Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(8): 1520-1533, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931053

RESUMEN

Germline PTEN variants (PTEN hamartoma tumor syndrome [PHTS]) confer up to 85% lifetime risk of female breast cancer (BC). BCs arising in PHTS are clinically distinct from sporadic BCs, including younger age of onset, multifocality, and an increased risk of second primary BCs. Yet, there is no previous investigation into the underlying genomic landscape of this entity. We sought to address the hypothesis that BCs arising in PHTS have a distinct genomic landscape compared to sporadic counterparts. We performed and analyzed exome sequencing data from 44 women with germline PTEN variants who developed BCs. The control cohort comprised of 497 women with sporadic BCs from The Cancer Genome Atlas (TCGA) dataset. We demonstrate that PHTS-derived BCs have a distinct somatic mutational landscape compared to the sporadic counterparts, namely second somatic hits in PTEN, distinct mutational signatures, and increased genomic instability. The PHTS group had a significantly higher frequency of somatic PTEN variants compared to TCGA (22.7% versus 5.6%; odds ratio [OR] 4.93; 95% confidence interval [CI] 2.21 to 10.98; p < 0.001) and a lower mutational frequency in PIK3CA (22.7% versus 33.4%; OR 0.59; 95% CI 0.28 to 1.22; p = 0.15). Somatic variants in PTEN and PIK3CA were mutually exclusive in PHTS (p = 0.01) but not in TCGA. Our findings have important implications for the personalized management of PTEN-related BCs, especially in the context of more accessible genetic testing.


Asunto(s)
Neoplasias de la Mama , Síndrome de Hamartoma Múltiple , Neoplasias de la Mama/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Exoma/genética , Femenino , Genómica , Células Germinativas/patología , Mutación de Línea Germinal/genética , Síndrome de Hamartoma Múltiple/genética , Humanos , Fosfohidrolasa PTEN/genética
2.
Gastroenterology ; 164(5): 800-811, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36717037

RESUMEN

The hamartomatous polyposis syndromes are a set of clinically distinct disorders characterized by the occurrence of hamartomatous polyps in the gastrointestinal tract. These syndromes include juvenile polyposis syndrome, Peutz-Jeghers syndrome, and PTEN hamartoma tumor syndrome. Although each of the syndromes has distinct phenotypes, the hamartomatous polyps can be challenging to differentiate histologically. Additionally, each of these syndromes is associated with increased lifetime risks of gene-specific and organ-specific cancers, including those outside of the gastrointestinal tract. Germline pathogenic variants can be identified in a subset of individuals with these syndromes, which facilitates molecular diagnosis and subsequent gene-enabled management in the setting of genetic counseling. Although the malignant potential of hamartomatous polyps remains elusive, timely recognition of these syndromes is important and enables presymptomatic cancer surveillance and management before symptom exacerbation. Presently, there are no standard agents to prevent the development of polyps and cancers in the hamartomatous polyposis syndromes.


Asunto(s)
Síndrome de Hamartoma Múltiple , Hamartoma , Síndromes Neoplásicos Hereditarios , Síndrome de Peutz-Jeghers , Humanos , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Hamartoma/diagnóstico , Hamartoma/genética , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/genética , Síndrome de Hamartoma Múltiple/diagnóstico , Síndrome de Hamartoma Múltiple/genética , Pólipos Intestinales
3.
Genet Med ; 26(7): 101128, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38829299

RESUMEN

PURPOSE: We previously described a combined risk score (CRS) that integrates a multiple-ancestry polygenic risk score (MA-PRS) with the Tyrer-Cuzick (TC) model to assess breast cancer (BC) risk. Here, we present a longitudinal validation of CRS in a real-world cohort. METHODS: This study included 130,058 patients referred for hereditary cancer genetic testing and negative for germline pathogenic variants in BC-associated genes. Data were obtained by linking genetic test results to medical claims (median follow-up 12.1 months). CRS calibration was evaluated by the ratio of observed to expected BCs. RESULTS: Three hundred forty BCs were observed over 148,349 patient-years. CRS was well-calibrated and demonstrated superior calibration compared with TC in high-risk deciles. MA-PRS alone had greater discriminatory accuracy than TC, and CRS had approximately 2-fold greater discriminatory accuracy than MA-PRS or TC. Among those classified as high risk by TC, 32.6% were low risk by CRS, and of those classified as low risk by TC, 4.3% were high risk by CRS. In cases where CRS and TC classifications disagreed, CRS was more accurate in predicting incident BC. CONCLUSION: CRS was well-calibrated and significantly improved BC risk stratification. Short-term follow-up suggests that clinical implementation of CRS should improve outcomes for patients of all ancestries through personalized risk-based screening and prevention.


Asunto(s)
Neoplasias de la Mama , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Herencia Multifactorial , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/diagnóstico , Medición de Riesgo/métodos , Herencia Multifactorial/genética , Persona de Mediana Edad , Adulto , Factores de Riesgo , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Anciano
4.
Hum Mol Genet ; 29(22): 3679-3690, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33326033

RESUMEN

Adrenocortical Carcinoma (ACC) is a rare endocrine tumor with poor overall prognosis and 1.5-fold overrepresentation in females. In children, ACC is associated with inherited cancer syndromes with 50-80% of childhood-ACC associated with TP53 germline variants. ACC in adolescents and young adults (AYA) is rarely due to germline TP53, IGF2, PRKAR1A and MEN1 variants. We analyzed exome sequencing data from 21 children (<15y), 32 AYA (15-39y), and 60 adults (>39y) with ACC, and retained all pathogenic, likely pathogenic, and highly prioritized variants of uncertain significance. We engineered a stable lentiviral-mutant ACC cell line, harboring an EGFR variant (p.Asp1080Asn) from a 21-year-old female without germline-TP53-variant and with aggressive ACC. We found that 4.8% of the children (P = 0.004) and 6.2% of AYA (P < 0.0001), all-female participants, harbored germline EGFR variants, compared to only 0.3% of the control group. Expanding our analysis to the RTK-RAS-MAPK pathway, we found that the RTK genes have the highest number of highly prioritized germline variants in these individuals amongst all three arms of this pathway. We showed EGFR mutant cells migrate faster and are characterized by a stem-like phenotype compared to wild type cells. While EGFR inhibitors did not affect the stemness of mutant cells, Sunitinib, a multireceptor tyrosine kinase inhibitor, significantly reduced their stem-like behavior. Our data suggest that EGFR could be a novel underlying germline predisposition factor for ACC, especially in the Childhood-AYA (C-AYA) population. Further clinical validation can improve precision oncology management of this disease, which is known to have limited therapeutic options.


Asunto(s)
Carcinoma Corticosuprarrenal/genética , Predisposición Genética a la Enfermedad , Proteína p53 Supresora de Tumor/genética , Adolescente , Carcinoma Corticosuprarrenal/patología , Adulto , Receptores ErbB/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genotipo , Mutación de Línea Germinal/genética , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Medicina de Precisión , Secuenciación del Exoma , Adulto Joven , Proteínas ras/genética
5.
N Engl J Med ; 382(22): 2103-2116, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32459922

RESUMEN

BACKGROUND: Patients with PTEN hamartoma tumor syndrome (PHTS) have germline mutations in the tumor-suppressor gene encoding phosphatase and tensin homologue (PTEN). Such mutations have been associated with a hereditary predisposition to multiple types of cancer, including the Cowden syndrome. However, a majority of patients who have PHTS-related phenotypes have tested negative for PTEN mutations. In a previous study, we found that the E3 ubiquitin ligase WWP1 negatively regulates the function of PTEN. METHODS: In a prospective cohort study conducted from 2005 through 2015, we enrolled 431 patients with wild-type PTEN who met at least the relaxed diagnostic criteria of the International Cowden Consortium. Patients were scanned for WWP1 germline variants. We used the Cancer Genome Atlas (TCGA) data set as representative of apparently sporadic cancers and the Exome Aggregation Consortium data set excluding TCGA (non-TCGA ExAC) and the noncancer Genome Aggregation Database (gnomAD) as representative of population controls without a reported cancer diagnosis. We established both in vitro and murine in vivo models to functionally characterize representative WWP1 variants. RESULTS: The existence of germline WWP1 variants was first established in a family with wild-type PTEN who had oligopolyposis and early-onset colon cancers. A validation series indicated that WWP1 germline variants occurred in 5 of 126 unrelated patients (4%) with oligopolyposis as a predominant phenotype. Germline WWP1 variants, particularly the WWP1 K740N and N745S alleles, were enriched in patients who did not have PHTS but had prevalent sporadic cancers, including PTEN-related cancer types in TCGA (odds ratio, 1.5; 95% confidence interval, 1.1 to 2.1; P = 0.01). The prioritized WWP1 variants resulted in gain-of-function effects, which led to aberrant enzymatic activation with consequent PTEN inactivation, thereby triggering hyperactive growth-promoting PI3K signaling in cellular and murine models. CONCLUSIONS: In this study involving patients with disorders resulting in a predisposition to the development of multiple malignant neoplasms without PTEN germline mutations, we confirmed the function of WWP1 as a cancer-susceptibility gene through direct aberrant regulation of the PTEN-PI3K signaling axis. (Funded by the National Institutes of Health and others.).


Asunto(s)
Mutación con Ganancia de Función , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Síndrome de Hamartoma Múltiple/genética , Fosfohidrolasa PTEN/genética , Ubiquitina-Proteína Ligasas/genética , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Variación Genética , Humanos , Masculino , Ratones , Ratones Mutantes , Linaje , Estudios Prospectivos
6.
J Am Acad Dermatol ; 89(1): 90-98, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35143913

RESUMEN

BACKGROUND: Dermatologic phenotypes in PTEN hamartoma tumor syndrome (PHTS) are heterogeneous and poorly documented. OBJECTIVE: To characterize dermatologic findings among PHTS and conduct an analysis of genotype-dermatologic phenotype associations. METHODS: Mucocutaneous findings were reviewed in a multicenter cohort study of PHTS. Genotype-dermatologic phenotype associations were tested using multivariable regression. RESULTS: A total of 201 patients were included. Children were significantly less likely than adults to have oral papillomas, vascular malformations, benign follicular neoplasms, and acral keratoses. There were no cases of skin cancer among children. Basal cell carcinoma, cutaneous squamous cell carcinoma, and melanoma developed in 5%, 2%, and 1% of White adults, respectively. After adjusting for age, missense mutations were associated with 60% lower odds of developing cutaneous papillomatous papules (odds ratio: 0.4; 95% confidence interval [0.2, 0.7]), oral papillomas (0.4; 95% confidence interval [0.2, 0.9]), and vascular malformations (0.4; 95% confidence interval [0.2, 0.8]). LIMITATIONS: Partly retrospective data. CONCLUSION: Children are less likely than adults to have certain dermatologic findings, likely due to age-related penetrance. The risk of pediatric melanoma and the lifetime risk of nonmelanoma skin cancer in PHTS may not be elevated. Missense variants may be associated with the development of fewer dermatologic findings but future validation is required.


Asunto(s)
Carcinoma de Células Escamosas , Síndrome de Hamartoma Múltiple , Melanoma , Papiloma , Neoplasias Cutáneas , Malformaciones Vasculares , Humanos , Síndrome de Hamartoma Múltiple/complicaciones , Síndrome de Hamartoma Múltiple/epidemiología , Síndrome de Hamartoma Múltiple/genética , Carcinoma de Células Escamosas/complicaciones , Estudios Retrospectivos , Estudios de Cohortes , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/complicaciones , Melanoma/complicaciones , Malformaciones Vasculares/complicaciones , Fosfohidrolasa PTEN/genética
7.
Hum Mol Genet ; 29(R2): R150-R157, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32568377

RESUMEN

Hereditary cancer syndromes represent ~10% of all incident cancers. It is important to identify individuals having these disorders because, unlike patients with sporadic cancer, these patients require specialised life-long care, with implications for their families. Importantly, the identification of alterations in cancer-predisposing genes facilitates gene-informed molecular diagnosis, cancer risk assessment and gene-specific clinical management. Moreover, knowledge about gene function in the inherited cancers offers insights towards biological processes pertinent to the more common sporadic cancers. Conversely, without a known gene, clinical management is less precise, and it is impossible to offer predictive testing of family members. PTEN hamartoma tumour syndrome (PHTS) is an umbrella term encompassing four overgrowth and cancer predisposition disorders associated with germline PTEN mutations. With time, it became evident that only a finite subset of individuals with PHTS-associated phenotypes harbour germline PTEN mutations. Therefore, non-PTEN aetiologies exist in PTEN wildtype patients. Indeed, gene discovery efforts over the last decade elucidated multiple candidate cancer predisposition genes. While a subset of genes (e.g. AKT1, PIK3CA) are biologically plausible as being key effectors within the PTEN signalling cascade, other genes required meticulous functional interrogation to explain their contribution to PHTS-related phenotypes. Collectively, the extensive phenotypic heterogeneity of the clinical syndromes typically united by PTEN is reflected by the genetic heterogeneity revealed through gene discovery. Validating these gene discoveries is critical because, while PTEN wildtype patients can be diagnosed clinically, they do not have the benefit of specific gene-informed risk assessment and subsequent management.


Asunto(s)
Biomarcadores de Tumor/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Síndrome de Hamartoma Múltiple/patología , Fosfohidrolasa PTEN/genética , Síndrome de Hamartoma Múltiple/genética , Humanos
8.
Annu Rev Med ; 71: 103-116, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31433956

RESUMEN

PTEN is a tumor suppressor gene that classically dampens the PI3K/AKT/mTOR growth-promoting signaling cascade. PTEN dysfunction causes dysregulation of this and other pathways, resulting in overgrowth. Cowden syndrome, a hereditary cancer predisposition and overgrowth disorder, was the first Mendelian condition associated with germline PTEN mutations. Since then, significant advances by the research and medical communities have elucidated how clinical phenotypic manifestations result from the underlying germline PTEN mutations. With time, it became evident that PTEN mutations can result in a broad phenotypic spectrum, causing seemingly disparate disorders from cancer to autism. Hence, the umbrella term of PTEN hamartoma tumor syndrome (PHTS) was coined. Timely diagnosis and understanding the natural history of PHTS are vital because early recognition enables gene-informed management, particularly as related to high-risk cancer surveillance and addressing the neurodevelopmental symptoms.


Asunto(s)
Detección Precoz del Cáncer/métodos , Predisposición Genética a la Enfermedad/genética , Mutación de Línea Germinal/genética , Neoplasias/genética , Femenino , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Incidencia , Masculino , Mutación/genética , Neoplasias/patología , Fosfohidrolasa PTEN , Fenotipo , Pronóstico , Medición de Riesgo
9.
Am J Hum Genet ; 105(4): 813-821, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564436

RESUMEN

Germline heterozygous PTEN mutations cause subsets of Cowden syndrome (CS) and Bannayan-Riley-Ruvalcaba syndrome (BRRS); these subsets are characterized by high risks of breast, thyroid, and other cancers and, in one subset, autism spectrum disorder (ASD). Up to 10% of individuals with PTENMUT CS, CS-like syndrome, or BRRS have germline SDHx (succinate dehydrogenase, mitochondrial complex II) variants, which modify cancer risk. PTEN contributes to metabolic reprogramming; this is a well-established role in a cancer context. Relatedly, SDH sits at the crossroad of the electron transport chain and tricarboxylic acid (TCA) cycle, two central bioenergetic pathways. Intriguingly, PTENMUT and SDHMUT individuals have reduced SDH catalytic activity, resulting in succinate accumulation; this indicates a common genotype-independent biochemical alteration. Here, we conducted a TCA targeted metabolomics study on 511 individuals with CS, CS-like syndrome, or BRRS with various genotypes (PTEN or SDHx, mutant or wild type [WT]) and phenotypes (cancer or ASD) and a series of 187 population controls. We found consistent TCA cycle metabolite alterations in cases with various genotypes and phenotypes compared to controls, and we found unique correlations of individual metabolites with particular genotype-phenotype combinations. Notably, increased isocitrate (p = 1.2 × 10-3), but reduced citrate (p = 5.0 × 10-4), were found to be associated with breast cancer in individuals with PTENMUT/SDHxWT. Conversely, increased lactate was associated with neurodevelopmental disorders regardless of genotype (p = 9.7 × 10-3); this finding was replicated in an independent validation series (n = 171) enriched for idiopathic ASD (PTENWT, p = 5.6 × 10-4). Importantly, we identified fumarate (p = 1.9 × 10-2) as a pertinent metabolite, distinguishing individuals who develop ASD from those who develop cancer. Our observations suggest that TCA cycle metabolite alterations are germane to the pathobiology of PTEN-related CS and BRRS, as well as genotype-independent ASD, with implications for potential biomarker and/or therapeutic value.


Asunto(s)
Trastorno Autístico/genética , Ciclo del Ácido Cítrico , Síndrome de Hamartoma Múltiple/genética , Neoplasias/genética , Fenotipo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
10.
PLoS Genet ; 14(4): e1007352, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29684080

RESUMEN

Patients with heritable cancer syndromes characterized by germline PTEN mutations (termed PTEN hamartoma tumor syndrome, PHTS) benefit from PTEN-enabled cancer risk assessment and clinical management. PTEN-wildtype patients (~50%) remain at increased risk of developing certain cancers. Existence of germline mutations in other known cancer susceptibility genes has not been explored in these patients, with implications for different medical management. We conducted a 4-year multicenter prospective study of incident patients with features of Cowden/Cowden-like (CS/CS-like) and Bannayan-Riley-Ruvalcaba syndromes (BRRS) without PTEN mutations. Exome sequencing and targeted analysis were performed including 59 clinically actionable genes from the American College of Medical Genetics and Genomics (ACMG) and 24 additional genes associated with inherited cancer syndromes. Pathogenic or likely pathogenic cancer susceptibility gene alterations were found in 7 of the 87 (8%) CS/CS-like and BRRS patients and included MUTYH, RET, TSC2, BRCA1, BRCA2, ERCC2 and HRAS. We found classic phenotypes associated with the identified genes in 5 of the 7 (71.4%) patients. Variant positive patients were enriched for the presence of second malignant neoplasms compared to patients without identified variants (OR = 6.101, 95% CI 1.143-35.98, p = 0.035). Germline variant spectrum and frequencies were compared to The Cancer Genome Atlas (TCGA), including 6 apparently sporadic cancers associated with PHTS. With comparable overall prevalence of germline variants, the spectrum of mutated genes was different in our patients compared to TCGA. Intriguingly, we also found notable enrichment of variants of uncertain significance (VUS) in our patients (OR = 2.3, 95% CI 1.5-3.5, p = 0.0002). Our data suggest that only a small subset of PTEN-wildtype CS/CS-like and BRRS patients could be accounted for by germline variants in some of the known cancer-related genes. Thus, the existence of alterations in other and more likely non-classic cancer-associated genes is plausible, reflecting the complexity of these heterogeneous hereditary cancer syndromes.


Asunto(s)
Mutación de Línea Germinal , Síndrome de Hamartoma Múltiple/genética , Oncogenes , Fosfohidrolasa PTEN/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , ADN Glicosilasas/genética , Análisis Mutacional de ADN , Femenino , Genes BRCA1 , Genes BRCA2 , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Neoplasias Primarias Múltiples/genética , Estudios Prospectivos , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Secuenciación del Exoma , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Adulto Joven
11.
Hum Mol Genet ; 27(18): 3154-3164, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29893852

RESUMEN

SEC23B is a component of coat protein complex II (COPII) vesicles that transport secretory proteins from the endoplasmic reticulum (ER) to the Golgi apparatus. Loss-of-function SEC23B mutations cause a rare form of anemia, resulting from decreased SEC23B levels. We recently identified germline heterozygous SEC23B variants as potentially cancer-predisposing. Mutant SEC23B associated with ER stress-mediated tumorigenesis, without decreased SEC23B expression. However, our understanding of the processes behind these observations remain limited. Here, we show mutant SEC23B exists within nucleoli, in addition to classical distribution at the ER/Golgi. This occurs independent of other COPII proteins and does not compromise secretory function. Mutant cells have increased ribosomal protein and translation-related gene expression, and enhanced translational capacity, in the presence of ER stress. We show that mutant SEC23B binds to UBF transcription factor, with increased UBF transcription factor binding at the ribosomal DNA promoter. Our data indicate SEC23B has potential non-canonical COPII-independent function, particularly within the ribosome biogenesis pathway, and that may contribute to the pathogenesis of cancer-predisposition.


Asunto(s)
Neoplasias/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Ribosomas/genética , Proteínas de Transporte Vesicular/genética , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Carcinogénesis/genética , Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/genética , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal/genética , Aparato de Golgi/genética , Heterocigoto , Humanos , Mutación con Pérdida de Función/genética , Neoplasias/patología , Unión Proteica
12.
Epilepsia ; 61(10): 2203-2213, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32945555

RESUMEN

OBJECTIVE: Memory dysfunction is prevalent in many neurological disorders and can have a significant negative impact on quality of life. The genetic contributions to memory impairment in epilepsy, particularly temporal lobe epilepsy (TLE), remain poorly understood. Here, we compare the brain transcriptome between TLE patients with and without verbal memory impairments to identify genes and signaling networks important for episodic memory. METHODS: Brain tissues were resected from 23 adults who underwent dominant temporal lobectomy for treatment of pharmacoresistant epilepsy. To control for potential effects of APOE on memory, only those homozygous for the APOE ε3 allele were included. A battery of memory tests was performed, and patients were stratified into two groups based on preoperative memory performance. The groups were well matched on demographic and disease-related variables. Total RNA-Seq and small RNA-Seq were performed on RNA extracted from the brain tissues. Pathway and integrative analyses were subsequently performed. RESULTS: We identified 1092 differentially expressed transcripts (DETs), with the majority (71%) being underexpressed in brain tissues from patients with impaired memory compared to those from patients with intact memory. Enrichment analysis revealed overrepresentation of genes in pathways pertaining to brain-related neurological dysfunction, including a subset associated with neurodegenerative diseases, memory, and cognition (APP, MAPT, PINK1). Despite including patients with identical APOE genotypes, we identify APOE as a differentially expressed gene associated with memory status. Small RNA-Seq identified four differentially expressed microRNAs (miRNAs) that were predicted to target a subset (22%) of all DETs. Integrative analysis showed that these miRNA-predicted DET targets impact brain-related pathways and biological processes also pertinent to memory and cognition. SIGNIFICANCE: TLE-associated memory status may be influenced by differences in gene expression profiles within the temporal lobe. Upstream processes influencing differential expression signatures, such as miRNAs, could serve as biomarkers and potential treatment targets for memory impairment in TLE.


Asunto(s)
Apolipoproteína E3/genética , Encéfalo/fisiología , Epilepsia del Lóbulo Temporal/genética , Trastornos de la Memoria/genética , Transcriptoma/genética , Aprendizaje Verbal/fisiología , Adulto , Anciano , Encéfalo/patología , Encéfalo/cirugía , Epilepsia del Lóbulo Temporal/diagnóstico , Epilepsia del Lóbulo Temporal/cirugía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Análisis de Secuencia de ARN/métodos , Adulto Joven
13.
Hum Mol Genet ; 26(2): 243-257, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28011713

RESUMEN

Cowden syndrome (CS) is an autosomal dominant disorder that predisposes to breast, thyroid, and other epithelial cancers. Differentiated thyroid carcinoma (DTC), as one of the major component cancers of CS, is the fastest rising incident cancer in the USA, and the most familial of all solid tumours. To identify additional candidate genes of CS and potentially DTC, we analysed a multi-generation CS-like family with papillary thyroid cancer (PTC), applying a combined linkage-based and whole-genome sequencing strategy and identified an in-frame germline compound heterozygous deletion, p.[Gln1478del];[Gln1476-Gln1478del] in USF3 (previously known as KIAA2018). Among 90 unrelated CS/CS-like individuals, 29% were found to have p.[Gln1478del];[Gln1476-Gln1478del]. Of 497 TCGA PTC individuals, 138 (27%) were found to carry this germline compound deletion, with somatically decreased tumour USF3 expression. We demonstrate an increased migration phenotype along with enhanced epithelial-to-mesenchymal transition (EMT) signature after USF3 knockdown or USF3 p.[Gln1478del];[Gln1476-Gln1478del] overexpression, which sensitizes cells to the endoplasmic reticulum (ER) stress. Loss of USF3 function induced cell necrosis-like features and impaired respiratory capacity while providing a glutamine-dependent cell survival advantage, strongly suggests a metabolic survival and migration-favouring microenvironment for carcinogenesis. Therefore, USF3 may be involved in the predisposition of thyroid cancer. Importantly, the results that glutamine-dependent survival and sensitivity to ER stress in USF3-deficient cells provide avenues for therapeutic and adjunct preventive interventions for both sporadic cancer as well as cancer predisposition syndromes with similar mechanisms.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma/genética , Predisposición Genética a la Enfermedad , Síndrome de Hamartoma Múltiple/genética , Neoplasias de la Tiroides/genética , Factores Estimuladores hacia 5'/genética , Carcinoma/patología , Carcinoma Papilar , Movimiento Celular , Estrés del Retículo Endoplásmico/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Genoma Humano , Genotipo , Mutación de Línea Germinal , Síndrome de Hamartoma Múltiple/patología , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Linaje , Péptidos/genética , Eliminación de Secuencia , Cáncer Papilar Tiroideo , Glándula Tiroides/patología , Neoplasias de la Tiroides/patología , Microambiente Tumoral/genética
14.
Am J Hum Genet ; 97(5): 661-76, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26522472

RESUMEN

Cancer-predisposing genes associated with inherited cancer syndromes help explain mechanisms of sporadic carcinogenesis and often inform normal development. Cowden syndrome (CS) is an autosomal-dominant disorder characterized by high lifetime risks of epithelial cancers, such that ∼50% of affected individuals are wild-type for known cancer-predisposing genes. Using whole-exome and Sanger sequencing of a multi-generation CS family affected by thyroid and other cancers, we identified a pathogenic missense heterozygous SEC23B variant (c.1781T>G [p.Val594Gly]) that segregates with the phenotype. We also found germline heterozygous SEC23B variants in 3/96 (3%) unrelated mutation-negative CS probands with thyroid cancer and in The Cancer Genome Atlas (TCGA), representing apparently sporadic cancers. We note that the TCGA thyroid cancer dataset is enriched with unique germline deleterious SEC23B variants associated with a significantly younger age of onset. SEC23B encodes Sec23 homolog B (S. cerevisiae), a component of coat protein complex II (COPII), which transports proteins from the endoplasmic reticulum (ER) to the Golgi apparatus. Interestingly, germline homozygous or compound-heterozygous SEC23B mutations cause an unrelated disorder, congenital dyserythropoietic anemia type II, and SEC23B-deficient mice suffer from secretory organ degeneration due to ER-stress-associated apoptosis. By characterizing the p.Val594Gly variant in a normal thyroid cell line, we show that it is a functional alteration that results in ER-stress-mediated cell-colony formation and survival, growth, and invasion, which reflect aspects of a cancer phenotype. Our findings suggest a different role for SEC23B, whereby germline heterozygous variants associate with cancer predisposition potentially mediated by ER stress "addiction."


Asunto(s)
Estrés del Retículo Endoplásmico , Mutación de Línea Germinal/genética , Síndrome de Hamartoma Múltiple/genética , Síndrome de Hamartoma Múltiple/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Proteínas de Transporte Vesicular/genética , Adolescente , Adulto , Anciano , Animales , Apoptosis , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Exoma/genética , Femenino , Técnica del Anticuerpo Fluorescente , Estudios de Seguimiento , Genotipo , Síndrome de Hamartoma Múltiple/metabolismo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Linaje , Fenotipo , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Tiroides/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/fisiología , Adulto Joven
15.
J Allergy Clin Immunol ; 139(2): 607-620.e15, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27477328

RESUMEN

BACKGROUND: Patients with heterozygous germline mutations in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) experience autoimmunity and lymphoid hyperplasia. OBJECTIVES: Because regulation of the phosphoinositide 3-kinase (PI3K) pathway is critical for maintaining regulatory T (Treg) cell functions, we investigate Treg cells in patients with heterozygous germline PTEN mutations (PTEN hamartoma tumor syndrome [PHTS]). METHODS: Patients with PHTS were assessed for immunologic conditions, lymphocyte subsets, forkhead box P3 (FOXP3)+ Treg cell levels, and phenotype. To determine the functional importance of phosphatases that control the PI3K pathway, we assessed Treg cell induction in vitro, mitochondrial depolarization, and recruitment of PTEN to the immunologic synapse. RESULTS: Autoimmunity and peripheral lymphoid hyperplasia were found in 43% of 79 patients with PHTS. Immune dysregulation in patients with PHTS included lymphopenia, CD4+ T-cell reduction, and changes in T- and B-cell subsets. Although total CD4+FOXP3+ Treg cell numbers are reduced, frequencies are maintained in the blood and intestine. Despite pathogenic PTEN mutations, the FOXP3+ T cells are phenotypically normal. We show that the phosphatase PH domain leucine-rich repeat protein phosphatase (PHLPP) downstream of PTEN is highly expressed in normal human Treg cells and provides complementary phosphatase activity. PHLPP is indispensable for the differentiation of induced Treg cells in vitro and Treg cell mitochondrial fitness. PTEN and PHLPP form a phosphatase network that is polarized at the immunologic synapse. CONCLUSION: Heterozygous loss of function of PTEN in human subjects has a significant effect on T- and B-cell immunity. Assembly of the PTEN-PHLPP phosphatase network allows coordinated phosphatase activities at the site of T-cell receptor activation, which is important for limiting PI3K hyperactivation in Treg cells despite PTEN haploinsufficiency.


Asunto(s)
Linfocitos B/fisiología , Síndrome de Hamartoma Múltiple/inmunología , Sinapsis Inmunológicas/metabolismo , Subgrupos Linfocitarios/fisiología , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Linfocitos T Reguladores/fisiología , Adolescente , Adulto , Anciano , Autoinmunidad , Células Cultivadas , Niño , Factores de Transcripción Forkhead/metabolismo , Síndrome de Hamartoma Múltiple/genética , Humanos , Hiperplasia , Masculino , Potencial de la Membrana Mitocondrial , Persona de Mediana Edad , Mutación/genética , Fosfohidrolasa PTEN/genética , Unión Proteica , Transporte de Proteínas , Transducción de Señal , Adulto Joven
16.
Gastroenterology ; 149(4): 886-9.e5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26122142

RESUMEN

Hamartomatous polyposis syndromes (HPS) account for a small but appreciable proportion of inherited gastrointestinal cancer predisposition syndromes; patients with HPS have an increased risk for colon and extracolonic malignancies. We present a unique case of familial juvenile polyposis syndrome associated with gastrointestinal ganglioneuromas of unknown etiology. The patient was tested for HPS-associated genes, but no mutation was detected. Exome sequencing identified a germline heterozygous mutation in SMAD9 (SMAD9(V90M)). This mutation was predicted to be an activating mutation. HEK cells transfected to express SMAD9(V90M) had reduced expression of phosphatase and tensin homolog; this reduction was also observed in a polyp from the patient. We have therefore identified a new susceptibility locus for HPS. Patients with hamartomatous polyposis in the colon associated with ganglioneuromatosis should be referred for genetic assessments.


Asunto(s)
Pólipos del Colon/genética , Neoplasias del Sistema Digestivo/genética , Exoma , Ganglioneuroma/genética , Mutación de Línea Germinal , Neoplasia Endocrina Múltiple Tipo 2b/genética , Fosfohidrolasa PTEN/metabolismo , Síndrome de Peutz-Jeghers/genética , Proteína Smad8/genética , Adulto , Pólipos del Colon/diagnóstico , Pólipos del Colon/enzimología , Análisis Mutacional de ADN , Neoplasias del Sistema Digestivo/diagnóstico , Neoplasias del Sistema Digestivo/enzimología , Regulación hacia Abajo , Femenino , Ganglioneuroma/diagnóstico , Ganglioneuroma/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 2b/diagnóstico , Neoplasia Endocrina Múltiple Tipo 2b/enzimología , Fosfohidrolasa PTEN/genética , Síndrome de Peutz-Jeghers/diagnóstico , Síndrome de Peutz-Jeghers/enzimología , Fenotipo , Proteína Smad8/metabolismo , Transfección
18.
Cell Rep Med ; 5(2): 101384, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38242121

RESUMEN

Individuals with PTEN hamartoma tumor syndrome (PHTS) harbor pathogenic germline PTEN variants that confer a significantly increased lifetime risk of various organ-specific cancers including second primary malignant neoplasms (SMNs). Currently, there are no reliable biomarkers that can predict individual-level cancer risk. Despite the highly promising value of cell-free DNA (cfDNA) as a biomarker for underlying sporadic cancers, the utility of cfDNA in individuals with known cancer-associated germline variants and subclinical cancers remains poorly understood. We perform ultra-low-pass whole-genome sequencing (ULP-WGS) of cfDNA from plasma samples from patients with PHTS and cancer as well as those without cancer. Analysis of cfDNA reveals that patients with PHTS and SMNs have distinct cfDNA size distribution, aberrant genome-wide fragmentation, and differential fragment end motif frequencies. Our work provides evidence that cfDNA profiles may be used as a marker for SMN risk in patients with PHTS.


Asunto(s)
Ácidos Nucleicos Libres de Células , Síndrome de Hamartoma Múltiple , Neoplasias , Humanos , Síndrome de Hamartoma Múltiple/diagnóstico , Síndrome de Hamartoma Múltiple/genética , Síndrome de Hamartoma Múltiple/complicaciones , Fosfohidrolasa PTEN/genética , Mutación de Línea Germinal , Ácidos Nucleicos Libres de Células/genética
19.
Res Sq ; 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38168271

RESUMEN

Individuals with germline PTEN variants (PHTS) have increased risks of the seemingly disparate phenotypes of cancer and neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD). Etiology of the phenotypic variability remains elusive. Here, we hypothesized that decreased genomic diversity, manifested by increased homozygosity, may be one etiology. Comprehensive analyses of 376 PHTS patients of European ancestry revealed significant enrichment of homozygous common variants in genes involved in inflammatory processes in the PHTS-NDD group and in genes involved in differentiation and chromatin structure regulation in the PHTS-ASD group. Pathway analysis revealed pathways germane to NDD/ASD, including neuroinflammation and synaptogenesis. Collapsing analysis of the homozygous variants identified suggestive modifier NDD/ASD genes. In contrast, we found enrichment of homozygous ultra-rare variants in genes modulating cell death in the PHTS-cancer group. Finally, homozygosity burden as a predictor of ASD versus cancer outcomes in our validated prediction model for NDD/ASD performed favorably.

20.
NPJ Genom Med ; 8(1): 14, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37407629

RESUMEN

Women with germline PTEN variants (PTEN hamartoma tumor syndrome, PHTS) have up to 85% lifetime risk of female breast cancer (BC). We previously showed that PHTS-derived BCs are distinct from sporadic BCs both at the clinical and genomic levels. In this study, we examined somatic copy number variations (CNV) and transcriptome data to further characterize the somatic landscape of PHTS-derived BCs. We analyzed exome sequencing data from 44 BCs from women with PHTS for CNV. The control group comprised of 558 women with sporadic BCs from The Cancer Genome Atlas (TCGA) dataset. Here, we found that PHTS-derived BCs have several distinct CNV peaks compared to TCGA. Furthermore, RNA sequencing data revealed that PHTS-derived BCs have a distinct immunologic cell type signature, which points toward cancer immune evasion. Transcriptomic data also revealed PHTS-derived BCs with pathogenic germline PTEN variants appear to have vitamin E degradation as a key pathway associated with tumorigenesis. In conclusion, our study revealed distinct CNV x transcript features in PHTS-derived BCs, which further facilitate understanding of BC biology arising in the setting of germline PTEN mutations.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA