronic and persistent disease that relapsed after destructive treatments. They showed no detectable anti-HPV blood T-cell responses and no T-cell intraepidermal vulvar infiltrate containing both CD4+ and CD8+ lymphocytes. The last patient had a complete clearance of viral lesions, 8 months after disease onset and 2 months after electrocoagulation of <50% of the VIN3 lesions. She showed high frequency anti-E6 and anti-E7 effector blood T cells by ex vivo ELISpot-IFNgamma assay before clinical regression. Immunohistochemical study of her vulvar biopsy revealed a marked dermal infiltrate containing a majority of CD4+ T lymphocytes and an epidermal infiltrate made up of both CD4(+) and CD8(+) T cells. This seems to be the first evidence of an association between spontaneous regression of VIN3 lesions and HPV-specific T-cell responses detectable in the blood. Hence, an increase of HPV-specific effector T lymphocyte responses by vaccine-based therapeutic strategies might be useful to clear the lesions in bowenoid papulosis disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Regresión Neoplásica Espontánea/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Neoplasias de la Vulva/inmunología , Neoplasias de la Vulva/virología , Adulto , Secuencia de Aminoácidos , Carcinoma in Situ/inmunología , Carcinoma in Situ/patología , Carcinoma in Situ/virología , Femenino , Humanos , Activación de Linfocitos/inmunología , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/complicaciones , Fragmentos de Péptidos/inmunología , Proteínas Represoras/inmunología , Neoplasias de la Vulva/patología
9.
Gastroenterology ; 125(2): 328-36, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12891533

RESUMEN

BACKGROUND & AIMS: Epidemiologic and experimental studies have suggested that aspirin intake reduces the risk for colorectal carcinogenesis. However, the available data are not sufficient to serve as the basis for firm recommendations. METHODS: We randomly assigned 272 patients with a history of colorectal adenomas (at least one more than 5 mm in diameter, or more than 3) to daily lysine acetylsalicylate (160 or 300 mg/day) or placebo for 4 years. The primary end points were adenoma recurrence after 1 and 4 years. These results are those of the year 1 colonoscopy. RESULTS: Among the 238 patients who completed the year 1 colonoscopy, at least one adenoma was observed in 38 patients of the 126 (30%) in the aspirin group and in 46 of the 112 (41%) in the placebo group; relative risk was 0.73 (95% confidence interval [CI]: 0.52-1.04; P = 0.08). At least one adenoma of more than 5 mm diameter was observed in 13 patients (10%) in the aspirin group and 26 (23%) in the placebo group (P = 0.01). The corresponding numbers for adenomas more than 10 mm in diameter were one (1%) and 7 (6%) (P = 0.05). Stepwise regression showed that independent factors associated with lower adenoma recurrence are aspirin treatment (adenoma >5 mm, P = 0.01), absence of personal history of adenoma before the entry colonoscopy (P = 0.01), and initial adenomatous polyp burden less than 10 mm (P = 0.001). CONCLUSIONS: Daily soluble aspirin is associated with a reduction in the risk for recurrent adenomas found at colonoscopy 1 year after starting treatment.


Asunto(s)
Adenoma/prevención & control , Aspirina/administración & dosificación , Neoplasias Colorrectales/prevención & control , Recurrencia Local de Neoplasia/prevención & control , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Cooperación del Paciente
10.
J Clin Endocrinol Metab ; 88(8): 3958-65, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12915693

RESUMEN

Various cellular and molecular alterations of the cAMP pathway have been observed in adrenal Cushing syndrome. We recently reported the loss of cAMP-responsive element-binding protein (CREB) expression in the adrenocortical cancer cell line H295R. CREB is the major nuclear target of the cAMP pathway. This study therefore aimed to analyze the status of the CREB protein in various types of human adrenocortical tumors and normal fetal adrenal cortex. CREB protein status was studied by Western blotting in adrenocortical adenomas (AAs, n = 27) and adrenocortical carcinomas (ACs, n = 24). A decrease of CREB protein was noticed in the majority of the adrenocortical tumors. The dramatic decrease in CREB protein levels was more pronounced in ACs than in AAs. Levels of the phosphorylated form of CREB were also low in adrenocortical tumors, with a greater decrease in ACs than in AAs. EMSAs also showed decreases in the amounts of CREB- containing complexes in nuclear extracts from adrenocortical tumors. The secretory status of adenomas was strongly correlated with CREB levels, significantly lower in nonfunctioning AAs (n = 9) than in functioning AAs (n = 9). CREB levels, determined by Western blotting and immunohistochemistry, were very low in the fetal zone of human fetal adrenal cortex, whereas they were normal in the definitive zone. In tumors, adrenocortical cells in several zones were weakly immunohistochemically stained for CREB, whereas CREB was uniformly detected in nonendocrine cell nuclei (e.g. vascular cells, fibroblasts). These results suggest that the absence of CREB may be linked to the development of a highly aggressive tumor with a dedifferentiated benign (nonfunctioning AA) or malignant (AC) phenotype. These findings highlight the similarities between the normal human fetal adrenal gland and adrenal cancers previously observed in terms of parallelism in IGF-II production.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/biosíntesis , Desarrollo Embrionario y Fetal/fisiología , Adolescente , Neoplasias de la Corteza Suprarrenal/patología , Adulto , Anciano , Western Blotting , AMP Cíclico/fisiología , Ensayo de Cambio de Movilidad Electroforética , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Embarazo , Células Tumorales Cultivadas
11.
Am J Hum Genet ; 71(6): 1433-42, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12424709

RESUMEN

We studied 11 new kindreds with primary pigmented nodular adrenocortical disease (PPNAD) or Carney complex (CNC) and found that 82% of the kindreds had PRKAR1A gene defects (including seven novel inactivating mutations), most of which led to nonsense mRNA and, thus, were not expressed in patients' cells. However, a previously undescribed base substitution in intron 6 (exon 6 IVS +1G-->T) led to exon 6 skipping and an expressed shorter PRKAR1A protein. The mutant protein was present in patients' leukocytes and tumors, and in vitro studies indicated that the mutant PRKAR1A activated cAMP-dependent protein kinase A (PKA) signaling at the nuclear level. This is the first demonstration of an inactivating PRKAR1A mutation being expressed at the protein level and leading to stimulation of the PKA pathway in CNC patients. Along with the lack of allelic loss at the PRKAR1A locus in most of the tumors from this kindred, these data suggest that alteration of PRKAR1A function (not only its complete loss) is sufficient for augmenting PKA activity leading to tumorigenesis in tissues affected by CNC.


Asunto(s)
Enfermedades de las Glándulas Suprarrenales/enzimología , Enfermedades de las Glándulas Suprarrenales/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neoplasia Endocrina Múltiple/enzimología , Neoplasia Endocrina Múltiple/genética , Mutación/genética , Transducción de Señal , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Secuencia de Bases , Células COS , Codón Iniciador/genética , Análisis Mutacional de ADN , Exones/genética , Femenino , Humanos , Intrones/genética , Leucocitos , Pérdida de Heterocigocidad , Masculino , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Linaje , Regiones Promotoras Genéticas/genética , Subunidades de Proteína , Sitios de Empalme de ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
12.
J Clin Endocrinol Metab ; 87(9): 4324-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213893

RESUMEN

Primary pigmented nodular adrenocortical disease (PPNAD) is a cause of ACTH-independent Cushing's syndrome. This condition can be difficult to diagnose because hypercortisolism may be periodic and adrenal imaging may not demonstrate an adrenal tumor. PPNAD can be part of the Carney complex (CNC), an autosomal dominant multiple neoplasia syndrome. Germline mutations of the regulatory subunit R1A of PKA (PRKAR1A) have been observed in about 45% of CNC kindreds. To improve our understanding of sporadic PPNAD and develop a potential diagnostic tool, we investigated the genetics of patients with sporadic and isolated PPNAD. Patients undergoing surgery for bilateral ACTH-independent Cushing's syndrome in whom pathological examination revealed PPNAD were subjected to endocrinological investigations and a systematic search for other manifestations of CNC. The PRKAR1A gene was sequenced using DNA from frozen adrenal tissues and leukocytes from three patients with sporadic isolated PPNAD and using leukocyte DNA from two additional patients. Different inactivating germline mutations of the PRKAR1A gene were found in the five patients. For three cases, study of the parents' DNA demonstrated a de novo mutation. One patient presented with an unusual 2.5-cm macronodule of the right adrenal mimicking an adrenal adenoma. A somatic 16-bp deletion of PRKAR1A gene was also found in this macronodule. Inactivating germline mutations of PRKAR1A are frequent in sporadic and isolated cases of PPNAD. The wild-type allele can be inactivated by somatic mutations, consistent with the hypothesis of the gene being a tumor suppressor gene. Thus, genetic analysis can be of help to the clinician in the diagnosis of this difficult form of adrenal Cushing's syndrome.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/genética , Síndrome de Cushing/genética , Mutación , Trastornos de la Pigmentación/genética , Adolescente , Enfermedades de la Corteza Suprarrenal/diagnóstico por imagen , Adulto , Niño , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Humanos , Hidrocortisona/sangre , Masculino , Estudios Retrospectivos , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...